Network


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

Hotspot


Dive into the research topics where Anat Yanai is active.

Publication


Featured researches published by Anat Yanai.


Neuron | 2004

Huntingtin-Interacting Protein HIP14 Is a Palmitoyl Transferase Involved in Palmitoylation and Trafficking of Multiple Neuronal Proteins

Kun Huang; Anat Yanai; Rujun Kang; Pamela Arstikaitis; Roshni R. Singaraja; Martina Metzler; Asher Mullard; Brendan J. Haigh; Catherine Gauthier-Campbell; Claire-Anne Gutekunst; Michael R. Hayden; Alaa El-Husseini

In neurons, posttranslational modification by palmitate regulates the trafficking and function of signaling molecules, neurotransmitter receptors, and associated synaptic scaffolding proteins. However, the enzymatic machinery involved in protein palmitoylation has remained elusive. Here, using biochemical assays, we show that huntingtin (htt) interacting protein, HIP14, is a neuronal palmitoyl transferase (PAT). HIP14 shows remarkable substrate specificity for neuronal proteins, including SNAP-25, PSD-95, GAD65, synaptotagmin I, and htt. Conversely, HIP14 is catalytically invariant toward paralemmin and synaptotagmin VII. Exogenous HIP14 enhances palmitoylation-dependent vesicular trafficking of several acylated proteins in both heterologous cells and neurons. Moreover, interference with endogenous expression of HIP14 reduces clustering of PSD-95 and GAD65 in neurons. These findings define HIP14 as a mammalian palmitoyl transferase involved in the palmitoylation and trafficking of multiple neuronal proteins.


Nature Neuroscience | 2006

Palmitoylation of huntingtin by HIP14 is essential for its trafficking and function.

Anat Yanai; Kun Huang; Rujun Kang; Roshni R. Singaraja; Pamela Arstikaitis; Lu Gan; Paul C. Orban; Asher Mullard; Catherine M. Cowan; Lynn A. Raymond; Renaldo C. Drisdel; William N. Green; Brinda Ravikumar; David C. Rubinsztein; Alaa El-Husseini; Michael R. Hayden

Post-translational modification by the lipid palmitate is crucial for the correct targeting and function of many proteins. Here we show that huntingtin (htt) is normally palmitoylated at cysteine 214, which is essential for its trafficking and function. The palmitoylation and distribution of htt are regulated by the palmitoyl transferase huntingtin interacting protein 14 (HIP14). Expansion of the polyglutamine tract of htt, which causes Huntington disease, results in reduced interaction between mutant htt and HIP14 and consequently in a marked reduction in palmitoylation. Mutation of the palmitoylation site of htt, making it palmitoylation resistant, accelerates inclusion formation and increases neuronal toxicity. Downregulation of HIP14 in mouse neurons expressing wild-type and mutant htt increases inclusion formation, whereas overexpression of HIP14 substantially reduces inclusions. These results suggest that the expansion of the polyglutamine tract in htt results in decreased palmitoylation, which contributes to the formation of inclusion bodies and enhanced neuronal toxicity.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Intercellular propagated misfolding of wild-type Cu/Zn superoxide dismutase occurs via exosome-dependent and -independent mechanisms.

Leslie I. Grad; Justin J. Yerbury; Bradley J. Turner; Will Guest; Edward Pokrishevsky; Megan A. O'Neill; Anat Yanai; Judith M. Silverman; Rafaa Zeineddine; Lisa Corcoran; Janet R. Kumita; Leila M. Luheshi; Masoud Yousefi; Bradley M. Coleman; Andrew F. Hill; Steven S. Plotkin; Ian R. Mackenzie; Neil R. Cashman

