Network


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

Hotspot


Dive into the research topics where Jeniffer Chan is active.

Publication


Featured researches published by Jeniffer Chan.


Journal of Biological Chemistry | 2010

ATP-binding Cassette Transporter A1 Mediates the Beneficial Effects of the Liver X Receptor Agonist GW3965 on Object Recognition Memory and Amyloid Burden in Amyloid Precursor Protein/Presenilin 1 Mice

James Donkin; Sophie Stukas; Veronica Hirsch-Reinshagen; Dhananjay Namjoshi; Anna Wilkinson; Sharon May; Jeniffer Chan; Jianjia Fan; Jon L. Collins; Cheryl L. Wellington

The cholesterol transpoter ATP-binding cassette transporter A1 (ABCA1) moves lipids onto apolipoproteins including apolipoprotein E (apoE), which is the major cholesterol carrier in the brain and an established genetic risk factor for late-onset Alzheimer disease (AD). In amyloid mouse models of AD, ABCA1 deficiency exacerbates amyloidogenesis, whereas ABCA1 overexpression ameliorates amyloid load, suggesting a role for ABCA1 in Aβ metabolism. Agonists of liver X receptors (LXR), including GW3965, induce transcription of several genes including ABCA1 and apoE, and reduce Aβ levels and improve cognition in AD mice. However, the specific role of ABCA1 in mediating beneficial responses to LXR agonists in AD mice is unknown. We evaluated behavioral and neuropathogical outcomes in GW3965-treated female APP/PS1 mice with and without ABCA1. Treatment of APP/PS1 mice with GW3965 increased ABCA1 and apoE protein levels. ABCA1 was required to observe significantly elevated apoE levels in brain tissue and cerebrospinal fluid upon therapeutic (33 mg/kg/day) GW3965 treatment. At 33 mg/kg/day, GW3965 was also associated with a trend toward redistribution of Aβ to the carbonate-soluble pool independent of ABCA1. APP/PS1 mice treated with either 2.5 or 33 mg/kg/day of GW3965 showed a clear trend toward reduced amyloid burden in hippocampus and whole brain, whereas APP/PS1-treated mice lacking ABCA1 failed to display reduced amyloid load in the whole brain and showed trends toward increased hippocampal amyloid. Treatment of APP/PS1 mice with either dose of GW3965 completely restored novel object recognition memory to wild-type levels, which required ABCA1. These results suggest that ABCA1 contributes to several beneficial effects of the LXR agonist GW3965 in APP/PS1 mice.


Neurobiology of Disease | 2006

Elevated plasma triglyceride levels precede amyloid deposition in Alzheimer’s disease mouse models with abundant Aβ in plasma

Braydon L. Burgess; Sean McIsaac; Kathryn E. Naus; Jeniffer Chan; Gavin Tansley; Jing Yang; Fudan Miao; Colin Ross; Miranda Van Eck; Michael R. Hayden; William E. Van Nostrand; Peter St George-Hyslop; David Westaway; Cheryl L. Wellington

Dietary or pharmacological manipulation of plasma lipids markedly influences amyloid deposition in animal models of Alzheimers Disease (AD). However, it is not known whether baseline plasma lipids in AD models differ from wild-type littermates throughout the natural history of disease. To address this question, we measured plasma total cholesterol and triglyceride levels over time in three transgenic AD mouse models in the absence of dietary or pharmacological treatments. Total cholesterol levels were not significantly different between transgenic and wild-type mice during the development of AD neuropathology in all models tested. In contrast, elevated very-low-density lipoprotein (VLDL) triglyceride levels preceded amyloid deposition in two AD models with abundant plasma A beta. Elevated triglycerides were not accompanied by increased inflammatory markers nor decreased lipase activity, but were associated with a significant 30% increase in VLDL-triglyceride secretion rate. Our results suggest that the presence of A beta in plasma may affect peripheral lipid metabolism early in AD pathogenesis.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2008

Overexpression of Human ABCG1 Does Not Affect Atherosclerosis in Fat-Fed ApoE-Deficient Mice

Braydon L. Burgess; Kathryn E. Naus; Jeniffer Chan; Veronica Hirsch-Reinshagen; Gavin Tansley; Lisa Matzke; Benny Chan; Anna Wilkinson; Jianjia Fan; James Donkin; Danielle Balik; Tracie Tanaka; George Ou; Roger A. Dyer; Sheila M. Innis; Bruce M. McManus; Dieter Lütjohann; Cheryl L. Wellington

Objective—The purpose of this study was to evaluate the effects of whole body overexpression of human ABCG1 on atherosclerosis in apoE−/− mice. Methods and Results—We generated BAC transgenic mice in which human ABCG1 is expressed from endogenous regulatory signals, leading to a 3- to 7-fold increase in ABCG1 protein across various tissues. Although the ABCG1 BAC transgene rescued lung lipid accumulation in ABCG1−/− mice, it did not affect plasma lipid levels, macrophage cholesterol efflux to HDL, atherosclerotic lesion area in apoE−/− mice, or levels of tissue cholesterol, cholesterol ester, phospholipids, or triglycerides. Subtle changes in sterol biosynthetic intermediate levels were observed in liver, with chow-fed ABCG1 BAC Tg mice showing a nonsignificant trend toward decreased levels of lathosterol, lanosterol, and desmosterol, and fat-fed mice exhibiting significantly elevated levels of each intermediate. These changes were insufficient to alter ABCA1 expression in liver. Conclusions—Transgenic human ABCG1 does not influence atherosclerosis in apoE−/− mice but may participate in the regulation of tissue cholesterol biosynthesis.


Journal of Lipid Research | 2007

The cholesterol transporter ABCG1 modulates the subcellular distribution and proteolytic processing of β-amyloid precursor protein

Gavin Tansley; Braydon L. Burgess; Matt T. Bryan; Yuan Su; Veronica Hirsch-Reinshagen; Jonathan Pearce; Jeniffer Chan; Anna Wilkinson; Jeanette Evans; Kathryn E. Naus; Sean McIsaac; Kelley Bromley; Weihong Song; Hsui-Chiung Yang; Nan Wang; Ronald B. DeMattos; Cheryl L. Wellington

Although intracellular cholesterol levels are known to influence the proteolysis of β-amyloid precursor protein (APP), the effect of specific genes that regulate cholesterol metabolism on APP processing remains poorly understood. The cholesterol transporter ABCG1 facilitates cholesterol efflux to HDL and is expressed in brain. Notably, the human ABCG1 gene maps to chromosome 21q22.3, and individuals with Down syndrome (DS) typically manifest with Alzheimers disease (AD) neuropathology in their 30s. Here, we demonstrate that expression of ABCG1 enhances amyloid-β protein (Aβ) production in transfected HEK cells in a manner that requires functional cholesterol transporter activity. ABCG1-expressing cells also exhibit increased secreted APP (sAPP)α and sAPPβ secretion and display increased cell surface-associated APP. These results suggest that ABCG1 increases the availability of APP as a secretase substrate for both the amyloidogenic and nonamyloidogenic pathways. In vivo, ABCG1 mRNA levels are 2-fold more abundant in DS brain compared with age- and sex-matched normal controls. Finally, both Aβ and sAPPα levels are increased in DS cortex relative to normal controls. These findings suggest that altered cholesterol metabolism and APP trafficking mediated by ABCG1 may contribute to the accelerated onset of AD neuropathology in DS.


Journal of the American Heart Association | 2014

Intravenously Injected Human Apolipoprotein A‐I Rapidly Enters the Central Nervous System via the Choroid Plexus

Sophie Stukas; Jérôme Robert; Michael Lee; Iva Kulic; Michael Carr; Katherine Tourigny; Jianjia Fan; Dhananjay Namjoshi; Kalistyne Lemke; Nicole DeValle; Jeniffer Chan; Tammy Wilson; Anna Wilkinson; Rafi Chapanian; Jayachandran N. Kizhakkedathu; John R. Cirrito; Michael N. Oda; Cheryl L. Wellington

Background Brain lipoprotein metabolism is dependent on lipoprotein particles that resemble plasma high‐density lipoproteins but that contain apolipoprotein (apo) E rather than apoA‐I as their primary protein component. Astrocytes and microglia secrete apoE but not apoA‐I; however, apoA‐I is detectable in both cerebrospinal fluid and brain tissue lysates. The route by which plasma apoA‐I enters the central nervous system is unknown. Methods and Results Steady‐state levels of murine apoA‐I in cerebrospinal fluid and interstitial fluid are 0.664 and 0.120 μg/mL, respectively, whereas brain tissue apoA‐I is ≈10% to 15% of its levels in liver. Recombinant, fluorescently tagged human apoA‐I injected intravenously into mice localizes to the choroid plexus within 30 minutes and accumulates in a saturable, dose‐dependent manner in the brain. Recombinant, fluorescently tagged human apoA‐I accumulates in the brain for 2 hours, after which it is eliminated with a half‐life of 10.3 hours. In vitro, human apoA‐I is specifically bound, internalized, and transported across confluent monolayers of primary human choroid plexus epithelial cells and brain microvascular endothelial cells. Conclusions Following intravenous injection, recombinant human apoA‐I rapidly localizes predominantly to the choroid plexus. Because apoA‐I mRNA is undetectable in murine brain, our results suggest that plasma apoA‐I, which is secreted from the liver and intestine, gains access to the central nervous system primarily by crossing the blood–cerebrospinal fluid barrier via specific cellular mediated transport, although transport across the blood–brain barrier may also contribute to a lesser extent.


Biochimica et Biophysica Acta | 2012

The LXR agonist GW3965 increases apoA-I protein levels in the central nervous system independent of ABCA1.

Sophie Stukas; Sharon May; Anna Wilkinson; Jeniffer Chan; James Donkin; Cheryl L. Wellington

Lipoprotein metabolism in the central nervous system (CNS) is based on high-density lipoprotein-like particles that use apoE as their predominant apolipoprotein rather than apoA-I. Although apoA-I is not expressed in astrocytes and microglia, which produce CNS apoE, apoA-I is reported to be expressed in porcine brain capillary endothelial cells and also crosses the blood-brain barrier (BBB). These mechanisms allow apoA-I to reach concentrations in cerebrospinal fluid (CSF) that are approximately 0.5% of its plasma levels. Recently, apoA-I has been shown to enhance cognitive function and reduce cerebrovascular amyloid deposition in Alzheimers Disease (AD) mice, raising questions about the regulation and function of apoA-I in the CNS. Peripheral apoA-I metabolism is highly influenced by ABCA1, but less is known about how ABCA1 regulates CNS apoA-I. We report that ABCA1 deficiency leads to greater retention of apoA-I in the CNS than in the periphery. Additionally, treatment of symptomatic AD mice with GW3965, an LXR agonist that stimulates ABCA1 expression, increases apoA-I more dramatically in the CNS compared to the periphery. Furthermore, GW3965-mediated up-regulation of CNS apoA-I is independent of ABCA1. Our results suggest that apoA-I may be regulated by distinct mechanisms on either side of the BBB and that apoA-I may serve to integrate peripheral and CNS lipid metabolism. This article is part of a Special Issue entitled Advances in High Density Lipoprotein Formation and Metabolism: A Tribute to John F. Oram (1945-2010).


Journal of Lipid Research | 2013

Hormonal modulators of glial ABCA1 and apoE levels

Jianjia Fan; Yoko Shimizu; Jeniffer Chan; Anna Wilkinson; Ayaka Ito; Peter Tontonoz; Edie Dullaghan; Liisa A.M. Galea; Tom A. Pfeifer; Cheryl L. Wellington

Apolipoprotein E (apoE) is the major lipid carrier in the central nervous system. As apoE plays a major role in the pathogenesis of Alzheimer disease (AD) and also mediates repair pathways after several forms of acute brain injury, modulating the expression, secretion, or function of apoE may provide potential therapeutic approaches for several neurological disorders. Here we show that progesterone and a synthetic progestin, lynestrenol, significantly induce apoE secretion from human CCF-STTG1 astrocytoma cells, whereas estrogens and the progesterone metabolite allopregnanolone have negligible effects. Intriguingly, lynestrenol also increases expression of the cholesterol transporter ABCA1 in CCF-STTG1 astrocytoma cells, primary murine glia, and immortalized murine astrocytes that express human apoE3. The progesterone receptor inhibitor RU486 attenuates the effect of progestins on apoE expression in CCF-STTG1 astrocytoma cells but has no effect on ABCA1 expression in all glial cell models tested, suggesting that the progesterone receptor (PR) may participate in apoE but does not affect ABCA1 regulation.These results suggest that selective reproductive steroid hormones have the potential to influence glial lipid homeostasis through liver X receptor-dependent and progesterone receptor-dependent pathways.


Journal of Lipid Research | 2014

LCAT deficiency does not impair amyloid metabolism in APP/PS1 mice

Sophie Stukas; Lita Freeman; Michael Lee; Anna Wilkinson; Alice Ossoli; Boris Vaisman; Stephen J. Demosky; Jeniffer Chan; Veronica Hirsch-Reinshagen; Alan T. Remaley; Cheryl L. Wellington

A key step in plasma HDL maturation from discoidal to spherical particles is the esterification of cholesterol to cholesteryl ester, which is catalyzed by LCAT. HDL-like lipoproteins in cerebrospinal fluid (CSF) are also spherical, whereas nascent lipoprotein particles secreted from astrocytes are discoidal, suggesting that LCAT may play a similar role in the CNS. In plasma, apoA-I is the main LCAT activator, while in the CNS, it is believed to be apoE. apoE is directly involved in the pathological progression of Alzheimer’s disease, including facilitating β-amyloid (Aβ) clearance from the brain, a function that requires its lipidation by ABCA1. However, whether apoE particle maturation by LCAT is also required for Aβ clearance is unknown. Here we characterized the impact of LCAT deficiency on CNS lipoprotein metabolism and amyloid pathology. Deletion of LCAT from APP/PS1 mice resulted in a pronounced decrease of apoA-I in plasma that was paralleled by decreased apoA-I levels in CSF and brain tissue, whereas apoE levels were unaffected. Furthermore, LCAT deficiency did not increase Aβ or amyloid in APP/PS1 LCAT−/− mice. Finally, LCAT expression and plasma activity were unaffected by age or the onset of Alzheimer’s-like pathology in APP/PS1 mice. Taken together, these results suggest that apoE-containing discoidal HDLs do not require LCAT-dependent maturation to mediate efficient Aβ clearance.


Alzheimers & Dementia | 2006

P1-053: Alzheimer’s disease neuropathology is not mitigated by the physiological expression of human ABCA1 in APP/PS1 mice

Veronica Hirsch-Reinshagen; Jeniffer Chan; Sean McIsaac; Kathryn E. Naus; Luis F. Maia; Braydon L. Burgess; Roshni R. Singaraja; Michael R. Hayden; Cheryl L. Wellington

a reduction in synapse density per se is unlikely to underlie the cognitive deficits present in TASTPM mice from 6 months of age and the presence of -amyloid plaques may not be sufficient to cause synaptic die-back. However, the current morphological analysis could not elucidate the functional status of synapses. Moreover, at 6 10 months of age synaptic sprouting contemporaneous with a degenerative process may be sufficient to maintain synaptic density but not synaptic function and synapse loss in TASTPM mice may eventually be manifest at greater ages.


Journal of Biological Chemistry | 2004

Deficiency of ABCA1 impairs apolipoprotein E metabolism in brain.

Veronica Hirsch-Reinshagen; Steven Zhou; Braydon L. Burgess; Lise Bernier; Sean McIsaac; Jeniffer Chan; Gavin Tansley; Jeffrey S. Cohn; Michael R. Hayden; Cheryl L. Wellington

Collaboration


Dive into the Jeniffer Chan's collaboration.

Top Co-Authors

Avatar

Cheryl L. Wellington

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Anna Wilkinson

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Braydon L. Burgess

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sean McIsaac

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Sophie Stukas

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

James Donkin

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Jianjia Fan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Gavin Tansley

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Kathryn E. Naus

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge