Network


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

Hotspot


Dive into the research topics where Yijun Pan is active.

Publication


Featured researches published by Yijun Pan.


Molecular Pharmaceutics | 2015

Fatty Acid-Binding Protein 5 Facilitates the Blood-Brain Barrier Transport of Docosahexaenoic Acid.

Yijun Pan; Martin J. Scanlon; Yuji Owada; Yui Yamamoto; Christopher J. H. Porter; Joseph A. Nicolazzo

The brain has a limited ability to synthesize the essential polyunsaturated fatty acid (PUFA) docosahexaenoic acid (DHA) from its omega-3 fatty acid precursors. Therefore, to maintain brain concentrations of this PUFA at physiological levels, plasma-derived DHA must be transported across the blood-brain barrier (BBB). While DHA is able to partition into the luminal membrane of brain endothelial cells, its low aqueous solubility likely limits its cytosolic transfer to the abluminal membrane, necessitating the requirement of an intracellular carrier protein to facilitate trafficking of this PUFA across the BBB. As the intracellular carrier protein fatty acid-binding protein 5 (FABP5) is expressed at the human BBB, the current study assessed the putative role of FABP5 in the brain endothelial cell uptake and BBB transport of DHA in vitro and in vivo, respectively. hFAPB5 was recombinantly expressed and purified from Escherichia coli C41(DE3) cells and the binding affinity of DHA to hFABP5 assessed using isothermal titration calorimetry. The impact of FABP5 siRNA on uptake of (14)C-DHA into immortalized human brain microvascular endothelial (hCMEC/D3) cells was assessed. An in situ transcardiac perfusion method was optimized in C57BL/6 mice and subsequently used to compare the BBB influx rate (Kin) of (14)C-DHA between FABP5-deficient (FABP5(-/-)) and wild-type (FABP5(+/+)) C57BL/6 mice. DHA bound to hFABP5 with an equilibrium dissociation constant of 155 ± 8 nM (mean ± SEM). FABP5 siRNA transfection decreased hCMEC/D3 mRNA and protein expression of FABP5 by 53.2 ± 5.5% and 44.8 ± 13.7%, respectively, which was associated with a 14.1 ± 2.7% reduction in (14)C-DHA cellular uptake. By using optimized conditions for the in situ transcardiac perfusion (a 1 min preperfusion (10 mL/min) followed by perfusion of (14)C-DHA (1 min)), the Kin of (14)C-DHA was 0.04 ± 0.01 mL/g/s. Relative to FABP5(+/+) mice, the Kin of (14)C-DHA decreased 36.7 ± 12.4% in FABP5(-/-) mice. This study demonstrates that FABP5 binds to DHA and is involved in the brain endothelial cell uptake and subsequent BBB transport of DHA, confirming the importance of this cytoplasmic carrier protein in the CNS exposure of this PUFA essential for neuronal function.


The Journal of Neuroscience | 2016

Fatty Acid-Binding Protein 5 at the Blood-Brain Barrier Regulates Endogenous Brain Docosahexaenoic Acid Levels and Cognitive Function.

Yijun Pan; Jennifer L. Short; Kwok Ho Christopher Choy; Annie X. Zeng; Philip J. Marriott; Yuji Owada; Martin J. Scanlon; Christopher J. H. Porter; Joseph A. Nicolazzo

Fatty acid-binding protein 5 (FABP5) at the blood–brain barrier contributes to the brain uptake of docosahexaenoic acid (DHA), a blood-derived polyunsaturated fatty acid essential for maintenance of cognitive function. Given the importance of DHA in cognition, the aim of this study was to investigate whether deletion of FABP5 results in cognitive dysfunction and whether this is associated with reduced brain endothelial cell uptake of exogenous DHA and subsequent attenuation in the brain levels of endogenous DHA. Cognitive function was assessed in male and female FABP5+/+ and FABP5−/− mice using a battery of memory paradigms. FABP5−/− mice exhibited impaired working memory and short-term memory, and these cognitive deficits were associated with a 14.7 ± 5.7% reduction in endogenous brain DHA levels. The role of FABP5 in the blood–brain barrier transport of DHA was assessed by measuring 14C-DHA uptake into brain endothelial cells and capillaries isolated from FABP5+/+ and FABP5−/− mice. In line with a crucial role of FABP5 in the brain uptake of DHA, 14C-DHA uptake into brain endothelial cells and brain capillaries of FABP5−/− mice was reduced by 48.4 ± 14.5% and 14.0 ± 4.2%, respectively, relative to those of FABP5+/+ mice. These results strongly support the hypothesis that FABP5 is essential for maintaining brain endothelial cell uptake of DHA, and that cognitive deficits observed in FABP5−/− mice are associated with reduced CNS access of DHA. SIGNIFICANCE STATEMENT Genetic deletion of fatty acid-binding protein 5 (FABP5) in mice reduces uptake of exogenous docosahexaenoic acid (DHA) into brain endothelial cells and brain capillaries and reduces brain parenchymal levels of endogenous DHA. Therefore, FABP5 in the brain endothelial cell is a crucial contributor to the brain levels of DHA. Critically, lowered brain DHA levels in FABP5−/− mice occurred in tandem with cognitive deficits in a battery of memory paradigms. This study provides evidence of a critical role for FABP5 in the maintenance of cognitive function via regulating the brain uptake of DHA, and suggests that upregulation of FABP5 in neurodegenerative diseases, where brain DHA levels are possibly diminished (e.g., Alzheimers disease), may provide a novel therapeutic approach for restoring cognitive function.


Current Clinical Pharmacology | 2015

The Impact of Docosahexaenoic Acid on Alzheimer’s Disease: Is There a Role of the Blood-Brain Barrier?

Yijun Pan; Hanan Khalil; Joseph A. Nicolazzo

There currently is no cure or established preventative treatment for Alzheimers disease (AD). Considering the increasing aging population and the subsequent high prevalence of AD worldwide, identifying a cost-effective way to prevent AD is an essential unmet medical need. Relative to healthy human brain samples, postmortem AD brain samples have been shown to exhibit lower docosahexaenoic acid (DHA) levels, an essential polyunsaturated fatty acid required for normal neuronal function. However, findings from different studies are controversial and it is not clear whether this alteration in DHA brain levels is a cause or consequence of AD. Animal studies have also demonstrated that administration of DHA can alleviate the underlying pathophysiology of AD, including but not limited to amyloid pathology, tau pathology, and neuroinflammation. Moreover, DHA has been suggested to exert cognitive-enhancing effects and epidemiological studies have suggested that regular consumption of fish or omega-3 fatty acid enriched diets can attenuate the cognitive decline in AD and/or lower the risk of developing AD. However, the beneficial effects of DHA in AD have not been clearly demonstrated by current human randomised-control trials. In addition, the underlying reasons for the lower brain levels of DHA in AD remain to be fully characterised. However, given that the brain has limited capacity to produce DHA de novo and obtains DHA from the plasma, one plausible explanation for the lower brain levels of DHA in AD is reduced bloodbrain barrier (BBB) transport of this fatty acid in AD, as has been reported in one mouse model of AD. Unfortunately, the actual mechanisms governing the BBB transport of DHA in healthy conditions are not clearly understood, complicating the relationship between reduced BBB transport of DHA, attenuated DHA brain levels and AD pathology. The purpose of this review, therefore, is to summarise the findings of the biochemical, functional and epidemiological studies assessing the impact of DHA on the progression of AD, with a focus on how brain DHA levels alter in AD, the mechanisms thought to be held responsible for the apparent protective effects of DHA in AD, and the factors governing BBB transport of DHA in AD.


Journal of Pharmaceutical Sciences | 2014

Exploiting the buccal mucosa as an alternative route for the delivery of donepezil hydrochloride.

Thiago Caon; Yijun Pan; Cláudia Maria Oliveira Simões; Joseph A. Nicolazzo

The potential of the buccal mucosa as an alternative route for the systemic delivery of donepezil (DPZ) hydrochloride, and the impact of various skin penetration enhancers on DPZ buccal permeability, was assessed using an in vitro model. DPZ was applied to porcine buccal mucosa in modified Ussing chambers either alone (20 μg/mL) or with different treatment protocols of various enhancers including Azone® (AZ), deoxycholic acid (DA), polyethylene glycol (PEG) 400, and oleic acid (OA)-PEG 400. DPZ permeated the buccal mucosa very rapidly with a permeability coefficient of 35.6 ± 4.9 × 10(-6) cm/s, which was not significantly affected by AZ pretreatment. Coapplication of DA 0.6% (w/w), but not DA 0.01% (w/w), reduced the buccal permeation of DPZ (3.5-fold), and PEG 400 reduced the absorption of DPZ in a dose-dependent manner (1.6- and 18.0-fold reduction at 5% and 50%, w/w, PEG 400, respectively). Coapplication of a combination of OA 1% (v/w) and PEG 400 5% (w/w) further reduced DPZ permeability (5.5-fold), which was demonstrated to result from excipient-induced DPZ precipitation as assessed by light microscopy analysis. These results confirm the feasibility of a novel buccal delivery system for Alzheimers disease, and suggest various approaches that may be exploited for controlled buccal delivery of DPZ.


Journal of Pharmaceutical Sciences | 2017

Development and Validation of an In-Cell Western for Quantifying P-Glycoprotein Expression in Human Brain Microvascular Endothelial (hCMEC/D3) Cells

Mitchell P. McInerney; Yijun Pan; Jennifer L. Short; Joseph A. Nicolazzo

An in-cell western (ICW) protocol detecting the relative expression of P-glycoprotein (P-gp) in human cerebro-microvascular endothelial cells (hCMEC/D3) was developed and optimized, with the intention of improving throughput relative to western blotting (WB). For validation of the ICW protocol, hCMEC/D3 cells were incubated with known P-gp upregulators (10 μM rifampicin and 5 μM SR12813) and treated with siRNA targeted against MDR1, before measuring changes in P-gp expression, using both ICW and WB in parallel. To confirm a relationship between the detected P-gp expression and function, the uptake of the P-gp substrate rhodamine-123 was assessed following SR12813 treatment. Rifampicin and SR12813 significantly upregulated P-gp expression (1.5-fold and 1.9-fold, respectively) compared to control, as assessed by the ICW protocol. WB analysis of the same treatments revealed 1.4-fold and 1.5-fold upregulations. MDR1 siRNA reduced P-gp abundance by 20% and 35% when assessed by ICW and WB, respectively. SR12813 treatment reduced rhodamine-123 uptake by 18%, indicating that the observed changes in P-gp expression by ICW were associated with comparable functional changes. The correlation of P-gp upregulation by WB, rhodamine-123 uptake, and the ICW protocol provide validation of a new ICW method as an alternative method for quantification of P-gp in hCMEC/D3 cells.


Brain Behavior and Immunity | 2018

Cognitive benefits of lithium chloride in APP/PS1 mice are associated with enhanced brain clearance of β-amyloid

Yijun Pan; Jennifer L. Short; Stephanie A. Newman; Kwok Ho Christopher Choy; Durgesh Tiwari; Christopher Yap; Danielle Senyschyn; William A. Banks; Joseph A. Nicolazzo

Epidemiological evidence suggests that people with bipolar disorder prescribed lithium exhibit a lower risk of Alzheimers disease (AD) relative to those prescribed other mood-stabilizing medicines. Lithium chloride (LiCl) reduces brain β-amyloid (Aβ) levels, and the brain clearance of Aβ is reduced in AD. Therefore, the purpose of this study was to assess whether the cognitive benefits of LiCl are associated with enhanced brain clearance of exogenously-administered Aβ. The brain clearance of intracerebroventricularly (icv) administered 125I-Aβ42 was assessed in male Swiss outbred mice administered daily oral NaCl or LiCl (300 mg/kg for 21 days). LiCl exhibited a 31% increase in the brain clearance of 125I-Aβ42 over 10 min, which was associated with a 1.6-fold increase in brain microvascular expression of the blood-brain barrier efflux transporter low density lipoprotein receptor-related protein 1 (LRP1) and increased cerebrospinal fluid (CSF) bulk-flow. 8-month-old female wild type (WT) and APP/PS1 mice were also administered daily NaCl or LiCl for 21 days, which was followed by cognitive assessment by novel object recognition and water maze, and measurement of soluble Aβ42, plaque-associated Aβ42, and brain efflux of 125I-Aβ42. LiCl treatment restored the long-term spatial memory deficit observed in APP/PS1 mice as assessed by the water maze (back to similar levels of escape latency as WT mice), but the short-term memory deficit remained unaffected by LiCl treatment. While LiCl did not affect plaque-associated Aβ42, soluble Aβ42 levels were reduced by 49.9% in APP/PS1 mice receiving LiCl. The brain clearance of 125I-Aβ42 decreased by 27.8% in APP/PS1 mice, relative to WT mice, however, LiCl treatment restored brain 125I-Aβ42 clearance in APP/PS1 mice to a rate similar to that observed in WT mice. These findings suggest that the cognitive benefits and brain Aβ42 lowering effects of LiCl are associated with enhanced brain clearance of Aβ42, possibly via brain microvascular LRP1 upregulation and increased CSF bulk-flow, identifying a novel mechanism of protection by LiCl for the treatment of AD.


Journal of Neurochemistry | 2018

Reduced blood-brain barrier expression of fatty acid-binding protein 5 is associated with increased vulnerability of APP/PS1 mice to cognitive deficits from low omega-3 fatty acid diets

Yijun Pan; Kwok Ho Christopher Choy; Philip J. Marriott; Siew Yeen Chai; Martin J. Scanlon; Christopher J. H. Porter; Jennifer L. Short; Joseph A. Nicolazzo

Lower levels of the cognitively beneficial docosahexaenoic acid (DHA) are often observed in Alzheimers disease (AD) brains. Brain DHA levels are regulated by the blood‐brain barrier (BBB) transport of plasma‐derived DHA, a process facilitated by fatty acid‐binding protein 5 (FABP5). This study reports a 42.1 ± 12.6% decrease in the BBB transport of 14C‐DHA in 8‐month‐old AD transgenic mice (APPswe,PSEN1∆E9) relative to wild‐type mice, associated with a 34.5 ± 6.7% reduction in FABP5 expression in isolated brain capillaries of AD mice. Furthermore, short‐term spatial and recognition memory deficits were observed in AD mice on a 6‐month n‐3 fatty acid‐depleted diet, but not in AD mice on control diet. This intervention led to a dramatic reduction (41.5 ± 11.9%) of brain DHA levels in AD mice. This study demonstrates FABP5 deficiency and impaired DHA transport at the BBB are associated with increased vulnerability to cognitive deficits in mice fed an n‐3 fatty acid‐depleted diet, in line with our previous studies demonstrating a crucial role of FABP5 in BBB transport of DHA and cognitive function.


Advanced Drug Delivery Reviews | 2018

Impact of aging, Alzheimer's disease and Parkinson's disease on the blood-brain barrier transport of therapeutics

Yijun Pan; Joseph A. Nicolazzo

Older people are at a greater risk of medicine-induced toxicity resulting from either increased drug sensitivity or age-related pharmacokinetic changes. The scenario is further complicated with the two most prevalent age-related neurodegenerative diseases, Alzheimers disease (AD) and Parkinsons disease (PD). With aging, AD and PD, there is growing evidence of altered structure and function of the blood-brain barrier (BBB), including modifications to tight junctions and efflux transporters, such as P-glycoprotein. The subsequent impact on CNS drug exposure and risk of neurotoxicity from systemically-acting medicines is less well characterized. The purpose of this review, therefore, is to provide an overview of the multiple changes that occur to the BBB as a result of aging, AD and PD, and the impact that such changes have on CNS exposure of drugs, based on studies conducted in aged rodents or rodent models of disease, and in elderly people with and without AD or PD.


Journal of Diabetes Research and Clinical Metabolism | 2013

Multiple pharmacological interventions targeting cardiovascular disease risk factors in individuals with type 2 diabetes-systematic review

Hanan Khalil; Kevin Peter McNamara; Yijun Pan

Background: The use of pharmacological agents has been shown to slow down the progression of microvascular and macrovascular complications. Most clinical trials address one pharmacological intervention at a time. To date, only a few studies explored multi-factorial pharmacological interventions in T2DM individuals for preventing CVD related complications. Given the current therapeutic inertia in pharmacological management of CVD risk factors, it is important to establish the benefits of a more holistic approach. Therefore, the aim of this review is to assess the efficacy of multiple pharmacological interventions for cardiovascular diseases (CVD) risk factors with or without conventional care in reducing all cause mortality, CVD mortality, stroke and cardiovascular events among adults with type 2 diabetes. Current evidence fails to support the benefit of multiple pharmacological interventions on all cause mortality and death from cardiovascular causes. However, beneficial effects were seen on the reduction of the overall number of cardiovascular events and there were promising trends for secondary outcomes such as stroke, myocardial infarction, revascularisation and amputation.


Molecular Pharmaceutics | 2018

Altered expression of small intestinal drug transporters and hepatic metabolic enzymes in a mouse model of familial Alzheimer’s disease

Yijun Pan; Kotaro Omori; Izna Ali; Masanori Tachikawa; Tetsuya Terasaki; Kim L. R. Brouwer; Joseph A. Nicolazzo

Drug transporter expression and function at the blood-brain barrier is altered in Alzheimers disease (AD). However, the impact of AD on the expression of transporters and metabolizing enzymes in peripheral tissues has received little attention. The current study evaluated the expression of drug transporters and metabolizing enzymes in the small intestine and liver from 8- to 9-month-old female wild-type (WT) and APPswe/PSEN 1dE9 (APP/PS1) transgenic mice, a widely used AD model, using a quantitative targeted absolute proteomics (QTAP) approach. Furthermore, the general morphological appearance of the liver was assessed by immunohistochemistry, and lipid content was visualized using Oil Red O staining. The small intestines of APP/PS1 mice exhibited a significant 2.3-fold increase in multidrug resistance-associated protein 2 (Mrp2), a 1.9-fold decrease in monocarboxylate transporter 1 (Mct1), and a 3.6-fold increase in UDP-glucuronosyltransferase (Ugt) 2b5 relative to those from WT mice based on QTAP analysis. While the liver from APP/PS1 mice exhibited no changes in drug transporter expression, there was a 1.3-fold elevation in cytochrome P450 (Cyp) 51a1 and a 1.2-fold reduction in Cyp2c29 protein expression, and this was associated with morphological alterations including accumulation of hepatocyte lipids. These studies are the first to demonstrate that the protein expression of transporters and metabolizing enzymes important in oral drug absorption are modified in a mouse model of familial AD, which may lead to altered disposition of some orally administered drugs in AD.

Collaboration


Dive into the Yijun Pan'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

Kwok Ho Christopher Choy

Mental Health Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge