Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
1.
Handb Exp Pharmacol ; 273: 97-120, 2022.
Article in English | MEDLINE | ID: mdl-33474672

ABSTRACT

Neuropharmacokinetics considers cerebral drug distribution as a critical process for central nervous system drug action as well as for drug penetration through the CNS barriers. Brain distribution of small molecules obeys classical rules of drug partition, permeability, binding to fluid proteins or tissue components, and tissue perfusion. The biodistribution of all drugs, including both small molecules and biologics, may also be influenced by specific brain properties related to brain anatomy and physiological barriers, fluid dynamics, and cellular and biochemical composition, each of which can exhibit significant interspecies differences. All of these properties contribute to select optimal dosing paradigms and routes of drug delivery to reach brain targets for classical small molecule drugs as well as for biologics. The importance of these properties for brain delivery and exposure also highlights the need for efficient new analytical technologies to more comprehensively investigate drug distribution in the CNS, a complex multi-compartmentalized organ system.


Subject(s)
Biological Products , Brain , Biological Products/pharmacokinetics , Blood-Brain Barrier/metabolism , Brain/metabolism , Drug Delivery Systems , Humans , Pharmaceutical Preparations/metabolism , Species Specificity , Tissue Distribution
2.
J Epidemiol Glob Health ; 11(4): 321-325, 2021 12.
Article in English | MEDLINE | ID: mdl-34734383

ABSTRACT

Various key performance indicators (KPIs) are communicated daily to the public by health authorities since the COVID-19 pandemic has started. "Upstream" KPIs mainly include the incidence of detected Sars-CoV-2-positive cases in the population, and "downstream" KPIs include daily hospitalizations, intensive care unit admissions and fatalities. Whereas "downstream" KPIs are essential to evaluate and adapt hospital organization, "upstream" KPIs are the most appropriate to decide on the strength of restrictions such as lockdown set up and evaluate their effectiveness. Here, we suggested tools derived from pharmacokinetic calculations to improve understanding the epidemic progression. From the time course of the number of new cases of SARS-coV-2 infection in the population, it is possible to calculate the infection rate constant using a simple linear regression and determine its corresponding half-life. This epidemic regression half-life is helpful to measure the potential benefits of restriction measures and to estimate the adequate duration of lockdown if implemented by policymakers in relation to the decided public health objectives. In France, during the first lockdown, we reported an epidemic half-life of 10 days. Our tools allow clearly acknowledging that the zero-COVID target is difficult to reach after a period of lockdown as seven half-lives are required to clear 99.2% of the epidemic and more than 10 half-lives to almost reach the objective of eliminating 100% of the contaminations.


Subject(s)
COVID-19 , SARS-CoV-2 , Communicable Disease Control , Hospitalization , Humans , Pandemics
3.
J Gen Intern Med ; 36(9): 2731-2738, 2021 09.
Article in English | MEDLINE | ID: mdl-34131877

ABSTRACT

BACKGROUND: Since late summer 2020, the French authorities implemented a curfew/lightened lockdown-alternating strategy instead of strict lockdown, to improve acceptability and limit socioeconomic consequences. However, data on curfew-related efficacy to control the epidemic are scarce. OBJECTIVE: To investigate the effects on COVID-19 spread in France of curfew combined to local and/or nationwide restrictions from late summer 2020 to mid-February 2021. DESIGN: We conducted a comparative evaluation using a susceptible-infected-recovered (SIR)-based model completed with epidemiokinetic tools. MAIN MEASURES: We analyzed the time-course of epidemic progression rate under curfew in French Guyana and five metropolitan regions where additional restrictions were implemented at different times. Using linear regressions of the decay/increase rates in daily contaminations, we calculated the epidemic regression half-lives (t1/2ß) for each identified period. KEY RESULTS: In French Guyana, two decay periods with rapid regression (t1/2ß of ~10 days) were observed under curfew, with slowing (t1/2ß of ~43 days) when curfew was lightened. During the 2-week pre-lockdown curfew (2020/10/17-2020/11/02) in Provence-Alpes-Côte-d'Azur, Auvergne-Rhône-Alpes, and Ile-de-France, the epidemic progression was unchanged. During the post-lockdown curfew (2020/12/15-2020/02/14), the epidemic slowly regressed in Grand-Est (t1/2ß of ~37 days), whereas its progression rate plateaued in Auvergne-Rhône-Alpes and increased immediately in Provence-Alpes-Côte-d'Azur, Ile-de-France, and Nouvelle-Aquitaine, whatever the curfew starting time was (06:00 or 08:00 pm). Interestingly, a delayed slow decay (17 days < t1/2ß < 64 days) occurred under curfew in all regions except Ile-de-France. CONCLUSIONS: Curfew allowed the temporary control of SARS-CoV-2 epidemic, however variably in the French regions, without preventing lockdown necessity. To accelerate the epidemic regression such as observed in French Guyana, curfew should be implemented timely with additional restrictions.


