己知a÷b二50,当axiological,b÷io当,商是多少

林子乔喜欢的音乐 - 歌单 - 网易云音乐
林子乔喜欢的音乐
播放:55次
网易云音乐多端下载
同步歌单,随时畅听320k好音乐
网易公司版权所有(C)杭州乐读科技有限公司运营:Associated material
Related literature
Other articles by authors
Related articles/pages
Download to ...
Share this article
Email updates
Activation of microglial cells triggers a release of brain-derived neurotrophic factor (BDNF) inducing their proliferation in an adenosine A2A receptor-dependent manner: A2A receptor blockade prevents BDNF release and proliferation of microglia
Catarina Gomes*, Raquel Ferreira, Jimmy George, Rui Sanches, Diana I Rodrigues, Nélio Gon?alves and Rodrigo A Cunha
Corresponding author:
Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, Coimbra, , Portugal
FMUC - Faculty of Medicine, University of Coimbra, Coimbra, , Portugal
Faculty of Pharmacy, University of Coimbra, Coimbra, , Portugal
For all author emails, please .
Journal of Neuroinflammation 2013, 10:16&
doi:10.94-10-16
The electronic version of this article is the complete one and can be found online at:
Received:7 March 2012
Accepted:18 January 2013
Published:30 January 2013
& 2013 Gomes et al.; licensee BioMed Central Ltd.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Background
Brain-derived neurotrophic factor (BDNF) has been shown to control microglial responses
in neuropathic pain. Since adenosine A2A receptors (A2ARs) control neuroinflammation, as well as the production and function of BDNF, we
tested to see if A2AR controls the microglia-dependent secretion of BDNF and the proliferation of microglial
cells, a crucial event in neuroinflammation.
Murine N9 microglial cells were challenged with lipopolysaccharide (LPS, 100 ng/mL)
in the absence or in the presence of the A2AR antagonist, SCH58261 (50 nM), as well as other modulators of A2AR signaling. The BDNF cellular content and secretion were quantified by Western blotting
and ELISA, A2AR density was probed by Western blotting and immunocytochemistry and cell proliferation
was assessed by BrdU incorporation. Additionally, the A2AR modulation of LPS-driven cell proliferation was also tested in primary cultures
of mouse microglia.
LPS induced time-dependent changes of the intra- and extracellular levels of BDNF
and increased microglial proliferation. The maximal LPS-induced BDNF release was time-coincident
with an LPS-induced increase of the A2AR density. Notably, removing endogenous extracellular adenosine or blocking A2AR prevented the LPS-mediated increase of both BDNF secretion and proliferation, as
well as exogenous BDNF-induced proliferation.
Conclusions
We conclude that A2AR activation plays a mandatory role controlling the release of BDNF from activated
microglia, as well as the autocrine/paracrine proliferative role of BDNF.
Keywords: Adenosine A2A N M BDNFBackground
Neurodegenerative pathologies, such as Alzheimer’s or Parkinson’s disease, progress
with a sustained, chronic neuroinflammatory process [,]. Microglial cells, which display specialized immune functions in the central nervous
system, play a crucial role in neuroinflammation [,], undergoing a process globally known as microgliosis. Microgliosis encompasses morphological
changes, cell proliferation and modifications in the synthesis and secretion of both
pro- and anti-inflammatory substances [], whose temporal balance determines the contention or the perpetuation of neuroinflammation.
Thus, neuroinflammation is a tightly regulated process involving several modulators,
including adenosine []. The relation between adenosine and neuroinflammation is heralded by the key role
of adenosine A2A receptors (A2AR) in peripheral inflammation [], as well as by the localization of A2AR in microglial cells and the ability of A2AR to control several microglial functions, namely chemotaxis [] and the production of inflammatory mediators [,].
One of the most notable modulatory properties of adenosine is the ability of A2AR activation to potentiate the production, secretion and effects of different trophic
factors []. This effect has been considered neuroprotective, in accordance with the ability
of trophic factors to prevent neuronal damage or to promote neuronal repair [-]. However, it seems to contradict the well-described neuroprotective role mediated
by A2AR antagonism [,]. BDNF (brain-derived neurotrophic factor), which is mostly recognized as a pro-survival
factor, has also been shown to support microglial activation in vivo[]. This could result in a potential amplification of microgliosis and neuroinflammation,
as occurs in neuropathic pain [,], with BDNF release resulting either through neuronal activity-dependent exocytosis
or from microglia using an exocytotic pathway or a constitutive release pathway present
in all cell types (reviewed in []), both of which are prone to modulation [,]. Therefore, BDNF emerges as a potential candidate for mediating the switch between
acute neuroinflammation (which is beneficial in resolving brain-noxious stimuli) and
chronic neuroinflammation, which is thought to be a main player in the progression
of neurodegenerative diseases [].
To date, it has not been explored whether A2AR controls microglial levels of neurotrophic factors or its involvement in autocrine/paracrine
actions potentially sustaining microgliosis over time. Thus, this study was designed
to test whether the A2AR blockade modulates BDNF secretion and microglial proliferation induced by the inflammatory
trigger lipopolysaccharide (LPS), which would indicate that A2AR acts as a key controller of microglial function and neuroinflammation.
Cell line and primary microglial cultures
A murine microglial cell line, N9 (a kind gift from Professor Claudia Verderio, CNR
Institute of Neuroscience, Cellular and Molecular Pharmacology, Milan, Italy), was
grown in an RPMI medium supplemented with 30 mM glucose (Sigma, Sintra, Portugal),
100 U/mL penicillin and 100 μg/mL streptomycin (GIBCO, Invitrogen, Porto, Portugal).
Primary microglial cultures were prepared as previously described []. Briefly, primary cultures of glial cells were obtained from a postnatal (P1-P5)
C57BL6 mouse and maintained for 15 days in the DMEM-F12 medium with glutamax (Invitrogen)
containing 10% fetal bovine serum (Invitrogen), 0.25% gentamycin (Invitrogen) and
0.25 ng/mL M-CSF (murine-colony stimulating factor, Peprotech, Rocky Hill, New Jersey,
USA). Microglia were then separated from the mixed primary culture by shaking (200
rpm for 2 hours), and plated in the DMEM-F12 medium with glutamax containing 0.25%
gentamycin (Invitrogen).
Cells were kept at 37°C under a humidified atmosphere with 95% O2 and 5% CO2. Viable cells (identified by counting trypan-blue-excluding cellular elements) were
plated at a density of 5 × 105 cells per cm2 in 6 well trays for Western blotting and enzyme-linked immunosorbent assay (ELISA)
or 1 × 105 cells per cm2 in 12 well trays for proliferation and immunocytochemistry studies.
Microglial cell pharmacological treatment
In order to evaluate the ability of an inflammatory trigger to control the cellular
content of BDNF over time, N9 cells were challenged with 100 ng/mL LPS (from Escherichia coli, serotype 055:B5 from Sigma) for 3, 6 and 12 hours. This concentration of LPS was
chosen since it was previously shown to induce changes of A2AR density in microglial cells [], which we now aim to pharmacologically manipulate to modulate BDNF secretion/function.
Thus, microglial cells were pre-incubated (20 minutes before LPS) with a supra-maximal
and selective concentration (50 nM) of an A2AR antagonist, SCH58261 [], which was present until the end of the experiment (3, 6 or 12 hours). Given that
we were able to identify the tipping time point of the changes in BDNF levels and
A2AR modulation effects, subsequent experiments were carried out at this time point (6
hours). In all these experiments, 20 to 30 minutes before adding LPS, N9 cells were
incubated with different modulators used in supra-maximal and selective concentrations
gauged from our previous experience in different preparations [,]: adenosine deaminase (ADA, 1 U/mL, which removes
Sigma), H89
(1 μM, a protein kinase A (PKA) Tocris, Madrid, Spain), chelerythrine (6
μM, a protein kinase C (PKC) Calbiochem, Lisbon, Portugal), or anti-human
BDNF polyclonal antibody (10 μg/mL; Promega, Lisbon, Portugal), a concentration chosen
according to []. ADA and the anti-human BDNF polyclonal antibody were directly diluted in the culture
H89 and chelerythrine were made up to a 25 mM stock solution in water to dilute
in the culture medium.
In the experiments carried out in the absence of LPS, N9 cells were incubated with
PKA upstream modulators used in concentrations gauged from our previous experience
in different preparations (for example [,]), namely: CGS21680 (30 nM; Sigma), an A2AR forskolin (1 μM; Ascent Scientific, Cambridge, UK), an activator
8-Br-AMP (5 μM; Tocris), a cyclic AMP (cAMP) analog, or with
exogenously added BDNF (Sigma) used in a concentration (20 ng/mL) able to modulate
A2AR-mediated neuronal functions []. BDNF was directly diluted in the culture medium, whereas CGS21680 and forskolin
were made up in dimethyl sulfoxide (100 mM) and 8-Br-AMP was made up in water (100
mM) to dilute in the culture medium. We always tested the impact of the vehicles of
the drugs and modulators on each measure and found that all the used vehicles were
devoid of effects (data not shown).
Western blotting
Cell lysates were obtained in a lysis solution containing 150 mM NaCl, 50 mM Tris–HCl,
1 mM ethylenediamine tetraacetic acid (EDTA), 1% NP-40 Igepal (Sigma, Sintra, Portugal),
0.1% sodium dodecyl sulfate (SDS), 0.5% sodium deoxycolate, 1 mM (phenylmethylsulfonyl
fluoride (PMSF), 1 mM sodium ortovanadate, 1 mM NaF, 1 μg/mL CLAP (protease inhibitor
Sigma). After cell scraping for homogenization, the total amount of protein
was quantified using the bicinchoninic acid (BCA; Thermo Scientific, Loures, Portugal)
method. Samples were then loaded onto gels with 7.5% (to detect A2AR, 50 kDa and pro-BDNF, 37.5 kDa) or 15% (to detect mBDNF, 13 kDa) of acrylamide plus
bisacrylamide (BioRad, Amadora, Portugal); proteins were separated by electrophoresis
(100 to 120 V for 1 hour) using a bicine-buffered solution (20 mM Tris, 192 mM bicine
and 0.1% SDS, pH 8.3) and then transferred (300 mV, 100 minutes, 4°C) to polyvinylidene
difluoride (PVDF) membranes (0.45 μm pore diameter) (GE Healthcare, Little Chalfont,
Buckinghamshire, UK). Blots were then blocked for 1 hour at room temperature (RT)
with 5% low-fat milk in Tris-buffered saline (20 mM Tris, 140 mM NaCl, pH 7.6, TBS)
with 0.1% Tween 20 (TBS-T) and incubated overnight at 4°C with primary antibodies
diluted in TBS-T with 0.5% low-fat milk. The tested primary antibodies were mouse
monoclonal anti-BDNF (1:1000; Sigma) and mouse anti-A2AR (1:1000; Millipore, Lisbon, Portugal). After rinsing three times with 0.5% low-fat
milk in TBS-T, membranes were incubated for 1 hour at RT with alkaline phosphatase-conjugated
secondary antibodies (1:2000; Amersham, Piscataway, New Jersey, USA). Protein immunoreactive
bands were visualized in a Versa Doc Imaging System (Model 3000, BioRad Laboratories),
after the incubation of the membrane with enhanced chemofluorescence reagent (ECF;
GE Healthcare).
Re-probing of the same membranes with a different antibody was achieved by washing
the ECF in 40% methanol for 30 minutes and stripping the previous antibodies in a
solution of 0.2 M glycine with 0.1% SDS and 1% (v/v) Tween 20, pH 2.2, for 1 hour.
After washing (3 times with TBS-T for 20 minutes), membranes were blocked and incubated
with primary and respective secondary antibodies, namely for mouse anti-α-tubulin
(1:20000; Sigma) to confirm that similar amounts of sample were loaded to the different
Immunocytochemistry
Cells were fixed with 4% paraformaldehyde (Sigma) and permeabilized for 20 minutes
in 0.25% Triton X-100 (Sigma) in a phosphate-buffered saline (PBS) solution (137 mM
NaCl, 2.7 mM KCl, 10 mM NaH2PO4, 1.8 mM KH2PO4, pH 7.4). Unspecific binding was prevented by incubating cells in PBS with 3% bovine
serum albumin (BSA) and 5% normal horse serum. Cells were incubated overnight at 4°C
in PBS with 3% BSA and 5% normal horse serum and including the primary antibody (mouse
anti-A2A, 1:250; Millipore). Cells were then washed and incubated for 1 hour at RT with an
Alexa Fluor 488 donkey anti-mouse (1:400; Molecular Probes, Lisbon, Portugal) secondary
antibody. Membrane ruffling, which is characteristic of activated microglia, was probed
using a marker for filamentous actin, phalloidin, by incubating cells for 20 minutes
at RT with PBS containing actin-stain 670 fluorescent phalloidin (1:75; Cytoskeleton,
Denver, USA). For nuclear labeling, N9 cells were stained with DAPI (0.1 mg/mL; Invitrogen).
The coverslips were mounted in the Prolong Gold Antifade fluorescent medium (Invitrogen).
In order to check for non-specific labeling of the secondary antibodies or cross-reactivity
between secondary antibodies, staining was tested in the absence of each primary antibody
and in the absence of both primary antibodies. Fluorescent images were acquired using
a Zeiss Imager Z2 fluorescence microscope equipped with an AxioCam HRm and 63x Plan-ApoChromat
oil objective (1.4 numerical aperture), with Axiovision SE64 4.8.2 software. Ten images
were randomly taken from each coverslip (three per condition in each experiment).
BrdU incorporation assay
Microglial proliferation was evaluated by measuring the incorporation of 5-bromo-2'-deoxyuridine
(BrdU; Sigma), a synthetic nucleoside that can be incorporated into newly synthesized
DNA, replacing thymidine during cell replication. Cells were incubated with BrdU (10
μM) for the last 2 hours of pharmacological treatment, fixed in 4% paraformaldehyde,
washed in TBS with 0.3% Triton X-100 and maintained in 1 M HCl at 37°C for 30 minutes.
