Network


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

Hotspot


Dive into the research topics where Julia A. Kaye is active.

Publication


Featured researches published by Julia A. Kaye.


Cell Stem Cell | 2012

Induced Pluripotent Stem Cells from Patients with Huntington’s Disease : Show CAG Repeat-Expansion-Associated Phenotypes

Virginia B. Mattis; Soshana Svendsen; Allison D. Ebert; Clive N. Svendsen; Alvin R. King; Malcolm Casale; Sara T. Winokur; Gayani Batugedara; Marquis P. Vawter; Peter J. Donovan; Leslie F. Lock; Leslie M. Thompson; Yu Zhu; Elisa Fossale; Ranjit S. Atwal; Tammy Gillis; Jayalakshmi S. Mysore; Jian Hong Li; Ihn Sik Seong; Yiping Shen; Xiaoli Chen; Vanessa C. Wheeler; Marcy E. MacDonald; James F. Gusella; Sergey Akimov; Nicolas Arbez; Tarja Juopperi; Tamara Ratovitski; Jason H. Chiang; Woon Roung Kim

Huntingtons disease (HD) is an inherited neurodegenerative disorder caused by an expanded stretch of CAG trinucleotide repeats that results in neuronal dysfunction and death. Here, The HD Consortium reports the generation and characterization of 14 induced pluripotent stem cell (iPSC) lines from HD patients and controls. Microarray profiling revealed CAG-repeat-expansion-associated gene expression patterns that distinguish patient lines from controls, and early onset versus late onset HD. Differentiated HD neural cells showed disease-associated changes in electrophysiology, metabolism, cell adhesion, and ultimately cell death for lines with both medium and longer CAG repeat expansions. The longer repeat lines were however the most vulnerable to cellular stressors and BDNF withdrawal, as assessed using a range of assays across consortium laboratories. The HD iPSC collection represents a unique and well-characterized resource to elucidate disease mechanisms in HD and provides a human stem cell platform for screening new candidate therapeutics.


Current Biology | 2004

DNA breaks promote genomic instability by impeding proper chromosome segregation.

Julia A. Kaye; Justine A. Melo; Stephanie K. Cheung; Moreshwar B. Vaze; James E. Haber; David P. Toczyski

BACKGROUND Unrepaired DNA double-stranded breaks (DSBs) can result in the whole or partial loss of chromosomes. Previously, we showed that the ends of broken chromosomes remain associated. Here, we have examined the machinery that holds broken chromosome ends together, and we have explored the behavior of broken chromosomes as they pass through mitosis. RESULTS Using GFP-localized arrays flanking an HO endonuclease site, we examined the association of broken chromosome ends in yeast cells that are checkpoint-arrested in metaphase. This association is partially dependent upon Rad50 and Rad52. After 6-8 hr, cells adapted to the checkpoint and resumed mitosis, segregating the broken chromosome. When this occurred, we found that the acentric fragments cosegregated into either the mother or daughter cell 95% of the time. Similarly, pedigree analysis showed that postmitotic repair of a broken chromosome (rejoining the centric and acentric fragments) occurred in either the mother or daughter cell, but rarely both, consistent with a model in which both acentric sister chromatid fragments are passaged into the same nucleus. CONCLUSIONS These data suggest two related phenomena: an intrachromosomal association that holds the halves of a single broken sister chromatid together in metaphase and an interchromosomal force that tethers broken sister chromatids to each other and promotes their missegregation. Strikingly, the interchromosomal association of DNA breaks also promotes the missegregation of centromeric chromosomal fragments, albeit to a lesser extent than acentric fragments. The DNA break-induced missegregation of acentric and centric chromosome fragments provides a novel mechanism for the loss of heterozygosity that precedes tumorigenesis in mammalian cells.


Molecular and Cellular Neuroscience | 2013

Modeling Huntington's disease with induced pluripotent stem cells

Julia A. Kaye; Steven Finkbeiner

Huntingtons disease (HD) causes severe motor dysfunction, behavioral abnormalities, cognitive impairment and death. Investigations into its molecular pathology have primarily relied on murine tissues; however, the recent discovery of induced pluripotent stem cells (iPSCs) has opened new possibilities to model neurodegenerative disease using cells derived directly from patients, and therefore may provide a human-cell-based platform for unique insights into the pathogenesis of HD. Here, we will examine the practical implementation of iPSCs to study HD, such as approaches to differentiate embryonic stem cells (ESCs) or iPSCs into medium spiny neurons, the cell type most susceptible in HD. We will explore the HD-related phenotypes identified in iPSCs and ESCs and review how brain development and neurogenesis may actually be altered early, before the onset of HD symptoms, which could inform the search for drugs that delay disease onset. Finally, we will speculate on the exciting possibility that ESCs or iPSCs might be used as therapeutics to restore or replace dying neurons in HD brains.