Subject(s)
COVID-19 , Communicable Disease Control/methods , COVID-19/prevention & control , France/epidemiology , Humans
4.
Neuropharmacology ; 191: 108588, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33940010

ABSTRACT

The inducible p25 overexpression mouse model recapitulate many hallmark features of Alzheimer's disase including progressive neuronal loss, elevated Aß, tau pathology, cognitive dysfunction, and impaired synaptic plasticity. We chose p25 mice to evaluate the physical and functional integrity of the blood-brain barrier (BBB) in a context of Tau pathology (pTau) and severe neurodegeneration, at an early (3 weeks ON) and a late (6 weeks ON) stage of the pathology. Using in situ brain perfusion and confocal imaging, we found that the brain vascular surface area and the physical integrity of the BBB were unaltered in p25 mice. However, there was a significant 14% decrease in cerebrovascular volume in 6 weeks ON mice, possibly explained by a significant 27% increase of collagen IV in the basement membrane of brain capillaries. The function of the BBB transporters GLUT1 and LAT1 was evaluated by measuring brain uptake of d-glucose and phenylalanine, respectively. In 6 weeks ON p25 mice, d-glucose brain uptake was significantly reduced by about 17% compared with WT, without any change in the levels of GLUT1 protein or mRNA in brain capillaries. The brain uptake of phenylalanine was not significantly reduced in p25 mice compared with WT. Lack of BBB integrity, impaired BBB d-glucose transport have been observed in several mouse models of AD. In contrast, reduced cerebrovascular volume and an increased basement membrane thickness may be more specifically associated with pTau in mouse models of neurodegeneration.


Subject(s)
Alzheimer Disease/chemically induced , Blood-Brain Barrier/physiopathology , Cerebrovascular Circulation/physiology , Disease Models, Animal , Animals , Atrophy , Biological Transport , Blood Vessels/pathology , Blood-Brain Barrier/physiology , Brain/metabolism , Brain/pathology , Glucose/metabolism , Green Fluorescent Proteins , Mice , Mice, Transgenic , tau Proteins/metabolism
5.
J Gen Intern Med ; 36(3): 746-752, 2021 03.
Article in English | MEDLINE | ID: mdl-33442818

ABSTRACT

BACKGROUND: To date, the risk/benefit balance of lockdown in controlling severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) epidemic is controversial. OBJECTIVE: We aimed to investigate the effectiveness of lockdown on SARS-CoV-2 epidemic progression in nine different countries (New Zealand, France, Spain, Germany, the Netherlands, Italy, the UK, Sweden, and the USA). DESIGN: We conducted a cross-country comparative evaluation using a susceptible-infected-recovered (SIR)-based model completed with pharmacokinetic approaches. MAIN MEASURES: The rate of new daily SARS-CoV-2 cases in the nine countries was calculated from the World Health Organization's published data. Using a SIR-based model, we determined the infection (ß) and recovery (γ) rate constants; their corresponding half-lives (t1/2ß and t1/2γ); the basic reproduction numbers (R0 as ß/γ); the rates of susceptible S(t), infected I(t), and recovered R(t) compartments; and the effectiveness of lockdown. Since this approach requires the epidemic termination to build the (I) compartment, we determined S(t) at an early epidemic stage using simple linear regressions. KEY RESULTS: In New Zealand, France, Spain, Germany, the Netherlands, Italy, and the UK, early-onset stay-at-home orders and restrictions followed by gradual deconfinement allowed rapid reduction in SARS-CoV-2-infected individuals (t1/2ß ≤ 14 days) with R0 ≤ 1.5 and rapid recovery (t1/2γ ≤ 18 days). By contrast, in Sweden (no lockdown) and the USA (heterogeneous state-dependent lockdown followed by abrupt deconfinement scenarios), a prolonged plateau of SARS-CoV-2-infected individuals (terminal t1/2ß of 23 and 40 days, respectively) with elevated R0 (4.9 and 4.4, respectively) and non-ending recovery (terminal t1/2γ of 112 and 179 days, respectively) was observed. CONCLUSIONS: Early-onset lockdown with gradual deconfinement allowed shortening the SARS-CoV-2 epidemic and reducing contaminations. Lockdown should be considered as an effective public health intervention to halt epidemic progression.