Non-specific binding was prevented by incubation for 1 hour in TBS with 3% BSA and
1% Triton X-100. Cells were incubated overnight at 4°C with a primary rat antibody
anti-BrdU (1:100; Serotec, Oxford, UK) in a 0.1% Triton X-100 and 0.3% BSA solution,
washed and incubated for 2 hours at RT with an Alexa Fluor 594 donkey anti-rat secondary
antibody (1:200; Molecular Probes). For nuclear staining, cells were incubated for
5 minutes at RT with Hoechst 33342 (10 μg/mL; Molecular Probes) in 0.3% BSA, and mounted
in Dakocytomation fluorescent medium (Dakocytomation Inc., California, USA). Fluorescent
images were acquired using an Axioskop 2 Plus fluorescence microscope (Zeiss, PG-Hitec,
Lisbon, Portugal). The number of proliferating cells (BrdU-positive) was counted and
expressed as a percentage of the total cells stained with Hoechst 33342 [].
BDNF enzyme-linked immunosorbent assay
The extracellular levels of BDNF were measured in the supernatant of N9 cells using
the BDNF Emax Immunoassay System (Promega, Madison, USA). Each well of a 96-well polystyrene
plate was incubated overnight at 4°C with 80 μL of anti-BDNF monoclonal antibody (1:1000)
in coating carbonate buffer (50 mM, pH 9.7). Non-adsorbed antibody was discarded and
rinsed off by washing once in a TBS-T buffer (20 mM Tris–HCl, pH 7.6, 150 mM NaCl
and 0.05% (v/v) Tween 20). Unspecific binding was prevented by blocking with 80 μL
Promega 1x Block and Sample Buffer (BB 1x) for 1 hour at RT. Plates were then washed
(as mentioned before) and 80 μL of each sample or standard (7.8 pg/mL to 500 pg/mL)
were loaded in triplicate to the plates (2 hours with 300 rpm shaking at RT). After
washing (5 times in TBS-T wash buffer), 80 μL of anti-human BDNF polyclonal antibody
(1:500 in BB 1x) was added to each well and the plates were incubated at RT (2 hours
with 300 rpm shaking). After washing (5 times in TBS-T wash buffer), the plates were
incubated for 1 hour with shaking (300 rpm) with 80 μL anti-IgY horseradish peroxidase
conjugate (1:200 in BB 1x). After the last wash with the TBS-T buffer, 80 μl of TMB
One solution was used as developer and the reaction was stopped by adding 80 μL of
HCl 1 M. Absorbance was measured at 450 nm. BDNF levels are reported as pg/mL normalized
per total amount of protein.
Data analysis
Values are presented as mean ± standard error of the mean (SEM) of n experiments. Either a Student’s t test for independent means or a one-way analysis of variance (ANOVA) followed by
a Newman–Keuls post hoc test, was used to define statistical differences between absolute values, which were
considered significant at P & 0.05 unless otherwise specified. Note that although the impact of several drugs
and modulators are presented as percentage values for the sake of clarity, the statistical
comparisons were always carried out using the absolute values.
LPS induces time-dependent changes in BDNF cellular levels, an effect dependent upon
adenosine A2A receptor tonic activation
To explore the ability of A2AR to modulate BDNF levels in microglial cells in response to an inflammatory stimulus,
we used a microglial cell line (N9), which has been successfully used previously to
dissect classical microglial responses (secretion of inflammatory mediators, microglial
proliferation and phagocytosis) in inflammatory-like conditions [,]. N9 cells were activated with LPS, a component of the Gram-negative bacteria cell
membrane, which is a well-characterized inflammatory stimulus able to induce microglial
activation and the subsequent secretion of trophic factors, including BDNF (for example
We began testing the time course (3 up to 12 hours) of the impact of LPS (100 ng/mL)
on the intracellular levels of BDNF. BDNF is synthesized as a precursor protein (pro-BDNF),
which is subsequently cleaved intra- and/or extracellularly (by peptidases and convertases)
to form the mature protein (mBDNF) [,,], which can be detected by Western blot analysis (37.5 and 13 kDa, respectively).
In the present experimental conditions, LPS (100 ng/mL) did not affect the cellular
levels of pro-BDNF at any given time point (Figure A,B, P & 0.05 compared with non-treated cells). However, 6 hours of exposure to LPS induced
a decrease of the cellular levels of the mature protein (74.9 ± 4.3%, n = 7, P & 0.001 compared with non-treated cells), which returned to values similar to those
observed in non-treated cells 6 hours later, that is, at 12 hours of exposure to LPS
(Figure A,B).
Effect of LPS on the intracellular levels of pro- and mature BDNF in murine N9 microglial
cells. (A) N9 cells were exposed to LPS (100 ng/mL) for 3, 6 and 12 hours, then lysed and homogenized
for Western blot analysis of pro- (open bars) and mature BDNF (filled bars) immunoreactivities
(37.5 and 13 kDa, respectively). Results are expressed as mean ± SEM of n (as indicated in each bar) independent experiments (*** P & 0.001, compared with non-treated cells, using the Newman–Keuls multiple comparison
test) and 100% represents the pro- and mBDNF in cells that were not exposed to LPS.
(B) Representative blot of the LPS (100 ng/mL for 6 hours)-mediated decrease of intracellular
mature BDNF and its inability to interfere with the intracellular levels of pro-BDNF;
the blots compare BDNF immunoreactivity from cells exposed (+) or not (-) to LPS.
BDNF, brain-derived LPS, mBDNF, mature protein
BDNF; SEM, standard error of the mean.
We have previously reported that LPS is able to induce microgliosis and the production
of pro-inflammatory mediators under A2AR control []. In parallel, it is known that A2AR modulates BDNF levels both in neurons [] and in native tissue []. Since this effect has not yet been reported in microglia, we tested the ability
of a supra-maximal and selective concentration (50 nM) of an A2AR antagonist, SCH58261 [], to control BDNF levels in the absence and in the presence of LPS. As shown in Figure
, the A2AR antagonist prevented the LPS-induced decrease of mBDNF at 6 hours (129.5 ± 23.04%,
n = 5, P & 0.01 compared with LPS-treated cells, Figures B,D), whereas SCH58261 was devoid of effects on BDNF levels either when LPS has no
effects (that is, at 3 or 12 Figure A,C) or in the absence of LPS (P & 0.05 compared with non-treated cells or LPS-treated cells for 3 and 12 hours).
Effect of LPS on the intracellular levels of pro- and mature BDNF in the presence
of a selective A2AR antagonist in murine N9 microglial cells. The A2AR antagonist SCH58261 (50 nM) prevented the LPS (100 ng/mL)-induced modification of
mature BDNF levels selectively at 6 hours (B). In contrast, the A2AR blockade failed to affect the levels of pro- and mature BDNF at 3 hours (A) or 12 hours (C) of LPS exposure. Note that A2AR only affected BDNF levels when they were challenged with LPS. Results are expressed
as mean ± SEM of n (as indicated in each bar) independent experiments (*** P & 0.001, compared with non- ·· P & 0.01, compared with LPS-treated cells, using the Newman–Keuls multiple comparison
test) and 100% represents the pro- and mBDNF in cells that were not exposed to LPS.