Methods in Enzymology | 2012

High-Throughput Screening in Primary Neurons

Punita Sharma; D. Michael Ando; Aaron Daub; Julia A. Kaye; Steven Finkbeiner

Despite years of incremental progress in our understanding of diseases such as Alzheimers disease (AD), Parkinsons disease (PD), Huntingtons disease (HD), and amyotrophic lateral sclerosis (ALS), there are still no disease-modifying therapeutics. The discrepancy between the number of lead compounds and approved drugs may partially be a result of the methods used to generate the leads and highlights the need for new technology to obtain more detailed and physiologically relevant information on cellular processes in normal and diseased states. Our high-throughput screening (HTS) system in a primary neuron model can help address this unmet need. HTS allows scientists to assay thousands of conditions in a short period of time which can reveal completely new aspects of biology and identify potential therapeutics in the span of a few months when conventional methods could take years or fail all together. HTS in primary neurons combines the advantages of HTS with the biological relevance of intact, fully differentiated neurons which can capture the critical cellular events or homeostatic states that make neurons uniquely susceptible to disease-associated proteins. We detail methodologies of our primary neuron HTS assay workflow from sample preparation to data reporting. We also discuss the adaptation of our HTS system into high-content screening (HCS), a type of HTS that uses multichannel fluorescence images to capture biological events in situ, and is uniquely suited to study dynamical processes in living cells.


Human Molecular Genetics | 2016

Targeting TEAD/YAP-transcription-dependent necrosis, TRIAD, ameliorates Huntington's disease pathology.

Ying Mao; Xigui Chen; Min Xu; Kyota Fujita; Kazumi Motoki; Toshikazu Sasabe; Hidenori Homma; Miho Murata; Kazuhiko Tagawa; Takuya Tamura; Julia A. Kaye; Steven Finkbeiner; Giovanni Blandino; Marius Sudol; Hitoshi Okazawa

Neuronal cell death in neurodegenerative diseases is not fully understood. Here we report that mutant huntingtin (Htt), a causative gene product of Huntington’s diseases (HD) selectively induces a new form of necrotic cell death, in which endoplasmic reticulum (ER) enlarges and cell body asymmetrically balloons and finally ruptures. Pharmacological and genetic analyses revealed that the necrotic cell death is distinct from the RIP1/3 pathway-dependent necroptosis, but mediated by a functional deficiency of TEAD/YAP-dependent transcription. In addition, we revealed that a cell cycle regulator, Plk1, switches the balance between TEAD/YAP-dependent necrosis and p73/YAP-dependent apoptosis by shifting the interaction partner of YAP from TEAD to p73 through YAP phosphorylation at Thr77. In vivo ER imaging with two-photon microscopy detects similar ER enlargement, and viral vector-mediated delivery of YAP as well as chemical inhibitors of the Hippo pathway such as S1P recover the ER instability and necrosis in HD model mice. Intriguingly S1P completely stops the decline of motor function of HD model mice even after the onset of symptom. Collectively, we suggest approaches targeting the signalling pathway of TEAD/YAP-transcription-dependent necrosis (TRIAD) could lead to a therapeutic development against HD.


American Journal of Human Genetics | 2015

Sequence-Level Analysis of the Major European Huntington Disease Haplotype

Jong-Min Lee; Kyung Hee Kim; Aram Shin; Michael J. Chao; Kawther Abu Elneel; Tammy Gillis; Jayalakshmi S. Mysore; Julia A. Kaye; Hengameh Zahed; Ian H. Kratter; Aaron Daub; Steven Finkbeiner; Hong Li; Jared C. Roach; Nathan Goodman; Leroy Hood; Richard H. Myers; Marcy E. MacDonald; James F. Gusella