Subject(s)
COVID-19/epidemiology , COVID-19/prevention & control , Communicable Disease Control/organization & administration , Quarantine/statistics & numerical data , France/epidemiology , Global Health , Humans , Italy/epidemiology , Netherlands/epidemiology , New Zealand/epidemiology , Outcome Assessment, Health Care , Social Isolation , Spain/epidemiology , Sweden/epidemiology , United States/epidemiology
6.
Eur J Pharm Sci ; 117: 68-79, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29427702

ABSTRACT

Efficacy of drugs aimed at treating central nervous system (CNS) disorders rely partly on their ability to cross the cerebral endothelium, also called the blood-brain barrier (BBB), which constitutes the main interface modulating exchanges of compounds between the brain and blood. In this work, we used both, conventional pharmacokinetics (PK) approach and in situ brain perfusion technique to study the blood and brain PK of PKRinh, an inhibitor of the double-stranded RNA-dependent protein kinase (PKR) activation, in mice. PKRinh showed a supra dose-proportional blood exposure that was not observed in the brain, and a brain to blood AUC ratio of unbound drug smaller than 1 at all tested doses. These data suggested the implication of an active efflux at the BBB. Using in situ brain perfusion technique, we showed that PKRinh has a very high brain uptake clearance which saturates with increasing concentrations. Fitting the data to a Michaelis-Menten equation revealed that PKRinh transport through the BBB is composed of a passive unsaturable flux and an active saturable protein-mediated efflux with a km of ≅ 3 µM. We were able to show that the ATP-binding cassette (ABC) transporter P-gp (Abcb1), but not Bcrp (Abcg2), was involved in the brain to blood efflux of PKRinh. At the circulating PKRinh concentrations of this study, the P-gp was not saturated, in accordance with the linear brain PKRinh PK. Finally, PKRinh had high brain uptake clearance (14 µl/g/s) despite it is a good P-gp substrate (P-gp Efflux ratio ≅ 3.6), and reached similar values than the cerebral blood flow reference, diazepam, in P-gp saturation conditions. With its very unique brain transport properties, PKRinh improves our knowledge about P-gp-mediated efflux across the BBB for the development of new CNS directed drugs.


Subject(s)
Brain/metabolism , Central Nervous System Agents/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , Animals , Biological Transport , Central Nervous System Agents/blood , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase Inhibitors/blood
7.
Sci Rep ; 7(1): 13393, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29042617

ABSTRACT

ABCG4 is an ATP-binding cassette transmembrane protein which has been shown, in vitro, to participate in the cellular efflux of desmosterol and amyloid-ß peptide (Aß). ABCG4 is highly expressed in the brain, but its localization and function at the blood-brain barrier (BBB) level remain unknown. We demonstrate by qRT-PCR and confocal imaging that mouse Abcg4 is expressed in the brain capillary endothelial cells. Modelling studies of the Abcg4 dimer suggested that desmosterol showed thermodynamically favorable binding at the putative sterol-binding site, and this was greater than for cholesterol. Additionally, unbiased docking also showed Aß binding at this site. Using a novel Abcg4-deficient mouse model, we show that Abcg4 was able to export Aß and desmosterol at the BBB level and these processes could be inhibited by probucol and L-thyroxine. Our assay also showed that desmosterol antagonized the export of Aß, presumably as both bind at the sterol-binding site on Abcg4. We show for the first time that Abcg4 may function in vivo to export Aß at the BBB, in a process that can be antagonized by its putative natural ligand, desmosterol (and possibly cholesterol).


Subject(s)
ATP Binding Cassette Transporter, Subfamily G/genetics , ATP Binding Cassette Transporter, Subfamily G/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Gene Expression , ATP Binding Cassette Transporter, Subfamily G/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Amyloid beta-Peptides/chemistry , Animals , Biomarkers , Capillary Permeability , Cell Membrane Permeability , Desmosterol/metabolism , Fluorescent Antibody Technique , Gene Targeting , Genetic Loci , Mice , Mice, Knockout , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , Structure-Activity Relationship
8.
Br J Pharmacol ; 173(3): 497-510, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26507673

ABSTRACT

BACKGROUND AND PURPOSE: The respective impact and interplay between ABC (P-glycoprotein/P-gp/Abcb1a, BCRP/ABCG2, MRP/ABCC) and SLC transporter functions at the blood-brain barrier (BBB) and blood-retinal barriers (BRB) are incompletely understood. EXPERIMENTAL APPROACH: We measured the initial cerebral and retinal distribution of selected ABC substrates by in situ carotid perfusion using P-gp/Bcrp knockout mice and chemical ABC/SLC modulation strategies. P-gp, Bcrp, Mrp1 and Mrp4 were studied by confocal retina imaging. KEY RESULTS: Chemical or physical disruption of P-gp increased [(3) H]-verapamil transport by ~10-fold at the BBB and ~1.5-fold at the BRB. [(3) H]-Verapamil transport involved influx-mediated by an organic cation clonidine-sensitive/diphenhydramine-sensitive proton antiporter at both barriers; this effect was unmasked when P-gp was partially or fully inhibited/disrupted at the BBB. Studies of [(3) H]-mitoxantrone and [(3) H]-zidovudine transport suggested, respectively, that Bcrp efflux was less involved at the BRB than BBB, whereas Mrps were significantly and similarly involved at both barriers. Confocal imaging showed that P-gp and Bcrp were expressed in intra-retinal vessels (inner BRB/iBRB) but absent from the blood/basal membrane of cells of the retinal pigment epithelium (outer BRB/oBRB/RPE) where, in contrast, Mrp1 and Mrp4 were localized. CONCLUSIONS AND IMPLICATIONS: P-gp, Bcrp, Mrp1 and Mrp4 are differentially expressed at the outer and inner BRB, resulting in an altered ability to limit substrate distribution at the retina as compared with the BBB. [(3) H]-Verapamil distribution is not P-gp-specific and involves a proton antiporter at both the BBB and BRB. However, this transport is concealed by P-gp at the BBB, but not at the BRB, where P-gp activity is reduced.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Blood-Brain Barrier/metabolism , Blood-Retinal Barrier/metabolism , Verapamil/pharmacology , ATP-Binding Cassette Transporters/genetics , Animals , Biological Transport , Male , Mice, Knockout , Mitoxantrone/pharmacology , Zidovudine/pharmacology
9.
J Alzheimers Dis ; 49(2): 287-300, 2016.
Article in English | MEDLINE | ID: mdl-26484906

