Network


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

Hotspot


Dive into the research topics where Kathryn Chase is active.

Publication


Featured researches published by Kathryn Chase.


Neuron | 1995

Huntingtin is a cytoplasmic protein associated with vesicles in human and rat brain neurons.

Marian DiFiglia; Ellen Sapp; Kathryn Chase; Cordula Schwarz; Alison R. Meloni; Christine Young; Eileen J. Martin; Jean-Paul Vonsattel; Robert E. Carraway; Steven A Reeves; Frederick M. Boyce; Neil Aronin

The gene defective in Huntingtons disease encodes a protein, huntingtin, with unknown function. Antisera generated against three separate regions of huntingtin identified a single high molecular weight protein of approximately 320 kDa on immunoblots of human neuroblastoma extracts. The same protein species was detected in human and rat cortex synaptosomes and in sucrose density gradients of vesicle-enriched fractions, where huntingtin immunoreactivity overlapped with the distribution of vesicle membrane proteins (SV2, transferrin receptor, and synaptophysin). Immunohistochemistry in human and rat brain revealed widespread cytoplasmic labeling of huntingtin within neurons, particularly cell bodies and dendrites, rather than the more selective pattern of axon terminal labeling characteristic of many vesicle-associated proteins. At the ultrastructural level, immunoreactivity in cortical neurons was detected in the matrix of the cytoplasm and around the membranes of the vesicles. The ubiquitous cytoplasmic distribution of huntingtin in neurons and its association with vesicles suggest that huntingtin may have a role in vesicle trafficking.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Therapeutic silencing of mutant huntingtin with siRNA attenuates striatal and cortical neuropathology and behavioral deficits

Marian DiFiglia; Miguel Sena-Esteves; Kathryn Chase; Ellen Sapp; Edith L. Pfister; Meghan Sass; Jennifer Yoder; Patrick B. Reeves; Rajendra K. Pandey; Kallanthottathil G. Rajeev; Muthiah Manoharan; Dinah Sah; Phillip D. Zamore; Neil Aronin

Huntingtons disease (HD) is a neurodegenerative disorder caused by expansion of a CAG repeat in the huntingtin (Htt) gene. HD is autosomal dominant and, in theory, amenable to therapeutic RNA silencing. We introduced cholesterol-conjugated small interfering RNA duplexes (cc-siRNA) targeting human Htt mRNA (siRNA-Htt) into mouse striata that also received adeno-associated virus containing either expanded (100 CAG) or wild-type (18 CAG) Htt cDNA encoding huntingtin (Htt) 1–400. Adeno-associated virus delivery to striatum and overlying cortex of the mutant Htt gene, but not the wild type, produced neuropathology and motor deficits. Treatment with cc-siRNA-Htt in mice with mutant Htt prolonged survival of striatal neurons, reduced neuropil aggregates, diminished inclusion size, and lowered the frequency of clasping and footslips on balance beam. cc-siRNA-Htt was designed to target human wild-type and mutant Htt and decreased levels of both in the striatum. Our findings indicate that a single administration into the adult striatum of an siRNA targeting Htt can silence mutant Htt, attenuate neuronal pathology, and delay the abnormal behavioral phenotype observed in a rapid-onset, viral transgenic mouse model of HD.


Experimental Neurology | 1998

Wild-Type and Mutant Huntingtins Function in Vesicle Trafficking in the Secretory and Endocytic Pathways☆

James Velier; Manho Kim; Cordula Schwarz; Tae Wan Kim; Ellen Sapp; Kathryn Chase; Neil Aronin; Marian DiFiglia