Huntington disease (HD) reflects the dominant consequences of a CAG-repeat expansion in HTT. Analysis of common SNP-based haplotypes has revealed that most European HD subjects have distinguishable HTT haplotypes on their normal and disease chromosomes and that ∼50% of the latter share the same major HD haplotype. We reasoned that sequence-level investigation of this founder haplotype could provide significant insights into the history of HD and valuable information for gene-targeting approaches. Consequently, we performed whole-genome sequencing of HD and control subjects from four independent families in whom the major European HD haplotype segregates with the disease. Analysis of the full-sequence-based HTT haplotype indicated that these four families share a common ancestor sufficiently distant to have permitted the accumulation of family-specific variants. Confirmation of new CAG-expansion mutations on this haplotype suggests that unlike most founders of human disease, the common ancestor of HD-affected families with the major haplotype most likely did not have HD. Further, availability of the full sequence data validated the use of SNP imputation to predict the optimal variants for capturing heterozygosity in personalized allele-specific gene-silencing approaches. As few as ten SNPs are capable of revealing heterozygosity in more than 97% of European HD subjects. Extension of allele-specific silencing strategies to the few remaining homozygous individuals is likely to be achievable through additional known SNPs and discovery of private variants by complete sequencing of HTT. These data suggest that the current development of gene-based targeting for HD could be extended to personalized allele-specific approaches in essentially all HD individuals of European ancestry.


Scientific Reports | 2017

Contribution of the cyclic nucleotide gated channel subunit, CNG-3, to olfactory plasticity in Caenorhabditis elegans

Damien M. O’Halloran; Svetlana Altshuler-Keylin; Xiao Dong Zhang; Chao He; Christopher Morales-Phan; Yawei Yu; Julia A. Kaye; Chantal Brueggemann; Tsung Yu Chen; Noelle D. L’Etoile

In Caenorhabditis elegans, the AWC neurons are thought to deploy a cGMP signaling cascade in the detection of and response to AWC sensed odors. Prolonged exposure to an AWC sensed odor in the absence of food leads to reversible decreases in the animal’s attraction to that odor. This adaptation exhibits two stages referred to as short-term and long-term adaptation. Previously, the protein kinase G (PKG), EGL-4/PKG-1, was shown necessary for both stages of adaptation and phosphorylation of its target, the beta-type cyclic nucleotide gated (CNG) channel subunit, TAX-2, was implicated in the short term stage. Here we uncover a novel role for the CNG channel subunit, CNG-3, in short term adaptation. We demonstrate that CNG-3 is required in the AWC for adaptation to short (thirty minute) exposures of odor, and contains a candidate PKG phosphorylation site required to tune odor sensitivity. We also provide in vivo data suggesting that CNG-3 forms a complex with both TAX-2 and TAX-4 CNG channel subunits in AWC. Finally, we examine the physiology of different CNG channel subunit combinations.


Scientific Reports | 2016

Identification of hepta-histidine as a candidate drug for Huntington's disease by in silico-in vitro- in vivo-integrated screens of chemical libraries

Tomomi Imamura; Kyota Fujita; Kazuhiko Tagawa; Teikichi Ikura; Xigui Chen; Hidenori Homma; Takuya Tamura; Ying Mao; Juliana Bosso Taniguchi; Kazumi Motoki; Makoto Nakabayashi; Nobutoshi Ito; Kazunori D. Yamada; Kentaro Tomii; Hideyuki Okano; Julia A. Kaye; Steven Finkbeiner; Hitoshi Okazawa

We identified drug seeds for treating Huntington’s disease (HD) by combining in vitro single molecule fluorescence spectroscopy, in silico molecular docking simulations, and in vivo fly and mouse HD models to screen for inhibitors of abnormal interactions between mutant Htt and physiological Ku70, an essential DNA damage repair protein in neurons whose function is known to be impaired by mutant Htt. From 19,468 and 3,010,321 chemicals in actual and virtual libraries, fifty-six chemicals were selected from combined in vitro-in silico screens; six of these were further confirmed to have an in vivo effect on lifespan in a fly HD model, and two chemicals exerted an in vivo effect on the lifespan, body weight and motor function in a mouse HD model. Two oligopeptides, hepta-histidine (7H) and Angiotensin III, rescued the morphological abnormalities of primary neurons differentiated from iPS cells of human HD patients. For these selected drug seeds, we proposed a possible common structure. Unexpectedly, the selected chemicals enhanced rather than inhibited Htt aggregation, as indicated by dynamic light scattering analysis. Taken together, these integrated screens revealed a new pathway for the molecular targeted therapy of HD.


bioRxiv | 2018

The cellular NMD pathway restricts Zika virus infection and is targeted by the viral capsid protein