ABSTRACT

The involvement of transporters located at the blood-brain barrier (BBB) has been suggested in the control of cerebral Aß levels, and thereby in Alzheimer's disease (AD). However, little is known about the regulation of these transporters at the BBB in animal models of AD. In this study, we investigated the BBB expression of Aß influx (Rage) and efflux (Abcb1-Abcg2-Abcg4-Lrp-1) transporters and cholesterol transporter (Abca1) in 3-18-month-old 3xTg-AD and control mice. The age-dependent effect of BBB transporters regulation on the brain uptake clearance (Clup) of [3H]cholesterol and [3H]Aß1 - 40 was then evaluated in these mice, using the in situ brain perfusion technique. Our data suggest that transgenes expression led to the BBB increase in Aß influx receptor (Rage) and decrease in efflux receptor (Lrp-1). Our data also indicate that mice have mechanisms counteracting this increased net influx. Indeed, Abcg4 and Abca1 are up regulated in 3- and 3/6-month-old 3xTg-AD mice, respectively. Our data show that the balance between the BBB influx and efflux of Aß is maintained in 3 and 6-month-old 3xTg-AD mice, suggesting that Abcg4 and Abca1 control the efflux of Aß through the BBB by a direct (Abcg4) or indirect (Abca1) mechanism. At 18 months, the BBB Aß efflux is significantly increased in 3xTg-AD mice compared to controls. This could result from the significant up-regulation of both Abcg2 and Abcb1 in 3xTg-AD mice compared to control mice. Thus, age-dependent regulation of several Aß and cholesterol transporters at the BBB could ultimately limit the brain accumulation of Aß.


Subject(s)
Aging , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/physiopathology , Brain/metabolism , Peptide Fragments/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Alzheimer Disease/genetics , Animals , Biological Transport/genetics , Blood-Brain Barrier/metabolism , Carbon Isotopes/metabolism , Cholesterol/metabolism , Disease Models, Animal , Humans , Lipoproteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mice, Transgenic , Receptors, LDL/metabolism , Sucrose/metabolism , Tritium/metabolism , Tumor Suppressor Proteins/metabolism
10.
Neuropharmacology ; 81: 311-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24631967

ABSTRACT

We evaluated the integrity and function of the blood-brain barrier in 3xTg-AD mice aged 3-18 months and in APP/PS1 mice aged 8-months to determine the impacts of changes in amyloid and tau proteins on the brain vascular changes. The vascular volume (Vvasc) was sub-normal in 3xTg-AD mice aged from 6 to 18 months, but not in the APP/PS1 mice. The uptakes of [(3)H]-diazepam by the brains of 3xTg-AD, APP/PS1 and their age-matched control mice were similar at all the times studied, suggesting that the simple diffusion of small solutes is unchanged in transgenic animals. The uptake of d-glucose by the brains of 18-month old 3xTg-AD mice, but not by those of 8-month old APP/PS1 mice, was reduced compared to their age-matched controls. Accordingly, the amount of Glut-1 protein was 1.4 times lower in the brain capillaries of 18 month-old 3xTg-AD mice than in those of age-matched control mice. We conclude that the brain vascular volume is reduced early in 3xTg-AD mice, 6 months before the appearance of pathological lesions, and that this reduction persists until they are at least 18 months old. The absence of alterations in the BBB of APP/PS1 mice suggests that hyperphosphorylated tau proteins contribute to the vascular changes that occur in AD.


Subject(s)
Alzheimer Disease/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiology , Brain/pathology , Cerebrovascular Circulation/genetics , Glucose Transporter Type 1/metabolism , Age Factors , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Brain/drug effects , Brain/metabolism , Diazepam/metabolism , Disease Models, Animal , Functional Laterality , Glucose/metabolism , Humans , Mice , Mice, Transgenic , Microvessels/pathology , Microvessels/physiopathology , Mutation/genetics , Presenilin-1/genetics , Sucrose/metabolism , tau Proteins/genetics
11.
Mol Pharm ; 11(1): 243-53, 2014 Jan 06.
Article in English | MEDLINE | ID: mdl-24215184

ABSTRACT

Monoclonal antibodies (mAbs) targeting blood-brain barrier (BBB) transporters are being developed for brain drug targeting. However, brain uptake quantification remains a challenge, particularly for large compounds, and often requires the use of radioactivity. In this work, we adapted an in situ brain perfusion technique for a fluorescent mAb raised against the mouse transferrin receptor (TfR) (clone Ri7). We first confirmed in vitro that the internalization of fluorolabeled Ri7 mAbs is saturable and dependent on the TfR in N2A and bEnd5 cells. We next showed that the brain uptake coefficient (Clup) of 100 µg (∼220 nM) of Ri7 mAbs fluorolabeled with Alexa Fluor 750 (AF750) was 0.27 ± 0.05 µL g(-1) s(-1) after subtraction of values obtained with a control IgG. A linear relationship was observed between the distribution volume VD (µL g(-1)) and the perfusion time (s) over 30-120 s (r(2) = 0.997), confirming the metabolic stability of the AF750-Ri7 mAbs during perfusion. Co-perfusion of increasing quantities of unlabeled Ri7 decreased the AF750-Ri7 Clup down to control IgG levels over 500 nM, consistent with a saturable mechanism. Fluorescence microscopy analysis showed a vascular distribution of perfused AF750-Ri7 in the brain and colocalization with a marker of basal lamina. To our knowledge, this is the first reported use of the in situ brain perfusion technique combined with quantification of compounds labeled with near-infrared fluorophores. Furthermore, this study confirms the accumulation of the antitransferrin receptor Ri7 mAb in the brain of mice through a saturable uptake mechanism.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Brain/metabolism , Drug Delivery Systems , Genetic Vectors/administration & dosage , Neuroblastoma/drug therapy , Receptors, Transferrin/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Blood-Brain Barrier/drug effects , Blotting, Western , Brain/drug effects , Fluorescent Antibody Technique , Genetic Vectors/immunology , Genetic Vectors/pharmacokinetics , Male , Mice , Mice, Inbred BALB C , Neuroblastoma/immunology , Neuroblastoma/metabolism , Perfusion , Tissue Distribution , Tumor Cells, Cultured
12.
J Alzheimers Dis ; 36(3): 555-61, 2013.
Article in English | MEDLINE | ID: mdl-23635403

ABSTRACT

The influx of amyloid-ß peptide (Aß) across the blood-brain barrier is partly mediated by the receptor for advanced glycation end products (RAGE). But other transporters, like Oatp (organic anion transporter polypeptide, SLC21) transporters, could also be involved. We used in situ brain perfusion to show that rosuvastatin and taurocholate, two established Oatp1a4 substrates, decreased (5-fold) the Clup of [3H]Aß while L-thyroxine increased it (5.5-fold). We demonstrated an interaction between Aß and Oatp1a4 by co-immunoprecipitation and western blotting experiments, supporting the hypothesis that the rosuvastatin- and taurocholate-sensitive transporter was Oatp1a4. In conclusion, our results suggest that, in mice, the brain uptake of Aß is partly mediated by Oatp1a4 and that L-thyroxine may play a crucial role in the inhibition of brain Aß clearance.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Organic Cation Transport Proteins/metabolism , Protein Transport/physiology , Animals , Blood-Brain Barrier/drug effects , Fluorobenzenes/pharmacology , Mice , Protein Transport/drug effects , Pyrimidines/pharmacology , Rosuvastatin Calcium , Sulfonamides/pharmacology , Taurocholic Acid/pharmacology , Thyroxine/pharmacology
13.
BMC Neurosci ; 13: 84, 2012 Jul 23.
Article in English | MEDLINE | ID: mdl-22824057

ABSTRACT

BACKGROUND: Amyloid precursor protein (APP), a key molecule in Alzheimer's disease (AD), is metabolized in two alternative cleavages, generating either the amyloidogenic peptides involved in AD pathology or the soluble form of APP (sAPPα). The level of amyloidogenic peptides in human cerebrospinal fluid (CSF) is considered to be a biomarker of AD, whereas the level of sAPPα in CSF as a biomarker has not been clearly established. sAPPα has neurotrophic and neuroprotective properties. Stimulating its formation and secretion is a promising therapeutic target in AD research. To this end, very sensitive tests for preclinical and clinical research are required. METHODS: The tests are based on homogenous time-resolved fluorescence and require no washing steps. RESULTS: We describe two new rapid and sensitive tests for quantifying mouse and human sAPPα. These 20 µl-volume tests quantify the levels of: i) endogenous mouse sAPPα in the conditioned medium of mouse neuron primary cultures, as well as in the CSF of wild-type mice, ii) human sAPPα in the CSF of AD mouse models, and iii) human sAPPα in the CSF of AD and non-AD patients. These tests require only 5 µl of conditioned medium from 5 × 10(4) mouse primary neurons, 1 µl of CSF from wild-type and transgenic mice, and 0.5 µl of human CSF. CONCLUSIONS: The high sensitivity of the mouse sAPPα test will allow high-throughput investigations of molecules capable of increasing the secretion of endogenous sAPPα in primary neurons, as well as the in vivo validation of molecules of interest through the quantification of sAPPα in the CSF of treated wild-type mice. Active molecules could then be tested in the AD mouse models by quantifying human sAPPα in the CSF through the progression of the disease. Finally, the human sAPPα test could strengthen the biological diagnosis of AD in large clinical investigations. Taken together, these new tests have a wide field of applications in preclinical and clinical studies.


Subject(s)
Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Disease Models, Animal , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/cerebrospinal fluid , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/pathology , Cognition Disorders/etiology , Dose-Response Relationship, Drug , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Humans , Linear Models , Mice , Mice, Transgenic , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Presenilin-1/genetics , Spinal Puncture , Time Factors , tau Proteins/genetics , tau Proteins/metabolism
14.
Biomaterials ; 33(22): 5593-602, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22575831

ABSTRACT

PLGA-PEG nanocapsules containing a liquid core of perfluorooctyl bromide were synthesized by an emulsion-evaporation process and designed as contrast agents for (19)F MRI. Physico-chemical properties of plain and PEGylated nanocapsules were compared. The encapsulation efficiency of PFOB, estimated by (19)F NMR spectroscopy, is enhanced when using PLGA-PEG instead of PLGA. PLGA-PEG nanocapsule diameter, measured by Dynamic Light Scattering is around 120 nm, in agreement with Transmission Electron microscopy (TEM) observations. TEM and Scanning Electron Microscopy (SEM) reveal that spherical core-shell morphology is preserved. PEGylation is further confirmed by Zeta potential measurements and X-ray Photoelectron Spectroscopy. In vitro, stealthiness of the PEGylated nanocapsules is evidenced by weak complement activation. Accumulation kinetics in the liver and the spleen was performed by (19)F MRI in mice, during the first 90 min after intravenous injection. In the liver, plain nanocapsules accumulate faster than their PEGylated counterparts. We observe PEGylated nanocapsule accumulation in CT26 xenograft tumor 7 h after administration to mice, whereas plain nanocapsules remain undetectable, using (19)F MRI. Our results validate the use of diblock copolymers for PEGylation to increase the residence time of nanocapsules in the blood stream and to reach tumors by the Enhanced Permeation and Retention (EPR) effect.


Subject(s)
Colonic Neoplasms/pathology , Fluorocarbons , Magnetic Resonance Imaging/methods , Nanocapsules/chemistry , Animals , Cell Line, Tumor , Contrast Media/chemical synthesis , Female , Fluorocarbons/chemistry , Hydrocarbons, Brominated , Mice , Mice, Nude , Nanocapsules/ultrastructure , Particle Size
15.
Biomaterials ; 33(19): 4936-46, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22498298

ABSTRACT

Countless studies showed that [60]fullerene (C(60)) and derivatives could have many potential biomedical applications. However, while several independent research groups showed that C(60) has no acute or sub-acute toxicity in various experimental models, more than 25 years after its discovery the in vivo fate and the chronic effects of this fullerene remain unknown. If the potential of C(60) and derivatives in the biomedical field have to be fulfilled these issues must be addressed. Here we show that oral administration of C(60) dissolved in olive oil (0.8 mg/ml) at reiterated doses (1.7 mg/kg of body weight) to rats not only does not entail chronic toxicity but it almost doubles their lifespan. The effects of C(60)-olive oil solutions in an experimental model of CCl(4) intoxication in rat strongly suggest that the effect on lifespan is mainly due to the attenuation of age-associated increases in oxidative stress. Pharmacokinetic studies show that dissolved C(60) is absorbed by the gastro-intestinal tract and eliminated in a few tens of hours. These results of importance in the fields of medicine and toxicology should open the way for the many possible -and waited for- biomedical applications of C(60) including cancer therapy, neurodegenerative disorders, and ageing.


Subject(s)
Fullerenes/pharmacology , Plant Oils/chemistry , Administration, Oral , Aging/drug effects , Animals , Fullerenes/administration & dosage , Fullerenes/chemistry , Fullerenes/pharmacokinetics , Male , Olive Oil , Oxidative Stress/drug effects , Rats , Rats, Wistar
16.
Eur J Pharm Biopharm ; 81(2): 453-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22465096

ABSTRACT

Recent reports showed that subtle modifications of nanoparticle surface properties induced dramatic changes of interactions with serum proteins. The present work was aimed to investigate the effect of the conformation of dextran chains decorating the surface of poly(alkylcyanoacrylate) (PACA) nanoparticles on the pharmacokinetic and biodistribution of a model drug associated with the nanoparticles. Doxorubicin was associated with PACA nanoparticles prepared by anionic emulsion polymerization (AEP) (Dox-AEP) and redox radical emulsion polymerization (RREP) (Dox-RREP). Nanoparticles and the free drug (f-Dox) were injected intravenously to rats to determine the pharmacokinetic and biodistribution of doxorubicin. Curves of the pharmacokinetics showed a rapid phase of distribution followed by a slower elimination phase. Pharmacokinetic parameters of the distribution phase determined for the Dox-RREP were significantly different from those of f-Dox and Dox-AEP, while no difference was observed in the elimination phase of the three formulations. Rats treated with Dox-RREP showed lower Dox concentrations in liver but higher concentrations in heart, lungs, and kidneys compared to those treated with the other formulations. Dox-RREP exhibited a new type of stealth behavior characterized by a short circulation time and a rapid distribution in highly vascularized organs bypassing the MPS. The difference in pharmacokinetic and biodistribution observed between the drugs formulated with the two types of nanoparticles was attributed to the difference in the conformation of the dextran chains stranded on the nanoparticle surface.


Subject(s)
Dextrans/chemistry , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Nanoparticles/chemistry , Animals , Anions/chemistry , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Chemistry, Pharmaceutical/methods , Drug Carriers/chemistry , Emulsions/chemistry , Emulsions/pharmacokinetics , Male , Oxidation-Reduction , Polymerization , Rats , Rats, Wistar , Surface Properties , Tissue Distribution
17.
J Alzheimers Dis ; 30(1): 155-66, 2012.
Article in English | MEDLINE | ID: mdl-22391220

ABSTRACT

The accumulation of amyloid-ß peptide (Aß) in the brain is a critical hallmark of Alzheimer's disease. This high cerebral Aß concentration may be partly caused by impaired clearance of Aß across the blood-brain barrier (BBB). The low-density lipoprotein receptor-related protein-1 (LRP-1) and the ATP-binding cassette (ABC) protein ABCB1 (P-glycoprotein) are involved in the efflux of Aß across the BBB. We hypothesized that other ABC proteins, such as members of the G subfamily, are also involved in the BBB clearance of Aß. We therefore investigated the roles of ABCG2 (BCRP) and ABCG4 in the efflux of [3H] Aß1-40 from HEK293 cells stably transfected with human ABCG2 or mouse abcg4. We showed that ABCG2 and Abcg4 mediate the cellular efflux of [3H] Aß1-40. In addition, probucol fully inhibited the efflux of [3H] Aß1-40 from HEK293-abcg4 cells. Using the in situ brain perfusion technique, we showed that GF120918 (dual inhibitor of Abcb1 and Abcg2) strongly enhanced the uptake (Clup, µl/g/s) of [3H] Aß1-40 by the brains of Abcb1-deficient mice, but not by the brains of Abcb1/Abcg2-deficient mice, suggesting that Abcg2 is involved in the transport of Aß at the mouse BBB. Perfusing the brains of Abcb1/Abcg2- and Abca1-deficient mice with [3H] Aß1-40 plus probucol significantly increased the Clup of Aß. This suggests that a probucol-sensitive transporter that is different from Abca1, Abcb1, and Abcg2 is involved in the brain efflux of Aß. We suggest that this probucol-sensitive transporter is Abcg4. We conclude that Abcg4 acts in concert with Abcg2 to efflux Aß from the brain across the BBB.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Neoplasm Proteins/metabolism , Peptide Fragments/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Acridines/pharmacology , Analysis of Variance , Animals , Blood-Brain Barrier/drug effects , Brain/anatomy & histology , Brain/metabolism , Carbon Isotopes/metabolism , Cell Line, Transformed , Glucose/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/drug effects , Microvessels/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Perfusion , Tetrahydroisoquinolines/pharmacology , Time Factors , Transfection , Tritium/metabolism
18.
Mol Cell Biochem ; 357(1-2): 397-404, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21660464

ABSTRACT

We investigated the expression and function of Abca1 in wild-type C57BL/6, abca1(+/+), and abca1(-/-) mice brain capillaries forming the blood-brain barrier (BBB). We first demonstrated by quantitative RT-PCR and Western immunoblot that Abca1 was expressed and enriched in the wild-type mouse brain capillaries. In abca1(-/-) mice, we reported that the lack of Abca1 resulted in an 1.6-fold increase of the Abcg4 expression level compared to abca1(+/+) mice. Next, using the in situ brain perfusion technique, we showed that the [(3)H]cholesterol brain uptake clearance (Cl(up), µl/s/g brain), was significantly increased (107%) in abca1(-/-) mice compared to abca1(+/+) mice, meaning that the deficiency of Abca1 conducted to a significant decrease of the cholesterol efflux at the BBB level. In addition, the co-perfusion of probucol (Abca1 inhibitor) with [(3)H]cholesterol resulted in an increase of [(3)H]cholesterol Cl(up) (115%) in abca1(+/+) but not in abca1(-/-) mice, meaning that probucol inhibited selectively the efflux function of Abca1. In conclusion, our results demonstrated that Abca1 was expressed in the mouse brain capillaries and that Abca1 functions as an efflux transporter through the mouse BBB.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Blood-Brain Barrier/metabolism , Cholesterol/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G , ATP-Binding Cassette Transporters/drug effects , Animals , Brain/metabolism , Brain/surgery , Mice , Mice, Inbred C57BL , Mice, Transgenic , Probucol/pharmacology
19.
Neuropharmacology ; 56(4): 808-13, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19705573

ABSTRACT

Combined evidence from neuroimaging and neuropathological studies shows that signs of vascular pathology and brain hypoperfusion develop early in Alzheimer's disease (AD). To investigate the functional implication of these abnormalities, we have studied the cerebrovascular volume and selected markers of blood-brain barrier (BBB) integrity in 11-month-old 3 x Tg-AD mice, using the in situ brain perfusion technique. The cerebrovascular volume of distribution of two vascular space markers, [3H]-inulin and [14C]-sucrose, was significantly lower (-26% and -27%, respectively; p < 0.01) in the brain of 3 x Tg-AD mice compared to non-transgenic littermates. The vascular volume reduction was significant in the hippocampus (p < 0.01), but not in the frontal cortex and cerebellum. However, the brain transport coefficient (Clup) of [14C]-D-glucose (1 microM) and [3H]-diazepam was similar between 3xTg-AD mice and controls, suggesting no difference in the functional integrity of the BBB. We also report a 32% increase (p < 0.001) in the thickness of basement membranes surrounding cortical microvessels along with a 20% increase (p < 0.05) of brain collagen content in 3xTg-AD mice compared to controls. The present data indicate that the cerebrovascular space is reduced in a mouse model of Abeta and tau accumulation, an observation consistent with the presence of cerebrovascular pathology in AD.


Subject(s)
Alzheimer Disease/pathology , Blood Vessels/pathology , Cerebrovascular Circulation/physiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Blood Vessels/metabolism , Brain/pathology , Brain Chemistry/drug effects , Collagen Type I/metabolism , Collagen Type IV/metabolism , Diazepam/metabolism , GABA Modulators/metabolism , Glucose/metabolism , Humans , Immunohistochemistry , Mice , Mice, Transgenic , tau Proteins/metabolism
20.
Neurochem Int ; 55(7): 476-82, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19442696

ABSTRACT

Docosahexaenoic (DHA) and eicosapentaenoic (EPA) acids are n-3 polyunsaturated fatty acids with a therapeutic potential for CNS diseases. Here, using an in situ brain perfusion technique in mice, we show that [(14)C]-DHA and [(14)C]-EPA readily cross the mouse blood-brain barrier (BBB) with brain transport coefficients (Clup) of 48+/-3microlg(-1)s(-1) and 52+/-4microlg(-1)s(-1), respectively. Mechanical capillary depletion of brain homogenates showed that less than 10% of [(14)C]-DHA or [(14)C]-EPA remained in endothelial cells of the brain vasculature, demonstrating that both molecules fully crossed the BBB. Addition of bovine serum albumin decreased the Clup of [(14)C]-DHA to 0.6+/-0.3microlg(-1)s(-1), indicating that binding to albumin reduced importantly, but not totally, the passage of DHA through the BBB. The Clup of [(14)C]-DHA or [(14)C]-EPA was not saturable at concentration up to 100microM, suggesting that these compounds crossed the BBB by simple diffusion. However, long-term high-DHA dietary consumption reduced the Clup of [(14)C]-DHA to 33+/-6microlg(-1)s(-1) (-20%, p<0.01). These results confirm that the brain uptake of DHA or EPA perfused with a physiological buffer is comparable to highly diffusible drugs like diazepam, and can be modulated by albumin binding and chronic dietary DHA intake.


Subject(s)
Blood-Brain Barrier/physiology , Cerebrovascular Circulation/physiology , Docosahexaenoic Acids/pharmacokinetics , Eicosapentaenoic Acid/pharmacokinetics , Algorithms , Animals , Brain Chemistry/physiology , Capillaries/physiology , Chromatography, Gas , Chromatography, High Pressure Liquid , Diet , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Perfusion
SELECTION OF CITATIONS
SEARCH DETAIL