(D) Representative blot of the modulation by SCH58261 of intracellular mature BDNF in
cells challenged for 6 hours with LPS. We verified (not shown) that none of the vehicles
of the tested drugs (water or dimethyl sulfoxide) modified BDNF levels. A2AR, A2A BDNF, brain-derived LPS, mBDNF,
mature protein BDNF; SEM, standard error of the mean.
LPS-induced decrease of BDNF cellular levels is related to an increase of BDNF secretion,
an effect dependent upon a tonic activation of A2AR
The LPS-induced decrease of mBDNF at 6 hours of exposure may be explained by changes
in BDNF expression and/or secretion. The fact that LPS did not influence the levels
of pro-BDNF (see Figures
and ) suggests that changes in secretion are more likely to be involved. Thus, we tested
the influence of LPS upon BDNF secretion by measuring free mature BDNF levels in the
culture medium, as assessed by ELISA. Endogenous levels of BDNF in non-treated cells
(196.2 ± 37.6 pg/mL, n = 9, Figure A) were increased by 147.3 ± 12% (n = 9, P & 0.01) in the presence of LPS (100 ng/mL for 6 hours), reaching extracellular values
of 272.1 ± 51.3 pg/mL (Figure A,B). The A2AR antagonist, SCH58261 (50 nM), notably prevented this LPS-induced BDNF secretion
(93.6 ± 8.7%, n = 9, P & 0.01 compared with LPS-treated cells, Figure B). These results indicate that A2AR activation by endogenous adenosine is required to allow the LPS-induced secretion
of BDNF. Accordingly, the presence of ADA (1 U/mL), which removes endogenous adenosine
thus impairing A2AR activation, also prevented the effect of LPS upon BDNF release (104.7 ± 4.2%, n = 5, P & 0.01 compared with LPS-treated cells, Figure B).
Endogenous extracellular adenosine, through activation of A2AR, mediates the LPS-induced enhancement of BDNF secretion from microglial N9 cells. Cells were incubated with LPS (100 ng/mL for 6 hours) in the absence or in the presence
of the A2AR antagonist, SCH58261 (50 nM) or in the absence or presence of adenosine deaminase
(ADA, 1 U/mL), which converts adenosine into its inactive metabolite, inosine. The
incubation medium was collected to quantify the extracellular free mature BDNF by
ELISA. (A) Comparison between absolute values of BDNF (pg/mL) in the absence and in the presence
of LPS. (B) LPS increased the extracellular levels of BDNF, an effect prevented by A2AR antagonism and by adenosine removal from the medium with ADA (
100% represents BDNF levels in cells that were not exposed to LPS). Results are expressed
as mean ± SEM of n (as indicated in each bar) independent experiments (** P & 0.01, compared with non- ·· P & 0.01, compared with LPS-treated cells, using the Newman–Keuls multiple comparison
test). We verified (not shown) that none of the vehicles of the tested drugs (water
or dimethyl sulfoxide) modified BDNF levels. A2AR, A2A ADA, BDNF, brain-derived LPS,
SEM, standard error of the mean.
A2AR control the LPS-induced secretion of BDNF through the cAMP-PKA pathway
The key role of A2AR in mediating the LPS-induced secretion of BDNF suggests that the direct activation
of A2AR should be able to trigger the release of BDNF from microglial N9 cells. Indeed,
the selective A2AR agonist, CGS21680 (30 nM), enhanced per se (that is, in the absence of LPS) the secretion of BDNF (146.2 ± 14.3%, n = 4, P & 0.05 compared with non-treated cells) to an extent similar to the effect of LPS
(P & 0.05 compared with LPS-treated cells), an effect prevented by the A2AR antagonist SCH58261 (50 nM) (96.3 ± 10%, n = 4, P & 0.01 compared with CGS21680-treated cells) (Figure A).
The LPS-induced enhancement of BDNF secretion mediated by A2AR involves the recruitment of the AC-cAMP-PKA transducing pathway, whereas PKC controls
the constitutive release of BDNF from N9 microglial cells. In all experiments, the medium was collected after 6 hours for quantitative analysis
of extracellular BDNF (ELISA). (A) Cells were incubated with the A2AR agonist CGS21680 (30 nM) in the absence or in the presence of the A2AR antagonist SCH58261 (50 nM) or LPS (100 ng/mL). (B) Cells were incubated with the adenylyl cyclase (AC) activator, forskolin (1 μM) or
with the cAMP analog, 8-Br-cAMP (5 μM) in the absence and in the presence of LPS (100
ng/mL). (C) Cells were incubated with the PKA inhibitor, H89 (1 μM) or with the PKC inhibitor,
chelerythrine (6 μM) in the absence and in the presence of LPS (100 ng/mL). Results
are expressed as mean ± SEM of n (as indicated in each bar) independent experiments (*** P & 0.001, ** P & 0.01, * P & 0.05, compared with non- ·· P & 0.01, compared with LPS-treated cells, using the Newman–Keuls multiple comparison
test) and 100% represents the pro- and mBDNF in cells that were not exposed to LPS.
We verified (not shown) that none of the vehicles of the tested drugs (water or dimethyl
sulfoxide) modified BDNF levels. A2AR, A2A ADA, BDNF, brain-derived cAMP,
cyclic AMP; LPS, PKA, protein kinase A; PKC, protein kinase C;
SEM, standard error of the mean.
A2AR are G protein-coupled receptors and most of their effects involve AC activation
and the subsequent raise in cAMP levels, which activate PKA (for a review, see, for
example, []). Accordingly, adenosine modulates peripheral immune responses through the activation
of A2AR and the recruitment of the cAMP-PKA pathway []. It has previously been reported that PKA notably plays a crucial role in the control
of the depolarization-evoked release of BDNF from neurons []. Thus, we tested to see if the AC-cAMP-PKA pathway would control BDNF secretion from
microglial cells. As shown in Figure B, the activation of AC with forskolin (1 μM) or the cAMP analog 8-bromo-cAMP (5 μM),
mimicked the effect of LPS, triggering BDNF secretion (147.4 ± 9.1%, n = 5, P & 0.001 compared with non-treated cells and 120.9 ± 3.4%, n = 5, P & 0.01 compared with non-treated cells) to an extent similar to that caused by LPS
(both have P & 0.05 compared with LPS-treated cells). Compatible with this scenario, we report
that the PKA inhibitor H89 (1 μM) prevented the effect of LPS upon BDNF secretion
(95.8 ± 7.7%, n = 6, P & 0.01 compared with LPS-treated cells, Figure C), further suggesting that the tonic activation by endogenous adenosine of A2AR operating through the cAMP-PKA pathway underlies the ability of LPS to trigger BDNF
secretion.
