14 research outputs found
Comparison of the Anti-inflammatory Activity and Cellular Interaction of Brush Polymer–<i>N</i>‑Acetyl Cysteine Conjugates in Human and Murine Microglial Cell Lines
Microglia-mediated neuroinflammation is commonly associated
with
neurodegeneration and has been implicated in several neurological
disorders, such as Alzheimer’s disease and Parkinson’s
disease. Therefore, it is crucial to develop a detailed understanding
of the interaction of potential nanocarriers with microglial cells
to efficiently deliver anti-inflammatory molecules. In this study,
we applied brush polymers as a modular platform to systematically
investigate their association with murine (BV-2) and human (HMC3)
microglial cell lines in the presence and absence of the pro-inflammatory
inducer lipopolysaccharide (LPS) using flow cytometry. Brush polymers
of different sizes and shapes, ranging from ellipsoid to worm-like
cylinders, were prepared through a combination of the two building
blocks carboxylated N-acylated poly(aminoester)s
(NPAEs)-based polymers and poly(2-ethyl-2-oxazoline)-NH2 (PEtOx-NH2) and characterized by 1H NMR spectroscopy,
size exclusion chromatography, and small-angle neutron scattering.
Generally, ellipsoidal particles showed the highest cellular association.
Moreover, while no significant differences in murine cell association
were observed, the brush polymers revealed a significant accumulation
in LPS-activated human microglia compared to resting cells, emphasizing
their higher affinity to activated HMC3 cells. Brush polymers with
the highest cell association were further modified with the anti-inflammatory
agent N-acetyl cysteine (NAC) in a reversible manner.
The brush polymer–NAC conjugates were found to significantly
attenuate the production of interleukin 6 (p <
0.001) in LPS-activated HMC3 cells compared to LPS-activated BV-2
cells. Thus, the presented brush polymer–NAC conjugates showed
a high anti-inflammatory activity in human microglia, suggesting their
potential for disease-targeted therapy of microglial-mediated neuroinflammation
in the future
Sex-Dependent Changes to the Intestinal and Hepatic Abundance of Drug Transporters and Metabolizing Enzymes in the SOD1<sup>G93A</sup> Mouse Model of Amyotrophic Lateral Sclerosis
Amyotrophic lateral sclerosis (ALS) is characterized
by death and
dysfunction of motor neurons that result in a rapidly progressing
loss of motor function. While there are some data on alterations at
the blood–brain barrier (BBB) in ALS and their potential impact
on CNS trafficking of drugs, little is reported on the impact of this
disease on the expression of drug-handling proteins in the small intestine
and liver. This may impact the dosing of the many medicines that individuals
with ALS are prescribed. In the present study, a proteomic evaluation
was performed on small intestine and liver samples from postnatal
day 120 SOD1G93A mice (a model of familial ALS that harbors
a human mutant form of superoxide dismutase 1) and wild-type (WT)
littermates (n = 7/genotype/sex). Untargeted, quantitative
proteomics was undertaken using either label-based [tandem mass tag
(TMT)] or label-free [data-independent acquisition (DIA)] acquisition
strategies on high-resolution mass spectrometric instrumentation.
Copper chaperone for superoxide dismutase (CCS) was significantly
higher in SOD1G93A samples compared to the WT samples for
both sexes and tissues, therefore representing a potential biomarker
for ALS in this mouse model. Relative to WT mice, male SOD1G93A mice had significantly different proteins (Padj 1.2) in the small intestine
(male
22, female 1) and liver (male 140, female 3). This included an up-regulation
of intestinal transporters for dietary glucose [solute carrier (SLC)
SLC5A1] and cholesterol (Niemann-Pick c1-like 1), as well as for several
drugs (e.g., SLC15A1), in the male SOD1G93A mice. There
was both an up-regulation (e.g., SLCO2A1) and down-regulation (ammonium
transporter rh type b) of transporters in the male SOD1G93A liver. In addition, there was both an up-regulation (e.g., phosphoenolpyruvate
carboxykinase) and down-regulation (e.g., carboxylesterase 1) of metabolizing
enzymes in the male SOD1G93A liver. This proteomic data
set identified male-specific changes to key small intestinal and hepatic
transporters and metabolizing enzymes that may have important implications
for the bioavailability of nutrients and drugs in individuals with
ALS
Fatty Acid-Binding Protein 5 Facilitates the Blood–Brain Barrier Transport of Docosahexaenoic Acid
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 <i>in vitro</i> and <i>in vivo</i>, respectively. hFAPB5 was recombinantly expressed and purified from <i>Escherichia coli</i> C41Â(DE3) cells and the binding affinity
of DHA to hFABP5 assessed using isothermal titration calorimetry.
The impact of FABP5 siRNA on uptake of <sup>14</sup>C-DHA into immortalized
human brain microvascular endothelial (hCMEC/D3) cells was assessed.
An <i>in situ</i> transcardiac perfusion method was optimized
in C57BL/6 mice and subsequently used to compare the BBB influx rate
(<i>K</i><sub>in</sub>) of <sup>14</sup>C-DHA between FABP5-deficient
(FABP5<sup>–/–</sup>) and wild-type (FABP5<sup>+/+</sup>) 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 <sup>14</sup>C-DHA cellular uptake.
By using optimized conditions for the <i>in situ</i> transcardiac
perfusion (a 1 min preperfusion (10 mL/min) followed by perfusion
of <sup>14</sup>C-DHA (1 min)), the <i>K</i><sub>in</sub> of <sup>14</sup>C-DHA was 0.04 ± 0.01 mL/g/s. Relative to FABP5<sup>+/+</sup> mice, the <i>K</i><sub>in</sub> of <sup>14</sup>C-DHA decreased 36.7 ± 12.4% in FABP5<sup>–/–</sup> 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 Lymphatic System Plays a Major Role in the Intravenous and Subcutaneous Pharmacokinetics of Trastuzumab in Rats
Therapeutic
monoclonal antibodies are currently delivered mainly
via the intravenous route, since large volumes are often required
to deliver a therapeutic dose. Administration via the subcutaneous
route would have several therapeutic advantages; the absorption mechanisms
for antibodies dosed subcutaneously are poorly understood. This study
was conducted to develop a better understanding of the mechanisms
governing the subcutaneous absorption and trafficking of monoclonal
antibodies. Specifically, the role of the lymphatic system in the
absorption and prolonged plasma exposure of trastuzumab was explored
in thoracic lymph duct-cannulated rats after SC and IV dosing. A population
pharmacokinetic model was developed in S-ADAPT to simultaneously fit
all plasma and lymph concentrations and to predict the pharmacokinetics
in nonlymph duct-cannulated animals. The estimated absolute bioavailability
of trastuzumab after SC administration in rats was 85.5%. Following
SC administration, 53.1% of the trastuzumab dose was absorbed via
a first-order process (mean absorption time: 99.6 h) into the peripheral
lymph compartment and 32.4% of the dose was absorbed by a Michaelis–Menten
process into the central compartment. Recovery in thoracic lymph over
30 h was 26.7% after SC and 44.1% after IV administration. This study
highlights for the first time the significant role of the lymphatic
system in maintaining the long plasma exposure of trastuzumab, with
the model predicting an extensive distribution of this monoclonal
antibody into the lymph following SC and IV administration. This extensive
direct absorption from the SC injection site into lymph may enable
novel therapeutic strategies for the treatment of lymph resident metastatic
cancer
Immunohistochemical detection of TDO/IDO-1 in the CA1 and CA3 regions of hippocampus in AD patients and age and sex-matched controls.
<p>Corresponding graphs of image analysis of TDO/IDO-1 immunoreactivity in human controls (white bars) and AD patients (black bars). Columns and bars represent mean ± SEM, n = 4, ****p<0.0001, in comparison to its counterpart, WT.</p
Immunohistochemical localization of TDO in the cerebellum of 3xTg AD mice.
<p>TDO was located mainly in neurons (NeuN+) and microglia (BS-Isolectin-B4+), with minor expression in astrocytes (GFAP). Columns 2, 3 and 4 show the higher magnification views of the boxed areas in the first column (merged image).</p
(A) TRP and (B) KYN concentrations quantified using HPLC; (C) K/T ratio depicting the expression of KP activation; (D) QUIN and (E) PIC production quantified using GC/MS in the hippocampus, cerebellum, cortex and remaining brain regions of 2–4 month, 6–8
<p>Bars represent mean ± SEM, n = 4.</p
Simplified schematic of the kynurenine pathway (KP).
<p>IDO-1, indoleamine dioxygenase; TDO, tryptophan dioxygenase; AFMID, arylformamidase; KAT 1–3, kynurenine amino transferase 1, 2 and 3; KMO, kynurenine 3-hydroxylase; KYNU, kynureninase; 3HAAO, 3-hydroxyanthranilic acid oxidase; ACMSD, amino-carboxymuconate-semialdehyde decarboxylase; QPRT, quinolinate phosphoribosyltransferase.</p
Immunohistochemical detection of TDO in the white matter of cerebellum in 4-month (A), 7-month (C) and 11-month (E) WT mice, compared to the same regions in 4-month (B), 7-month (D) and 11 month (F) 3xTg AD mice.
<p>(G) Corresponding graphs of image analysis of density of TDO-immunoreactivity in 4, 7 and 11-months WT (white bars) and 3xTg AD (black bars) mice brains; mean ± SEM (n = 3) *p<0.05, in comparison to its counterpart, WT.</p
The upper panel shows the representative bands of Western blots for TDO (30 kDa) and IDO1 (45 kDa) proteins in the hippocampus and cerebellum of 8-month WT and 3xTg AD mice.
<p>GAPDH was used as a loading control. The lower panel shows the quantitative comparison expressed as relative percentage change of TDO/GAPDH or IDO1/GAPDH ratio in both regions of 8-month WT (white bars) and 3xTg AD (black bars) mouse brains. Data are presented as mean ± SEM (n = 3), **p<0.01, in comparison to its counterpart, WT.</p