Huntingtin is a cytoplasmic protein that is found in neurons and somatic cells. In patients with Huntingtons disease (HD), the NH2-terminal region of huntingtin has an expanded polyglutamine tract. An abnormal protein interaction by mutant huntingtin has been proposed as a mechanism for HD pathogenesis. Huntingtin associates with vesicle membranes and interacts with proteins involved in vesicle trafficking. It is unclear where along vesicle transport pathways wild-type and mutant huntingtins are found and whether polyglutamine expansion affects this localization. To distinguish wild-type and mutant huntingtin, fibroblasts from normals and HD patients with two mutant alleles (homozygotes) were examined. Immunofluorescence confocal microscopy showed that mutant huntingtin localized with clathrin in membranes of the trans Golgi network and in clathrin-coated and noncoated endosomal vesicles in the cytoplasm and along plasma membranes. Separation of organelles in Nycodenz gradients showed that in normal and HD homozygote patient cells, huntingtin was present in membrane fractions enriched in clathrin. Similar results were obtained in fibroblasts from heterozyote juvenile HD patients who had a highly expanded polyglutamine tract in the HD allele. Western blot analysis of membrane fractions from rat brain showed that wild-type huntingtin was present in fractions that contained purified clathrin-coated membranes or a mixture of clathrin-coated and noncoated membranes. Electron microscopy of huntingtin immunoreactivity in rat brain revealed labeling along dendritic plasma membranes in association with clathrin-coated pits and clusters of noncoated endosomal vesicles 40-60 nm in diameter. These data suggest that wild-type and mutant huntingtin can influence vesicle transport in the secretory and endocytic pathways through associations with clathrin-coated vesicles.


Neuron | 1995

CAG EXPANSION AFFECTS THE EXPRESSION OF MUTANT HUNTINGTIN IN THE HUNTINGTON'S DISEASE BRAIN

Neil Aronin; Kathryn Chase; Christine Young; Ellen Sapp; Cordula Schwarz; Nahida Matta; Ruth Kornreich; Bernhard Lanwehrmeyer; Edward D. Bird; M. Flint Beal; Jean-Paul Vonsattel; Thomas J. Smith; Robert E. Carraway; Frederick M. Boyce; Anne B. Young; John B. Penney; Marian DiFiglia

A trinucleotide repeat (CAG) expansion in the huntingtin gene causes Huntingtons disease (HD). In brain tissue from HD heterozygotes with adult onset and more clinically severe juvenile onset, where the largest expansions occur, a mutant protein of equivalent intensity to wild-type huntingtin was detected in cortical synaptosomes, indicating that a mutant species is synthesized and transported with the normal protein to nerve endings. The increased size of mutant huntingtin relative to the wild type was highly correlated with CAG repeat expansion, thereby linking an altered electrophoretic mobility of the mutant protein to its abnormal function. Mutant huntingtin appeared in gray and white matter with no difference in expression in affected regions. The mutant protein was broader than the wild type and in 6 of 11 juvenile cases resolved as a complex of bands, consistent with evidence at the DNA level for somatic mosaicism. Thus, HD pathogenesis results from a gain of function by an aberrant protein that is widely expressed in brain and is harmful only to some neurons.


Neuroreport | 1997

Fast transport and retrograde movement of huntingtin and HAP 1 in axons

Jessie Block-Galarza; Kathryn Chase; Ellen Sapp; Kevin T. Vaughn; Richard B. Vallee; Marian DiFiglia; Neil Aronin

HUNTINGTIN, the protein product of the Huntingtons disease gene, associates with vesicle membranes and microtubules in neurons. Analysis of axonal transport with a stop-flow, double crush ligation approach in rat sciatic nerve showed that full length huntingtin (350 kDa) and an N-terminal cleavage product (50 kD) were increased within 6–12 h on both the proximal and distal sides of the crush site when compared with normal unligated nerve. The huntingtin associated protein HAP 1 and the retrograde motor protein dynein also accumulated on both sides of the crush, whereas the vesicle docking protein SNAP-25 was elevated only proximally. The cytoskeletal protein a-tubulin was unaffected. The rapid anterograde accumulation of huntingtin and HAP 1 is compatible with their axonal transport on vesicular membranes. Retrograde movement of both proteins, as seen by accumulation distal to the nerve crush, may be necessary for their degradation at the soma or for a function in retrograde membrane trafficking.


Neuroscience | 1991

N-methyl-d-aspartate receptor activation in the neostriatum increases c-fos and fos-related antigens selectively in medium-sized neurons

Neil Aronin; Kathryn Chase; S.M. Sagar; F.R. Sharp; Marian DiFiglia

In the neostriatum a selective loss of neurons occurs following exposure to N-methyl-D-aspartate receptor agonists. One hypothesis emerging from this observation is that an excitotoxic process via N-methyl-D-aspartate receptors may contribute to the pathogenesis of Huntingtons disease, which is characterized by the loss of medium-sized neurons. However, whether there is a selective distribution of N-methyl-D-aspartate receptors in specific populations of neostriatal neurons is unknown. In this study the expression of c-fos mRNA and protein was used to examine the response of neostriatal cells to N-methyl-D-aspartate receptor stimulation in the rat. After intrastriatal injection of the N-methyl-D-aspartate receptor agonist, quinolinic acid, an increase in c-fos mRNA concentrations was detected using in situ hybridization and Northern blot analysis. Western blot analysis showed that not only the c-Fos mRNA protein product but also other Fos-related antigens capable of binding to DNA were increased in response to N-methyl-D-aspartate receptor activation. The selectivity of the neuronal response to N-methyl-D-aspartate receptor activation was examined immunohistochemically at the light and ultrastructural levels. Our results indicate that N-methyl-D-aspartate receptor activation by quinolinic acid stimulates medium spiny neurons to increase c-Fos expression; to a lesser extent, medium aspiny interneurons and glial cells also respond. In contrast, negligible change in c-Fos expression is observed in large neurons. These results are consistent with other evidence that medium-sized spiny neurons are preferentially vulnerable to the toxic effects of excitatory amino acids acting at N-methyl-D-aspartate receptors. An additional implication of these findings is that activation of the N-methyl-D-aspartate receptor in medium spiny neurons leads to increased expression of candidate AP-1 transcription factors, thereby coupling the N-methyl-D-aspartate receptor and regulation of gene expression in signal transduction processes of the neostriatal medium spiny neuron.


Molecular and Cellular Biology | 2009

Mutant Huntingtin Impairs Vesicle Formation from Recycling Endosomes by Interfering with Rab11 Activity

Xueyi Li; Clive Standley; Ellen Sapp; Antonio Valencia; Zheng-Hong Qin; Kimberly B. Kegel; Jennifer Yoder; Laryssa A. Comer-Tierney; Miguel Esteves; Kathryn Chase; Jonathan Alexander; Nicholas Masso; Lindsay Sobin; Karl D. Bellve; Richard A. Tuft; Lawrence M. Lifshitz; Kevin E. Fogarty; Neil Aronin; Marian DiFiglia

ABSTRACT Huntingtin (Htt) localizes to endosomes, but its role in the endocytic pathway is not established. Recently, we found that Htt is important for the activation of Rab11, a GTPase involved in endosomal recycling. Here we studied fibroblasts of healthy individuals and patients with Huntingtons disease (HD), which is a movement disorder caused by polyglutamine expansion in Htt. The formation of endocytic vesicles containing transferrin at plasma membranes was the same in control and HD patient fibroblasts. However, HD fibroblasts were delayed in recycling biotin-transferrin back to the plasma membrane. Membranes of HD fibroblasts supported less nucleotide exchange on Rab11 than did control membranes. Rab11-positive vesicular and tubular structures in HD fibroblasts were abnormally large, suggesting that they were impaired in forming vesicles. We used total internal reflection fluorescence imaging of living fibroblasts to monitor fluorescence-labeled transferrin-carrying transport intermediates that emerged from recycling endosomes. HD fibroblasts had fewer small vesicles and more large vesicles and long tubules than did control fibroblasts. Dominant active Rab11 expressed in HD fibroblasts normalized the recycling of biotin-transferrin. We propose a novel mechanism for cellular dysfunction by the HD mutation arising from the inhibition of Rab11 activity and a deficit in vesicle formation at recycling endosomes.