Notably, we observed that H89 failed to modify the basal outflow of BDNF from microglial
N9 cells (Figure C), indicating that the AC-cAMP-PKA pathway can be recruited by A2AR to bolster the release of BDNF upon LPS-induced microgliosis, but does not mediate
the constitutive release of BDNF. In contrast, the PKC inhibitor, chelerythrine (6
μM), enhanced the constitutive outflow of BDNF (160.3 ± 12.3%, n = 4, P & 0.05 compared with non- Figure C), but failed to affect the LPS-induced secretion of BDNF (146.4 ± 15.0%, n = 4, P & 0.05 compared with LPS- Figure C). This indicates that the transducing pathways involved in the constitutive and
LPS-induced release of BDNF are different and that the LPS-induced enhancement of
BDNF secretion selectively recruits the A2AR operating the cAMP-PKA pathway.
In accordance with our working hypothesis that the LPS-induced release of BDNF requires
the recruitment of A2AR and the activation of the AC-cAMP pathway, we expected to record an ability for
CGS21680 (an activator of A2AR), forskolin (an activator of AC) or 8-Br-cAMP to occlude the ability of LPS to enhance
BDNF release. Surprisingly, we observed that the activation of the A2AR-AC-cAMP system before the LPS challenge actually changed the set-up of the N9 cells
to such an extent that the simultaneous presence of LPS and any of the activators
of the A2AR-AC-cAMP axis (30 nM CGS21680, 1 μM forskolin or 5 μM 8-Br-cAMP) now failed to modify
(P & 0.05) the release of BDNF compared to the control (Figures A,B). This prompts the hypothesis that the LPS signaling pathway or the ability to
release BDNF might be affected by this pre-exposure to these activators of the A2AR-AC-cAMP axis, a question that warrants further detailed mechanistic investigation.
This different role for A2AR in the control of LPS-induced enhancement of BDNF secretion (LPS requires the subsequent
activation of A2AR, whereas the pre-activation of A2AR inhibited the LPS-induced release of BDNF) led us to test the hypothesis that LPS
might up-regulate A2AR to bolster BDNF secretion from N9 microglial cells. As shown in Figure , both Western blot (Figure A,B) and immunocytochemical (Figure C) analysis of N9 cells revealed an enhancement of A2AR immunoreactivity at the same time point where LPS-induced enhancement of BDNF secretion
was observed (that is, at 6 hours). The temporal correlation between the enhancement of A2AR density and the PKA-dependent increase of BDNF secretion suggests that LPS-induced
PKC-PKA shifting in the regulation of BDNF secretion may be triggered by the increased
A2AR signaling subsequent to an increase in receptor density and activation.
Effect of LPS on the density of A2AR in N9 microglial cells. (A) Representative blot of the LPS (100 ng/mL for 6 hours)-mediated increase of A2AR the blot compares A2AR immunoreactivity from cells exposed (+) or not (-) to LPS in four independent experiments.
(B) Cells were exposed to LPS (100 ng/mL for 6 hours) and then lysed and homogenized
for Western blot analysis. Quantitative analysis of A2AR immunoreactivity in the presence of LPS was compared with non-treated cells (taken
as 100%). Results are expressed as mean ± SEM of n (as indicated in each bar) independent experiments (* P & 0.05, compared with non-treated cells, using Student’s t test). (C) Immunocytochemistry confirmed the LPS-induced increase in A2AR immunoreactivity (representative images of independent experiments). Double-labeling
for A2AR (green) and for the structural protein, phalloidin (far red), was performed in all
phalloidin-labeling is not shown to allow a clear visualization of A2AR immunoreactivity (the nuclei were labeled with DAPI, in blue). (D) Magnification of cellular elements 1 (microglia in basal conditions) and 2 (activated
cell) in (C), showing in detail the increase in A2AR density in activated cells (phalloidin-labeling is shown to allow the visualization
of LPS-induced morphological changes of microglial cells). A2AR, A2A LPS, SEM, standard error of the mean.
LPS promotes microglial proliferation, an effect dependent upon BDNF and A2AR tonic activation
Given that both BDNF and A2AR activation promote neuroinflammation and A2AR was previously proposed to be required for microglial proliferation [], we next aimed to extend understanding of the tight interplay between BDNF and A2AR in the context of the control of microglial proliferation, a key process for sustaining
neuroinflammation. We posited that one of the functions of BDNF released under LPS
stimulation of microglial cells might be to promote their proliferation in order to
‘feed’ microgliosis, an effect which might also be controlled by A2AR. To begin probing this hypothesis, we first tested the impact of an antibody that
specifically recognizes and sequesters BDNF on the LPS-induced proliferation of N9
microglial cells. As anticipated, LPS (100 ng/mL for 6 hours) induced a clear proliferation
of N9 microglial cells (170.5 ± 9.2%, n = 6, P & 0.01 compared with non-treated cells). Notably, this effect was prevented when
the cells were simultaneously incubated with the antibody anti-BDNF (79.4 ± 3.5%,
n = 3, P & 0.001 compared with LPS- Figure A,B). Furthermore, in line with the ability of A2AR to control LPS-induced BDNF secretion, we observed that the A2AR antagonist, SCH58261 (50 nM), prevented the LPS-induced increase of N9 microglial
cell proliferation (90.4 ± 8.1%, n = 3, P & 0.01 compared with LPS-treated cells, Figure A,C).
LPS triggers microglial N9 proliferation in a manner dependent on extracellular BDNF
and A2AR activation. Cells were exposed to LPS (100 ng/mL) for 6 hours in the presence of BrdU for the
last 2 hours. Proliferation was quantified as the number of BrdU-labeled nuclei (red)
and expressed as a percentage of the total number of DAPI-labeled nuclei (blue). (A) Representative images illustrating the ability of LPS to enhance N9 microglial cell
proliferation, an effect prevented both by an anti-BDNF antibody (10 μg/mL) and by
the selective A2AR antagonist, SCH58261 (50 nM). Average quantitative analysis shows that anti-BDNF
antibody (B) or SCH58261 (C) prevents LPS-induced proliferation. (D) Representative images illustrating the ability of added BDNF (20 ng/mL) to enhance
N9 microglial cells proliferation, an effect prevented both by the selective A2AR antagonist, SCH58261 (50 nM) and by adenosine deaminase (ADA, 1 U/mL). Average quantitative
analysis shows the ability of SCH58261 (50 nM) and ADA (1 U/mL) to prevent BDNF effects
(E). Results are expressed as mean ± SEM of n (as indicated in each bar) independent experiments (** P & 0.01, compared with non- ··· P & 0.001, ·· P & 0.01, compared with LPS- ·· P & 0.01, · P & 0.05 compared with BDNF-treated cells using the Newman–Keuls multiple comparison
test) and 100% represents proliferation of cells that were not exposed to LPS. A2AR, A2A ADA, BDNF, brain-derived LPS,
SEM, standard error of the mean.
In order to disentangle whether the A2AR blockade was only preventing BDNF secretion or also the impact of BDNF on microglial
proliferation, we then evaluated the ability of exogenously added BDNF to directly
trigger the proliferation of N9 microglial cells, as well as the ability of SCH58261
to modulate this eventual effect. BDNF (20 ng/mL) increased the proliferation of N9
microglial cells (242 ± 48%, n = 3, P & 0.01 compared with non-treated cells), an effect prevented by the antibody anti-BDNF
(104.3 ± 4.1%, n = 3, P & 0.05 compared with BDNF-treated cells, data not shown) and by the A2AR blockade (147.7 ± 47%, n = 3, P & 0.05 compared with BDNF-treated cells) or removal of endogenous adenosine with
1 U/mL of ADA (111.2 ± 29%, n = 3, P & 0.01 compared with BDNF-treated cells, Figure D,E).
Finally, we attempted to extend the A2AR-mediated control of BDNF-induced proliferation from the context of N9 microglial
cells to that of mouse microglia. These mouse primary cultures of microglia (n = 2 in triplicate) were exposed to LPS (100 ng/mL) or to BDNF (20 ng/mL) for 6 hours:
both LPS (251.1 ± 12.8%, P & 0.01 compared with non-treated cells, Figure A,B) and BDNF (201.1 ± 2.6%,P & 0.05 compared with non-treated cells, Figure A,B) increased primary microglial proliferation, an effect prevented by the A2AR blockade with SCH58261 (50 nM).
LPS and BDNF trigger primary microglial proliferation in a manner dependent on A2AR activation. Cells were exposed to LPS (100 ng/mL) for 6 hours in the presence of BrdU for the
last 2 hours. Proliferation was quantified as the number of BrdU-labeled nuclei (red)
and expressed as a percentage of the total number of DAPI-labeled nuclei (blue). (A) Representative images illustrating the ability of LPS and BDNF to enhance primary
microglial proliferation, an effect prevented by the selective A2AR antagonist, SCH58261 (50 nM) (B). Results are expressed as mean ± SEM of two independent experiments (** P & 0.01, *** P & 0.001, compared with non- ·· P & 0.01, · P & 0.05, compared with LPS-treated cells) and 100% represents proliferation of cells
that were not exposed to LPS. A2AR, A2A ADA, BDNF, brain-derived BrdU,
5-bromo-2'- LPS, SEM, standard error of the mean.
Discussion
The present study suggest two major conclusions: 1) the activation of microglial cells
with LPS triggers a release of BDNF, which plays a key autocrine or paracrine role
by stimulating micr 2) both the LPS-induced release of BDNF as
well as the resulting proliferation of microglia is prevented by the blockade of adenosine
In response to noxious stimuli, microglia become activated by changing their morphology,
proliferating and releasing molecules, some considered to be neurotoxic and others
neuroprotective []. Irrespective of the nature of the released substances, microglial responses in the
central nervous system must be tightly regulated since the sustained activation of
microglia results in a chronic neuroinflammatory process, which is involved in neurodegeneration.
Neurotrophic factors, namely BDNF, which are produced and secreted by microglial cells
[,,], have the ability to protect neurons and to promote neuronal repair in pathologic-like
conditions [-]. Although the physiological role of BDNF in neuronal survival/repair has been extensively
investigated, the autocrine/paracrine function of BDNF secreted by microglial cells
has not been addressed and may actually play a deleterious role. Thus, BDNF can actually
behave as a switch by promoting an autocrine action in microglial cells, which may
lead to their sustained activation, as occurs in spinal cord cells during neuropathic
pain []: in this disease, BDNF exacerbates pain hypersensitivity, and strategies to prevent
BDNF-mediated microgliosis are considered to have therapeutic potential [,]. The present study with cultured N9 microglial cells provides the first direct demonstration
for a key autocrine/paracrine role of BDNF in the LPS-induced proliferation of microglial
cells: thus, LPS triggers the secretion of BDNF, which we showed to be directly responsible
for the proliferation of microglial cells.
The second major novel conclusion of this study was the ability of the A2AR blockade to prevent LPS-induced microglial proliferation, as a result of the dual
ability of A2AR to control the LPS-induced secretion of BDNF and the ability of this BDNF to trigger
microglial proliferation. These observations are in agreement with the ability of
the A2AR blockade to control in vitro microglial proliferation []; they are also heralded by the numerous reports showing that A2AR controls microglial activation, namely responses triggered by LPS [,], as well as microgliosis in animal models of inflammatory disease [,-]. In particular, the present results that both the induced secretion of BDNF as well
as its impact on microglial cells are dependent on A2AR activation, provide the first direct demonstration for a tight interplay between
A2AR and BDNF signaling in glial cells, extending the previously reported tight interaction
between A2AR and different neurotrophic factors in the control of neuronal responses [-]. In contrast to neuronal interactions between BNDF and A2AR, which were revealed in physiological-like processes, the interaction between A2AR and BDNF in microglial cells seems to occur selectively under pathologic-like conditions
typified by microglial activation. Thus, the effect of A2AR is not observed in the absence of LPS and the LPS-induced release of BDNF is notably
time-dependent and time-coincident with the increase in A2AR density, strongly suggesting a role for the up-regulation of A2AR in activated microglia as a trigger for this interaction, in agreement with the
previously reported selective role of A2AR in the pathophysiology of neuroinflammation [,,-]. It is important to recognize that the present results are in general agreement with
the observation that it is the blockade of A2AR that prevents neuroinflammation, which is in clear contrast with the anti-inflammatory
effect resulting from A2AR activation in the control of peripheral inflammation [] or central pathological conditions, in particular when peripheral cells invade the
central nervous system, as occurs through disruption of the blood–brain barrier, where
immunosuppressive actions may be mediated by A2AR activation rather than blockade [,]. The mechanism underlying this contradiction remains to be elucidated. The impact
of A2AR in controlling inflammation may depend on the type of immune cell involved, as well
as the particular conditions found in the brain parenchyma [], as previously discussed []. Considering that the A2AR gene has independent promoters [], region-specific transcriptional control of A2AR may explain differences between central and peripheral immune responses.
The final interesting conclusion from this study is the observation that LPS redirects
the intracellular pathway involved in the regulation of BDNF secretion from PKC to
PKA. Considering the parallel increase in A2AR density, this shift is likely due to an increase in A2AR signaling. Indeed, the basal outflow of BDNF seems to be affected by a blockade
of PKC but not of PKA; in contrast, the LPS-induced outflow of BDNF, shown to be mediated
by the endogenous activation of the up-regulated A2AR, was prevented by PKA rather than PKC inhibition, suggesting a reorganization of
the control of BDNF outflow upon microglial activation by LPS (Figure ).
Schematic representation of the role of A2AR in the control of BDNF release and microglial proliferation selectively in activated
microglia. (A) Constitutive release of BDNF is under the control of PKC (non-treated cells). (B) In the presence of an inflammatory trigger, LPS, the increase in A2AR density (1) and the subsequent downstream activation of AC-cAMP-PKA (2) prevails
over PKC actions, inducing an increase in BDNF release (3), which increases microglial
proliferation (4). AC, cAMP, cyclic AMP; IN, intra
PKA, protein kinase A; PKC, protein kinase C.
Conclusion
In inflammatory conditions microglial cells are activated and proliferate, which is
a beneficial response if self-limited, but can become detrimental if sustained. The
present study shows that BDNF secretion upon microglial activation plays an autocrine/paracrine
role bolstering microglial proliferation, which may drive sustained microgliosis.
Furthermore, we provide the first demonstration that adenosine A2AR play a key dual role in the control of BDNF secretion upon microglial activation,
as well as on BDNF-induced proliferation of microglial cells. This provides the first
direct demonstration for the interplay between A2AR and BDNF in glial cells and provides a novel mechanistic insight into the therapeutic
potential of A2AR antagonists to control neurodegenerative conditions where chronic neuroinflammation
is present.
Abbreviations
A2AR: A2A AC: ADA: ANOVA:
BCA: BDNF: brain-derived BrdU: 5-bromo-2'-
cAMP: cyclic AMP; ECF: enhanced chemo
EDTA: ethylenediam ELISA: enzyme-linked
LPS: mBDNF: mature protein BDNF; PBS: phosphate-
PKA: protein kinase A; PKC: protein kinase C; PMSF: phenylmet
pro-BDNF: precursor protein BDNF; PVDF: polyv RT:
SDS: so SEM: standa TBS: Tris-buffered saline
Competing interests
The authors declare that they have no competing interests.
Authors’ contribution
CG was responsible for the quantification of extracellular BDNF (ELISA), intracellular
BDNF (Western bloting, with the help of RS), primary cultures of microglia (with the
help of JG) and for the experimental design, for coordinating the project and organizing
the manuscript with the help and under the supervision of RAC. RF was in charge with
proliferation assays and image selection and organization, as well as scientific advice.
JG performed Western blot analysis of p38 and IL-1beta detection (ELISA). DR performed
Western blot analysis and immunocytochemistry for A2AR. NG performed A2AR signaling
experiments. All authors read and approved the final manuscript.
Acknowledgments
The authors wish to thank Professor Claudia Verderio from the National Research Council,
Institute of Neuroscience and Department of Medical Pharmacology, Milan, Italy, for
her generous gift of the murine N9 microglial cell line and Elisabete Augusto for
her constructive remarks. The anti-BDNF antibody was kindly provided by Professor
Carlos Duarte from the Center for Neuroscience and Cell Biology, Portugal. This work
was supported by FCT, Portugal. The cell line was maintained in the facilities of
the Microbiology Department of the Faculty of Medicine, University of Coimbra, P
the authors wish to thank Professor Teresa Gon?alves for her generous help.
References
Richardson JC,
Gentleman SM,
Brooks DJ:
Inflammatory risk factors and pathologies associated with Alzheimer’s disease. Curr Alzheimer Res 2011,
Tansey MG,
Goldberg MS:
Neuroinflammation in Parkinson’s disease: its role in neuronal death and implications
for therapeutic intervention. Neurobiol Dis 2010,
37(3):510.
Epidemiology of Alzheimer disease. Nat Rev Neurol 2011,
Kettenmann H,
Hanisch UK,
Verkhratsky A:
Physiology of microglia. Physiol Rev 2011,
91(2):461.
Di Virgilio F,
Bramanti P,
Abbracchio MP:
Purinergic signalling in inflammation of the central nervous system. Trends Neurosci 2009,
Sitkovsky M:
Role of G-protein-coupled adenosine receptors in downregulation of inflammation and
protection from tissue damage. Nature 2001,
414(6866):916.
Traynelis SF:
Adenosine A(2A) receptor mediates microglial process retraction. Nat Neurosci 2009,
12(7):872.
Local glutamate level dictates adenosine A2A receptor regulation of neuroinflammation
and traumatic brain injury. J Neurosci 2010,
30(16):5802.
Sim?es AP,
Andrade GM,
Agostinho PM,
Adenosine A2A receptors control neuroinflammation and consequent hippocampal neuronal
dysfunction. J Neurochem 2011,
117(1):100.
Sebasti?o AM,
Ribeiro JA:
Triggering neurotrophic factor actions through adenosine A2A receptor activation:
implications for neuroprotection. Br J Pharmacol 2009,
158(1):15.
Batchelor PE,
Liberatore GT,
Porritt MJ,
Donnan GA,
Howells DW:
Inhibition of brain-derived neurotrophic factor and glial cell line-derived neurotrophic
factor expression reduces dopaminergic sprouting in the injured striatum. Eur J Neurosci 2000,
12(10):3462.
Espinosa-Oliva AM,
de Pablos RM,
Villarán RF,
Argüelles S,
Venero JL,
Machado A,
Stress is critical for LPS-induced activation of microglia and damage in the rat hippocampus. Neurobiol Aging 2011,
Madinier A,
Bertrand N,
Mossiat C,
Prigent-Tessier A,
Garnier P:
Microglial involvement in neuroplastic changes following focal brain ischemia in rats. PLoS One 2010,
4(12):e8101.
Sonsalla PK,
Domenici MR,
de Mendon?a A:
Adenosine A2A receptors and brain injury: broad spectrum of neuroprotection, multifaceted
actions and ‘fine tuning’ modulation. Prog Neurobiol 2007,
83(5):310.
Kaster MP,
Agostinho PM,
Adenosine receptors and brain diseases: neuroprotection and neurodegeneration. Biochim Biophys Acta 2011,
1808(5):1380.
Intranasal brain-derived neurotrophic factor protects brain from ischemic insult via
modulating local inflammation in rats. Neuroscience 2011,
Hatcher JP,
Hughes JP,
Chaumont S,
Conquet F,
Chessell IP,
Rassendren F:
Up-regulation of P2X4 receptors in spinal microglia after peripheral nerve injury
mediates BDNF release and neuropathic pain. J Neurosci 2008,
28(44):11263.
Brain-derived neurotrophic factor contributes to spinal long-term potentiation and
mechanical hypersensitivity by activation of spinal microglia in rat. Brain Behav Immun 2011,
25(2):322.
Lessmann V,
Brigadski T:
Mechanisms, locations, and kinetics of synaptic BDNF secretion: an update. Neurosci Res 2009,
Kuczewski N,
Porcher C,
Ferrand N,
Fiorentino H,
Pellegrino C,
Kolarow R,
Lessmann V,
Gaiarsa JL:
Backpropagating action potentials trigger dendritic release of BDNF during spontaneous
network activity. J Neurosci 2008,
Salter MW:
P2X4-receptor-mediated synthesis and release of brain-derived neurotrophic factor
in microglia is dependent on calcium and p38-mitogen-activated protein kinase activation. J Neurosci 2009,
29(11):3518.
Carreira BP,
Carvalho CM,
Araújo IM:
Assessing the influence of neuroinflammation on neurogenesis: in vitro models using
neural stem cells and microglia as valuable research tools. In Neural Stem Cells and Therapy.
Edited by Tao S.
Rodrigues RJ,
Oliveira CR,
Increased adenosine A2A immunoreactivity in activated rat microglia in culture. FENS Abstract 2004,
Halldner L,
Johansson B,
Fredholm BB,
Binding of the prototypical adenosine A(2A) receptor agonist CGS 21680 to the cerebral
cortex of adenosine A(1) and A(2A) receptor knockout mice. Br J Pharmacol 2004,
141(6):1006.
Ribeiro JA:
Purinergic modulation of [3H]GABA release from rat hippocampal nerve terminals. Neuropharmacology 2000,
Sebasti?o AM,
de Mendon?a A,
Oliveira CR,
Ribeiro JA,
Enhanced adenosine A2A receptor facilitation of synaptic transmission in the hippocampus
of aged rats. J Neurophysiol 2003,
Inagaki T,
Yoshimura Y,
Komatsu Y:
Brain-derived neurotrophic factor-mediated retrograde signaling required for the induction
of long-term potentiation at inhibitory synapses of visual cortical pyramidal neurons. Neurosci Res 2008,
Fontinha BM,
Diógenes MJ,
Ribeiro JA,
Sebasti?o AM:
Enhancement of long-term potentiation by brain-derived neurotrophic factor requires
adenosine A2A receptor activation by endogenous adenosine. Neuropharmacology 2008,
54(6):924.
Horner PJ,
Kempermann G,
Palmer TD,
Winkler J,
Proliferation and differentiation of progenitor cells throughout the intact adult
rat spinal cord). J Neurosci 2000,
20(5):2218.
Ferreira R,
Cochaud S,
Xapelli S,
Neuropeptide Y inhibits interleukin-1 beta-induced microglia motility. J Neurochem 2012,
120(1):93.
Ferreira R,
Bernardino L,
Vieira OV,
Neuropeptide Y inhibits interleukin-1β-induced phagocytosis by microglial cells. J Neuroinflammation 2011,
Kermani P,
Hempstead BL:
Regulation of cell survival by secreted proneurotrophins. Science 2001,
294(5548):1945.
Farhadi HF,
Morris SJ,
Seidah NG,
Murphy RA:
Biosynthesis and post-translational processing of the precursor to brain-derived neurotrophic
factor. J Biol Chem 2001,
276(16):12660.
Cheong JH,
Activation of adenosine A2A receptor up-regulates BDNF expression in rat primary cortical
neurons. Neurochem Res 2011,
36(12):2259.
Tebano MT,
Martire A,
Potenza RL,
Pepponi R,
Domenici MR,
Schwarzschild MA,
Adenosine A(2A) receptors are required for normal BDNF levels and BDNF-induced potentiation
of synaptic transmission in the mouse hippocampus. J Neurochem 2008,
104(1):279.
Fredholm BB,
Svenningsson P,
Vaugeois JM:
Adenosine and brain function. Int Rev Neurobiol 2005,
Gebicke-Haerter PJ,
Christoffel F,
Northoff H,
Van Calker D:
Both adenosine A1- and A2-receptors are required to stimulate microglial proliferation. Neurochem Int 1996,
Lysophosphatidic acid-induced membrane ruffling and brain-derived neurotrophic factor
gene expression are mediated by ATP release in primary microglia. J Neurochem 2008,
107(1):152.
Furukawa S,
Nakajima K,
Furukawa Y,
Kohsaka S:
Lipopolysaccharide enhances synthesis of brain-derived neurotrophic factor in cultured
rat microglia. J Neurosci Res 1997,
50(6):1023.
Hayashi Y,
Yokoyama T,
Kohsaka S,
Nakanishi H:
Microglial Ca2+-activated K+ channels are possible molecular targets for the analgesic
effects of S-ketamine on neuropathic pain. J Neurosci 2011,
31(48):17370.
Ejarque A,
Casadó V,
Tusell JM,
Moratalla R,
Schwarzschild MA,
Serratosa J:
Adenosine A2A receptor stimulation potentiates nitric oxide release by activated microglia. J Neurochem 2005,
95(4):919.
Brothers HM,
Marchalant Y,
Caffeine attenuates lipopolysaccharide-induced neuroinflammation. Neurosci Lett 2010,
480(2):97.
Chronic caffeine treatment attenuates experimental autoimmune encephalomyelitis induced
by guinea pig spinal cord homogenates in Wistar rats. Brain Res 2010,
Paternini I,
Cipriani S,
Esposito E,
Bramanti P,
Cuzzocrea P,
Selective adenosine A2A receptor agonists and antagonists protect against spinal cord
injury through peripheral and central effects. J Neuroinflammation 2011,
Assaife-Lopes N,
Pereira DB,
Ribeiro JA,
Sebasti?o AM:
Activation of adenosine A2A receptors induces TrkB translocation and increases BDNF-mediated
phospho-TrkB localization in lipid rafts: implications for neuromodulation. J Neurosci 2010,
30(25):8468.
Sim?es PF,
Sebasti?o AM,
Ribeiro JA:
GDNF control of the glutamatergic cortico-striatal pathway requires tonic activation
of adenosine A2A receptors. J Neurochem 2009,
108(5):1208.
Ribeiro JA,
Sebasti?o AM:
Glial cell line-derived neurotrophic factor (GDNF) enhances dopamine release from
striatal nerve endings in an adenosine A2A receptor-dependent manner. Brain Res 2006,
1113(1):129.
Trincavelli ML,
Cipriani S,
Martini C:
Regulation of A(2A) adenosine receptor expression and functioning following permanent
focal ischemia in rat brain. J Neurochem 2008,
104(2):479.
Van der Putten C,
Zuiderwijk-Sick EA,
van Straalen L,
de Geus ED,
Kondova I,
IJzerman AP,
Bajramovic JJ:
Differential expression of adenosine A3 receptors controls adenosine A2A receptor-mediated
inhibition of TLR responses in microglia. J Immunol 2009,
182(12):7603.
Coelho JE,
Araújo IM,
Ferrara J,
Müller CE,
Adenosine A2A receptor antagonists exert motor and neuroprotective effects by distinct
cellular mechanisms. Ann Neurol 2008,
Adenosine A2A receptor deficiency exacerbates white matter lesions and cognitive deficits
induced by chronic cerebral hypoperfusion in mice. J Neurol Sci 2009,
285(1–2):39.
Fotheringham J,
Del Bigio MR,
Peeling J,
Geiger JD:
Adenosine A2A receptor activation reduces proinflammatory events and decreases cell
death following intracerebral hemorrhage. Ann Neurol 2001,
49(6):727.
Sitkovski MV:
Opposite modulation of peripheral inflammation and neuroinflammation by adenosine
A2A receptors. In Interaction Between Neurons and Glia in Aging and Disease.
Edited by Malva JO, Rego AC, Cunha RA, Oliveira CR.
Berlin: Springer-V
Mariani J,
Moskowitz M,
Selective inactivation or reconstitution of adenosine A2A receptors in bone marrow
cells reveals their significant contribution to the development of ischemic brain
injury. Nat Med 2004,
Sign up to receive new article alerts from Journal of Neuroinflammation}

我要回帖

更多关于 java中的io流知识总结 的文章

更多推荐

版权声明:文章内容来源于网络,版权归原作者所有,如有侵权请点击这里与我们联系,我们将及时删除。

点击添加站长微信