Significance Amyotrophic lateral sclerosis (ALS), an incurable motor neuron disease, is associated with mutation and misfolding of the Cu/Zn superoxide dismutase (SOD1) protein. Prior studies found that mutant misfolded SOD1 can convert wild-type (WT) SOD1 to a misfolded form inside living cells in a prion-like fashion. We now report that misfolded WT SOD1 can be transmitted from cell to cell, and that propagated protein misfolding can be perpetuated. Misfolded SOD1 transmission between cells can be mediated through release and uptake of protein aggregates or via small membrane-bounded transport vesicles called exosomes. These mechanisms may help explain why sporadic ALS, without a known genetic cause, can spread systematically from region to region in a progressive manner. Amyotrophic lateral sclerosis (ALS) is predominantly sporadic, but associated with heritable genetic mutations in 5–10% of cases, including those in Cu/Zn superoxide dismutase (SOD1). We previously showed that misfolding of SOD1 can be transmitted to endogenous human wild-type SOD1 (HuWtSOD1) in an intracellular compartment. Using NSC-34 motor neuron-like cells, we now demonstrate that misfolded mutant and HuWtSOD1 can traverse between cells via two nonexclusive mechanisms: protein aggregates released from dying cells and taken up by macropinocytosis, and exosomes secreted from living cells. Furthermore, once HuWtSOD1 propagation has been established, misfolding of HuWtSOD1 can be efficiently and repeatedly propagated between HEK293 cell cultures via conditioned media over multiple passages, and to cultured mouse primary spinal cord cells transgenically expressing HuWtSOD1, but not to cells derived from nontransgenic littermates. Conditioned media transmission of HuWtSOD1 misfolding in HEK293 cells is blocked by HuWtSOD1 siRNA knockdown, consistent with human SOD1 being a substrate for conversion, and attenuated by ultracentrifugation or incubation with SOD1 misfolding-specific antibodies, indicating a relatively massive transmission particle which possesses antibody-accessible SOD1. Finally, misfolded and protease-sensitive HuWtSOD1 comprises up to 4% of total SOD1 in spinal cords of patients with sporadic ALS (SALS). Propagation of HuWtSOD1 misfolding, and its subsequent cell-to-cell transmission, is thus a candidate process for the molecular pathogenesis of SALS, which may provide novel treatment and biomarker targets for this devastating disease.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Intermolecular transmission of superoxide dismutase 1 misfolding in living cells

Leslie I. Grad; Will Guest; Anat Yanai; Edward Pokrishevsky; Megan A. O'Neill; Ebrima Gibbs; Valentyna Semenchenko; Masoud Yousefi; David S. Wishart; Steven S. Plotkin; Neil R. Cashman

Human wild-type superoxide dismutase-1 (wtSOD1) is known to coaggregate with mutant SOD1 in familial amyotrophic lateral sclerosis (FALS), in double transgenic models of FALS, and in cell culture systems, but the structural determinants of this process are unclear. Here we molecularly dissect the effects of intracellular and cell-free obligately misfolded SOD1 mutant proteins on natively structured wild-type SOD1. Expression of the enzymatically inactive, natural familial ALS SOD1 mutations G127X and G85R in human mesenchymal and neural cell lines induces misfolding of wild-type natively structured SOD1, as indicated by: acquisition of immunoreactivity with SOD1 misfolding-specific monoclonal antibodies; markedly enhanced protease sensitivity suggestive of structural loosening; and nonnative disulfide-linked oligomer and multimer formation. Expression of G127X and G85R in mouse cell lines did not induce misfolding of murine wtSOD1, and a species restriction element for human wtSOD1 conversion was mapped to a region of sequence divergence in loop II and β-strand 3 of the SOD1 β-barrel (residues 24–36), then further refined surprisingly to a single tryptophan residue at codon 32 (W32) in human SOD1. Time course experiments enabled by W32 restriction revealed that G127X and misfolded wtSOD1 can induce misfolding of cell-endogenous wtSOD1. Finally, aggregated recombinant G127X is capable of inducing misfolding and protease sensitivity of recombinant human wtSOD1 in a cell-free system containing reducing and chelating agents; cell-free wtSOD1 conversion was also restricted by W32. These observations demonstrate that misfolded SOD1 can induce misfolding of natively structured wtSOD1 in a physiological intracellular milieu, consistent with a direct protein–protein interaction.


The FASEB Journal | 2009

Neuronal palmitoyl acyl transferases exhibit distinct substrate specificity

Kun Huang; Shaun S. Sanders; Roshni R. Singaraja; Paul C. Orban; Tony Cijsouw; Pamela Arstikaitis; Anat Yanai; Michael R. Hayden; Alaa El-Husseini