Krystal A. Fontaine; Kristoffer E. Leon; Mir Mubashir Khalid; David Jimenez-Morales; Julia A. Kaye; Priya Shah; Steven Finkbeiner; Nevan J. Krogan; Melanie Ott

Zika virus (ZIKV) infection of neural progenitor cells (NPCs) in utero is associated with neurological disorders, such as microcephaly1–3, but a detailed molecular understanding of ZIKV-induced pathogenesis is lacking. Here we show that in vitro ZIKV infection of human cells, including NPCs, causes disruption of the nonsense-mediated mRNA decay (NMD) pathway. NMD is a cellular mRNA surveillance mechanism that is required for normal brain size in mice4–6. Using affinity purification-mass spectrometry, we identified multiple cellular NMD factors that bind to the viral capsid protein, including the central NMD regulator up-frameshift protein 1 (UPF1)7. Endogenous UPF1 interacted with the viral capsid protein in co-immunoprecipitation experiments, and capsid expression post-transcriptionally downregulated UPF1, a process that we confirmed occurs during de novo ZIKV infection. A further decrease in UPF1 levels by RNAi significantly enhanced ZIKV infection in NPC cultures. RNA electrophoretic mobility shift assays with UPF1-expressing cell lysates showed binding to ZIKV RNA in vitro, and UPF1 protein in ZIKV-infected NPCs colocalized with viral double-stranded RNA replication intermediates. Collectively, our data support a model where ZIKV, via the capsid protein, has evolved a strategy to dampen antiviral activities of NMD8,9, which subsequently contributes to neuropathology in vivo.


Journal of Neurology, Neurosurgery, and Psychiatry | 2012

A11 Induced pluripotent stem cells for basic and translational research on HD

Virginia B. Mattis; Soshana Svendsen; Allison D. Ebert; Clive N. Svendsen; Ar King; Malcolm Casale; Sara T. Winokur; G Batugedara; Marquis P. Vawter; Peter J. Donovan; Leslie F. Lock; Leslie M. Thompson; Y Zhu; Elisa Fossale; Ranjit S. Atwal; Tammy Gillis; Jayalakshmi S. Mysore; J-h Li; Ihn Sik Seong; Yiping Shen; X Chen; Vanessa C. Wheeler; Marcy E. MacDonald; James F. Gusella; S Akimov; Nicolas Arbez; T Juopperi; Tamara Ratovitski; Jh Chiang; Wr Kim

Background The expression of mutant HTT leads to many cellular alterations, including abnormal vesicle recycling, loss of signalling by brain-derived neurotrophic factor, excitotoxicity, perturbation of Ca2+ signalling, decreases in intracellular ATP, alterations of gene transcription, inhibition of protein clearance pathways, mitochondrial and metabolic disturbances, and ultimately cell death. While robust mammalian systems have been developed to model disease and extensive mechanistic insights have emerged, significant differences between rodent and human cells and between non-neuronal cells and neurons limit the utility of these models for accurately representing human disease. Human pluripotent stem cells can generate highly specified cell populations, including DARPP32-positive MSNs of the striatum, and provide a method for modelling HD in human neurons carrying the mutation. As it is caused by one single gene, HD is an ideal disorder for exploring the utility of modelling disease in induced pluripotent stem cells (iPSCs) through reprogramming adult cells from HD patients with known patterns of disease onset and duration. Aims Generate iPSC lines from HD patients and controls and identify CAG-repeat expansion associated phenotypes. Methods/techniques Through the efforts of an international consortium effort, 14 lines were generated, differentiated into neuronal populations and assessed for CAG-repeat dependent outcome measures. Results/outcomes HD iPSC lines have reproducible CAG expansion–associated phenotypes upon differentiation, including CAG expansion-associated changes in gene expression patterns and alterations in electrophysiology, metabolism, cell adhesion, and ultimately an increased risk of cell death. While the lines with the longest repeats (HD180) showed a phenotype across all assays, those with shorter repeats (HD60) showed phenotypes in a specific sub set of assays. The most sensitive assay for establishing repeat dependent effects was found to be calcium responses to stress. Conclusions This HD iPSC collection represents a unique and well-characterised resource to elucidate disease mechanisms in HD and provides a novel human stem cell platform for screening new candidate therapeutics. Funding NIH, CHDI, CIRM.

Collaboration


Dive into the Julia A. Kaye's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Malcolm Casale

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hidenori Homma

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Hitoshi Okazawa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Kazuhiko Tagawa

Tokyo Medical and Dental University

View shared research outputs
Researchain Logo
Decentralizing Knowledge