Journal of Neuroscience Research | 2010

Mutant Huntingtin and Glycogen Synthase Kinase 3-β Accumulate in Neuronal Lipid Rafts of a Presymptomatic Knock-In Mouse Model of Huntington's Disease

Antonio Valencia; Patrick B. Reeves; Ellen Sapp; Xueyi Li; Jonathan Alexander; Kimberly B. Kegel; Kathryn Chase; Neil Aronin; Marian DiFiglia

Patients with Huntingtons disease have an expanded polyglutamine tract in huntingtin and suffer severe brain atrophy and neurodegeneration. Because membrane dysfunction can occur in Huntingtons disease, we addressed whether mutant huntingtin in brain and primary neurons is present in lipid rafts, which are cholesterol‐enriched membrane domains that mediate growth and survival signals. Biochemical analysis of detergent‐resistant membranes from brains and primary neurons of wild‐type and presymptomatic Huntingtons disease knock‐in mice showed that wild‐type and mutant huntingtin were recovered in lipid raft‐enriched detergent‐resistant membranes. The association with lipid rafts was stronger for mutant huntingtin than wild‐type huntingtin. Lipid rafts extracted from Huntingtons disease mice had normal levels of lipid raft markers (Gαq, Ras, and flotillin) but significantly more glycogen synthase kinase 3‐β. Increases in glycogen synthase kinase 3‐β have been associated with apoptotic cell death. Treating Huntingtons disease primary neurons with inhibitors of glycogen synthase kinase 3‐β reduced neuronal death. We speculate that accumulation of mutant huntingtin and glycogen synthase kinase 3‐β in lipid rafts of presymptomatic Huntingtons disease mouse neurons contributes to neurodegeneration in Huntingtons disease.


Neurobiology of Disease | 2009

Disruption of Rab11 activity in a knock-in mouse model of Huntington's Disease

Xueyi Li; Ellen Sapp; Kathryn Chase; Laryssa A. Comer-Tierney; Nicholas Masso; Jonathan Alexander; Patrick Reeves; Kimberly B. Kegel; Antonio Valencia; Miguel Esteves; Neil Aronin; Marian DiFiglia

The Huntingtons disease (HD) mutation causes polyglutamine expansion in huntingtin (Htt) and neurodegeneration. Htt interacts with a complex containing Rab11GDP and is involved in activation of Rab11, which functions in endosomal recycling and neurite growth and long-term potentiation. Like other Rab proteins, Rab11GDP undergoes nucleotide exchange to Rab11GTP for its activation. Here we show that striatal membranes of HD(140Q/140Q) knock-in mice are impaired in supporting conversion of Rab11GDP to Rab11GTP. Dominant negative Rab11 expressed in the striatum and cortex of normal mice caused neuropathology and motor dysfunction, suggesting that a deficiency in Rab11 activity is pathogenic in vivo. Primary cortical neurons from HD(140Q/140Q) mice were delayed in recycling transferrin receptors back to the plasma membrane. Partial rescue from glutamate-induced cell death occurred in HD neurons expressing dominant active Rab11. We propose a novel mechanism of HD pathogenesis arising from diminished Rab11 activity at recycling endosomes.


Experimental Neurology | 2002

Maintenance of susceptibility to neurodegeneration following intrastriatal injections of quinolinic acid in a new transgenic mouse model of Huntington's disease.

Åsa Petersén; Kathryn Chase; Zoe Puschban; Marian DiFiglia; Patrik Brundin; Neil Aronin

A transgenic mouse model of Huntingtons disease (R6/1 and R6/2 lines) expressing exon 1 of the HD gene with 115-150 CAG repeats resisted striatal damage following injection of quinolinic acid and other neurotoxins. We examined whether excitotoxin resistance characterizes mice with mutant huntingtin transgenes. In a new transgenic mouse with 3 kb of mutant human huntingtin cDNA with 18, 46, or 100 CAG repeats, we found no change in susceptibility to intrastriatal injections of the excitotoxin quinolinic acid, compared to wild-type littermates. The new transgenic mice were injected with the same dose of quinolinic acid (30 nmol) as had been the R6 mice. Our findings highlight the importance of studying pathogenetic mechanisms in different transgenic models of a disease.

Collaboration


Dive into the Kathryn Chase's collaboration.

Top Co-Authors

Avatar

Neil Aronin

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edith L. Pfister

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lori A. Kennington

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Miguel Sena-Esteves

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Petr Vodicka

Academy of Sciences of the Czech Republic

View shared research outputs
Researchain Logo
Decentralizing Knowledge