Palmitoylation, a post‐translational modification of cysteine residues with the lipid palmitate, has recently emerged as an important mechanism for regulating protein trafficking and function. With the identification of 23 DHHC mammalian palmitoyl acyl transferases (PATs), a key question was the nature of substrate‐enzyme specificity for these PATs. Using the acyl‐biotin exchange palmitoylation assay, we compared the substrate specificity of four neuronal PATs, namely DHHC‐3, DHHC‐8, HIP14L (DHHC‐13), and HIP14 (DHHC‐17). Exogenous expression of enzymes and substrates in COS cells reveals that HIP14L and HIP14 modulate huntingtin palmitoylation, DHHC‐8 modulates paralemmin‐1 palmitoylation, and DHHC‐3 shows the least substrate specificity. These in vitro data were validated by lentiviral siRNA‐mediated knockdown of endogenous HIP14 and DHHC‐3 in cultured rat cortical neurons. PATs require the presence of palmitoylated cysteines in order to interact with their substrates. To understand the elements that influence enzyme/ substrate specificity further, we fused the HIP14 ankryin repeat domain to the N terminus of DHHC‐3, which is not a PAT for huntingtin. This modification enabled DHHC‐3 to behave similarly to HIP14 by modulating palmitoylation and trafficking of huntingtin. Taken together, this study indicates that individual PATs have specific substrate preference, determined by regulatory domains outside the DHHC domain of the enzymes.— Huang, K., Sanders, S., Singaraja, R., Orban, P., Cij‐souw, T., Arstikaitis, P., Yanai, A., Hayden, M. R., El‐Husseini, A. Neuronal palmitoyl acyl transferases exhibit distinct substrate specificity. FASEB J. 23, 2605–2615 (2009)


Cell Transplantation | 2012

Focused Magnetic Stem Cell Targeting to the Retina Using Superparamagnetic Iron Oxide Nanoparticles

Anat Yanai; Urs O. Häfeli; Andrew Metcalfe; Peter Soema; Lois Addo; Cheryl Y. Gregory-Evans; Kelvin Po; Xianghong Shan; Orson L. Moritz; Kevin Gregory-Evans

Developing new ways of delivering cells to diseased tissue will be a key factor in translating cell therapeutics research into clinical use. Magnetically targeting cells enables delivery of significant numbers of cells to key areas of specific organs. To demonstrate feasibility in neurological tissue, we targeted cells magnetically to the upper hemisphere of the rodent retina. Rat mesenchymal stem cells (MSCs) were magnetized using superparamagnetic iron oxide nanoparticles (SPIONs). In vitro studies suggested that magnetization with fluidMAG-D was well tolerated, that cells remained viable, and they retained their differentiation capabilities. FluidMAG-D-labeled MSCs were injected intravitreally or via the tail vein of the S334ter-4 transgenic rat model of retinal degeneration with or without placing a gold-plated neodymium disc magnet within the orbit, but outside the eye. Retinal flatmount and cryosection imaging demonstrated that after intravitreal injection cells localized to the inner retina in a tightly confined area corresponding to the position of the orbital magnet. After intravenous injection, similar retinal localization was achieved and remarkably was associated with a tenfold increase in magnetic MSC delivery to the retina. Cryosections demonstrated that cells had migrated into both the inner and outer retina. Magnetic MSC treatment with orbital magnet also resulted in significantly higher retinal concentrations of anti-inflammatory molecules interleukin-10 and hepatocyte growth factor. This suggested that intravenous MSC therapy also resulted in significant therapeutic benefit in the dystrophic retina. With minimal risk of collateral damage, these results suggest that magnetic cell delivery is the best approach for controlled delivery of cells to the outer retina—the focus for disease in age-related macular degeneration and retinitis pigmentosa.


Methods of Molecular Biology | 2013

Efficient Production of Photoreceptor Precursor Cells from Human Embryonic Stem Cells.

Anat Yanai; Christopher Laver; Aaron W. Joe; Kevin Gregory-Evans

Transplantation of photoreceptor precursor cells (PPCs) differentiated from human embryonic stem cells (hESCs) is a promising approach to treat common blinding diseases such as age-related macular degeneration and retinitis pigmentosa. However, existing PPC generation methods are inefficient. To enhance differentiation protocols for rapid and high-yield production of PPCs, we focused on optimizing the handling of the cells by including feeder-independent growth of hESCs, using size-controlled embryoid bodies (EBs), and addition of triiodothyronine (T3) and taurine to the differentiation medium, with subsequent removal of undifferentiated cells via negative cell-selection. Our novel protocol produces higher yields of PPCs than previously reported while reducing the time required for differentiation, which will help understand retinal diseases and facilitate large-scale preclinical trials.


IEEE Transactions on Magnetics | 2013

Influence of Iron Oxide Nanoparticles on Innate and Genetically Modified Secretion Profiles of Mesenchymal Stem Cells

Abu E. Bashar; Andrew Metcalfe; Anat Yanai; Christopher Laver; Urs O. Häfeli; Cheryl Y. Gregory-Evans; Orson L. Moritz; Joanne A. Matsubara; Kevin Gregory-Evans

Mesenchymal stem cells (MSCs) have well-established paracrine effects that are proving to be therapeutically useful. This potential is based on the ability of MSCs to secrete a range of neuroprotective and anti-inflammatory molecules. Previous work in our laboratory has demonstrated that intravenous injection of MSCs, treated with superparamagnetic iron oxide nanoparticle fluidMAG-D resulted in enhanced levels of glial-derived neurotrophic factor, ciliary neurotrophic factor, hepatocyte growth factor and interleukin-10 in the dystrophic rat retina. In this present study we investigated whether the concentration of fluidMAG-D in cell culture media affects the secretion of these four molecules in vitro. In addition, we assessed the effect of fluidMAG-D concentration on retinoschisin secretion from genetically modified MSCs. ELISA-assayed secretion of these molecules was measured using escalating concentrations of fluidMAG-D which resulted in MSC iron loads of 0, 7, 120, or 274 pg iron oxide per cell respectively. Our results demonstrated glial-derived neurotrophic factor and hepatocyte growth factor secretion was significantly decreased but only at the 96 hours time-point whereas no statistically significant effect was seen with ciliary neurotrophic factor secretion. Whereas no effect was observed on culture media concentrations of retinoschisin with increasing iron oxide load, a statistically significant increase in cell lysate retinoschisin concentration (p=0.01) was observed suggesting that increasing fluidMAG-D concentration did increase retinoschisin production but this did not lead to greater secretion. We hypothesize that higher concentrations of iron-oxide nanoparticle fluidMAG-D have an effect on the innate ability of MSCs to secrete therapeutically useful molecules and also on secretion from genetically modified cells. Further work is required to verify these in vitro finding using in vivo model systems.


Regenerative Medicine | 2018

The olfactory mucosa: a potential source of stem cells for hearing regeneration

Emily Young; Brian D. Westerberg; Anat Yanai; Kevin Gregory-Evans

The olfactory mucosa contains cells that enable it to generate new neurons and other supporting cells throughout life, allowing it to replace cells of the mucosa that have been damaged by exposure to various insults. In this article, we discuss the different types of stem cell found within the olfactory mucosa and their properties. In particular, the mesenchymal-like cells found within the lamina propria will be reviewed in detail. In addition, we discuss potential applications of olfactory-derived stem cells toward hearing regeneration secondary to either inner hair cell loss or primary or secondary auditory nerve degeneration.


Journal of Tissue Engineering and Regenerative Medicine | 2017

Anti-ageing glycoprotein promotes long-term survival of transplanted neurosensory precursor cells.

Anat Yanai; Ishaq A. Viringipurampeer; Emran Bashar; Kevin Gregory-Evans

Cell therapy, to replace lost tissue, is a promising approach for the treatment of various neurodegenerative diseases. Many studies suggest, however, that the percentage of transplanted cells that survive and undergo functional integration remains low as a result of immune rejection, suboptimal precursor cell type, trauma during cell transplantation, toxic compounds released by dying tissues or nutritional deficiencies. We recently developed an ex vivo system to facilitate identification of factors contributing to the death of transplanted neuronal (photoreceptor) cells and compounds that block these toxic effects. In this system, photoreceptor precursor cells (PPCs) are sandwiched between a neurosensory retinal explant and retinal pigment epithelium derived from human embryonic stem cells. Explant medium was collected to identify toxic components and PPC survival was assessed by flow cytometry. We also assessed the potential for AAGP™, a cryopreservative molecule, to improve PPC survival. We identified elevated prostaglandin E2 (PGE2) in the explant medium and demonstrated that AAGP™ reduced PGE2 levels by 2.6‐fold. A pro‐inflammatory stress assay suggested that this may result from AAGP™ inhibition of cyclo‐oxygenase‐2 (COX‐2) expression. We confirmed that PGE2 reduced the viability of cultured PPCs by 44% and found that the survival rate of PPCs pretreated with AAGP™ was 2.8‐fold higher than in untreated PPCs. These data suggest that PGE2 release from necrotic tissue may be one factor that reduces the survival of transplanted precursor cells and that the pro‐survival molecule AAGP™ may improve long‐term transplanted cell viability. Copyright

Collaboration


Dive into the Anat Yanai's collaboration.

Top Co-Authors

Avatar

Kevin Gregory-Evans

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Christopher Laver

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Andrew Metcalfe

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Cheryl Y. Gregory-Evans

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael R. Hayden

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aaron W. Joe

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Alaa El-Husseini

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Kun Huang

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge