Joseph A. Nicolazzo
Monash University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Joseph A. Nicolazzo.
Neuron | 2008
Paul A. Adlard; Robert A. Cherny; David Finkelstein; Elisabeth Gautier; Elysia Robb; Mikhalina Cortes; Irene Volitakis; Xiang Liu; Jeffrey P. Smith; Keyla Perez; Katrina M. Laughton; Qiao-Xin Li; Susan A. Charman; Joseph A. Nicolazzo; Simon Wilkins; Karolina Deleva; Toni Lynch; Gaik Beng Kok; Craig W. Ritchie; Rudolph E. Tanzi; Roberto Cappai; Colin L. Masters; Kevin J. Barnham; Ashley I. Bush
As a disease-modifying approach for Alzheimers disease (AD), clioquinol (CQ) targets beta-amyloid (Abeta) reactions with synaptic Zn and Cu yet promotes metal uptake. Here we characterize the second-generation 8-hydroxy quinoline analog PBT2, which also targets metal-induced aggregation of Abeta, but is more effective as a Zn/Cu ionophore and has greater blood-brain barrier permeability. Given orally to two types of amyloid-bearing transgenic mouse models of AD, PBT2 outperformed CQ by markedly decreasing soluble interstitial brain Abeta within hours and improving cognitive performance to exceed that of normal littermate controls within days. Nontransgenic mice were unaffected by PBT2. The current data demonstrate that ionophore activity, inhibition of in vitro metal-mediated Abeta reactions, and blood-brain barrier permeability are indices that predict a potential disease-modifying drug for AD. The speed of recovery of the animals underscores the acutely reversible nature of the cognitive deficits associated with transgenic models of AD.
European Journal of Pharmaceutical Sciences | 2010
Admire Dube; Joseph A. Nicolazzo; Ian Larson
Catechins found in green tea have received considerable attention due to their favourable biological properties which include cardioprotective, neuroprotective and anti-cancer effects. However, their therapeutic potential is limited by their low oral bioavailability, attributed to poor stability and intestinal absorption. We encapsulated (+)-catechin (C) and (-)-epigallocatechin gallate (EGCg) in chitosan nanoparticles (CS NP) as a means of enhancing their intestinal absorption. Using excised mouse jejunum in Ussing chambers, encapsulation significantly enhanced (p<0.05) intestinal absorption. The cumulative amounts transported after encapsulation were significantly higher (p<0.05), i.e. 302.1+/-46.1 vs 206.8+/-12.6ng/cm(2) and 102.7+/-12.4 vs 57.9+/-7.9ng/cm(2) for C and EGCg, respectively. The mechanism by which absorption was enhanced was not through an effect of CS NPs on intestinal paracellular or passive transcellular transport processes (as shown by transport of (14)C-mannitol and (3)H-propranolol) or an effect on efflux proteins (as shown by transport of (3)H-digoxin) but was likely due to stabilization of catechins after encapsulation (99.7+/-0.7 vs 94.9+/-3.8% and 56.9+/-3.0 vs 1.3+/-1.7% of the initial C and EGCg concentration remaining, respectively). This study demonstrates that encapsulation of catechins in CS NPs enhances their intestinal absorption and is a promising strategy for improving their bioavailability.
Journal of Pharmacy and Pharmacology | 2006
Joseph A. Nicolazzo; Susan A. Charman; William N. Charman
Much research has focussed on the development of novel therapeutic agents to target various central nervous system disorders, however less attention has been given to determining the potential of such agents to permeate the blood‐brain barrier (BBB), a factor that will ultimately govern the effectiveness of these agents in man. In order to assess the potential for novel compounds to permeate the BBB, various in‐vitro, in‐vivo and in‐silico methods may be employed. Although in‐vitro models (such as primary cell culture and immortalized cell lines) are useful as a screening method and can appropriately rank compounds in order of BBB permeability, they often correlate poorly to in‐vivo brain uptake due to down‐regulation of some BBB‐specific transporters. In‐vivo models (such as the internal carotid artery single injection or perfusion, intravenous bolus injection, brain efflux index and intracerebral microdialysis) provide more accurate information regarding brain uptake, and these can be complemented with novel imaging techniques (such as magnetic resonance imaging and positron emission tomography), although such methods are not suited to high‐throughput permeability assessment. This paper reviews current methods used for assessing BBB permeability and highlights the particular advantages and disadvantages associated with each method, with a particular focus on methods suitable for moderate‐ to high‐throughput screening.
PLOS ONE | 2013
Wei Wu; Joseph A. Nicolazzo; Li Wen; Roger S. Chung; Roger Stankovic; Shisan Bao; Chai K. Lim; Bruce J. Brew; Karen M. Cullen; Gilles J. Guillemin
To assess the role of the kynurenine pathway in the pathology of Alzheimers disease (AD), the expression and localization of key components of the kynurenine pathway including the key regulatory enzyme tryptophan 2,3 dioxygenase (TDO), and the metabolites tryptophan, kynurenine, kynurenic acid, quinolinic acid and picolinic acid were assessed in different brain regions of triple transgenic AD mice. The expression and cell distribution of TDO and quinolinic acid, and their co-localization with neurofibrillary tangles and senile β amyloid deposition were also determined in hippocampal sections from human AD brains. The expression of TDO mRNA was significantly increased in the cerebellum of AD mouse brain. Immunohistochemistry demonstrated that the density of TDO immuno-positive cells was significantly higher in the AD mice. The production of the excitotoxin quinolinic acid strongly increased in the hippocampus in a progressive and age-dependent manner in AD mice. Significantly higher TDO and indoleamine 2,3 dioxygenase 1 immunoreactivity was observed in the hippocampus of AD patients. Furthermore, TDO co-localizes with quinolinic acid, neurofibrillary tangles-tau and amyloid deposits in the hippocampus of AD. These results show that the kynurenine pathway is over-activated in AD mice. This is the first report demonstrating that TDO is highly expressed in the brains of AD mice and in AD patients, suggesting that TDO-mediated activation of the kynurenine pathway could be involved in neurofibrillary tangles formation and associated with senile plaque. Our study adds to the evidence that the kynurenine pathway may play important roles in the neurodegenerative processes of AD.
European Journal of Pharmaceutical Sciences | 2011
Admire Dube; Joseph A. Nicolazzo; Ian Larson
The green tea catechin (-)-epigallocatechin gallate (EGCG) has attracted significant research interest due to its beneficial therapeutic effects, which include anti-oxidant, neuro-protective and anti-cancer effects. However, the therapeutic potential of EGCG following oral consumption is limited by its poor absorption. To address this issue, EGCG has been encapsulated in chitosan-tripolyphosphate nanoparticles (CS NPs) and the oral absorption of EGCG evaluated in Swiss Outbred mice. Administration of the CS NPs enhanced the plasma exposure of total EGCG by a factor of 1.5 relative to an EGCG solution, with plasma AUC((0-5 h)) values of 116.4±4.1 and 179.3±10.8 nM.h (mean±s.d., n=3-5) for the EGCG solution and CS NPs, respectively. Associated with the increased plasma exposure of EGCG was an enhancement in concentrations of EGCG in the stomach and jejunum of mice following CS NP administration. A 2.3-fold increase in the apparent exposure of EGCG to the jejunum (AUC(j)) was observed following CS NP encapsulation, with AUC(j(0-5 h)) values of 5.3±1.1 and 12.3±1.5 μM.h (mean±s.d., n=3-5) for the EGCG solution and CS NPs, respectively. The enhanced exposure of EGCG to the jejunum was likely responsible for the increased plasma concentrations of EGCG. The findings from this study suggest that CS NPs may be a useful approach for enhancing oral delivery, and therapeutic application, of EGCG in a number of disease conditions.
Current Topics in Medicinal Chemistry | 2009
Joseph A. Nicolazzo; Kasiram Katneni
With the discovery of novel therapeutic targets within the central nervous system (CNS), there has been a significant effort to synthesize a multitude of drug molecules with increasing potency and selectivity. However, the impact of the blood-brain barrier (BBB) in limiting effective concentrations of drug candidate from reaching the brain parenchyma is often ignored, resulting in a lack of efficacy when administered to animal models or humans. Intercellular drug transport across the BBB is negligible due to the impermeable tight junctions formed by interconnecting endothelial cells. Furthermore, drug permeability via the transcellular route cannot be assumed for all molecules due to the high expression of drug efflux transport proteins, which effectively extrude compounds from the brain endothelial cell back into the cerebral vasculature. In addition to the extensively-studied P-glycoprotein (P-gp, ABCB1), the brain endothelial cells also express multidrug resistance associated proteins (MRP, ABCC) and breast cancer resistance protein (BCRP, ABCG2), amongst other efflux transporters. While more research has focussed on the impact of P-gp and MRP on drug transport across the BBB, the role of ABCG2 in limiting exposure of drug molecules to the CNS is now becoming more clearly understood. The purpose of this review, therefore, is to summarise the findings of the various studies assessing the expression profile of ABCG2 at the BBB, to provide an overview on the current research being undertaken to identify specific ABCG2 inhibitors with therapeutic benefit, and to critically assess the functional role of ABCG2 on drug transport across the BBB.
Antimicrobial Agents and Chemotherapy | 2011
Liang Jin; Jian Li; Roger L. Nation; Joseph A. Nicolazzo
ABSTRACT The aim of this study was to investigate the factors limiting the blood-brain barrier (BBB) transport of colistin in healthy mice and to assess the impact of systemic inflammation on the transport of this antibiotic across the BBB. Colistin sulfate (40 mg/kg) was administered subcutaneously to Swiss outbred mice as single and multiple doses to determine any relationship between brain uptake and plasma concentrations of colistin. To assess the effect of P-glycoprotein (P-gp) on BBB transport, colistin sulfate (5 mg/kg) was concomitantly administered intravenously with PSC833 or GF120918 (10 mg/kg). Systemic inflammation was induced by three intraperitoneal injections of lipopolysaccharide (LPS; 3 mg/kg), and BBB transport of colistin was subsequently measured following subcutaneous administration and by an in situ brain perfusion. The brain uptake of colistin was low following single and multiple subcutaneous doses, with brain-to-plasma concentration ratios ranging between 0.021 and 0.037, and this was not significantly enhanced by coadministration of GF120918 or PSC833 (P > 0.05). LPS significantly increased the brain uptake of subcutaneously administered colistin with area under the brain concentration time curve (AUCbrain) values of 11.7 ± 2.7 μg·h/g and 4.0 ± 0.3 μg·h/g for LPS- and saline-treated mice, respectively (mean ± standard deviation). Similarly, in situ perfusion of colistin led to higher antibiotic brain concentrations in LPS-treated animals than in saline-treated animals, with colistin brain-to-perfusate concentration ratios of 0.019 ± 0.001 and 0.014 ± 0.001, respectively. This study demonstrates that the BBB transport of colistin is negligible in healthy mice; however, brain concentrations of colistin can be significantly enhanced during systemic inflammation, as might be observed in infected patients.
Molecular Pharmaceutics | 2015
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.
Antimicrobial Agents and Chemotherapy | 2009
Liang Jin; Jian Li; Roger L. Nation; Joseph A. Nicolazzo
ABSTRACT A sensitive and reliable liquid chromatographic method was developed and validated for the determination of colistin concentrations in mouse brain homogenate. With a mobile phase consisting of acetonitrile-tetrahydrofuran-water (50:25:25 [vol/vol]) at a flow rate of 1 ml/min, a linear correlation between peak area and colistin concentration was observed over the concentration range of 93.8 to 3,000 ng/g in brain tissue (R2 > 0.994). Intra- and interday coefficients of variation were 5.1 to 8.3% and 5.8 to 8.5%, respectively, and the recovery ranged from 85% to 94%. This assay was then utilized to determine the amount of colistin that permeated the blood-brain barrier over a 2-h period following bolus intravenous administration of colistin sulfate to mice. After a single dose of 5 mg/kg of body weight to mice, brain homogenate concentrations of colistin were very low, relative to plasma colistin concentrations, suggesting that colistin permeability across the healthy blood-brain barrier is negligible during this experimental period.
Journal of Medicinal Chemistry | 2014
Simon J. Mountford; Anthony L. Albiston; William N. Charman; Leelee Ng; Jessica K. Holien; Michael W. Parker; Joseph A. Nicolazzo; Philip E. Thompson; Siew Yeen Chai
Peptide inhibitors of insulin-regulated aminopeptidase (IRAP) enhance fear avoidance and spatial memory and accelerate spatial learning in a number of memory paradigms. Using a virtual screening approach, a series of benzopyran compounds was identified that inhibited the catalytic activity of IRAP, ultimately resulting in the identification of potent and specific inhibitors. The present study describes the medicinal chemistry campaign that led to the development of the lead candidate, 3, highlighting the key structural features considered as critical for binding. Furthermore, the in vivo pharmacokinetics and brain uptake of compounds (1 and 3) were assessed in rats and were complemented with in vitro human and rat microsomal stability studies. Following intravenous administration to rodents, 3 exhibits brain exposure, albeit it is rapidly converted to 1, a compound which also exhibits potent inhibition of IRAP.