You are on page 1of 18

GLIA 00:000–000 (2012)

Effects of Aging and Sensory Loss on Glial Cells


in Mouse Visual and Auditory Cortices
MARIE-ÈVE TREMBLAY,1 MARTHA L. ZETTEL,1 JAMES R. ISON,1,2 PAUL D. ALLEN,1 AND ANIA K. MAJEWSKA1*
1
Department of Neurobiology and Anatomy and Center for Visual Science, University of Rochester, Rochester, New York
2
Department of Brain and Cognitive Science, University of Rochester, Rochester, New York

KEY WORDS may be secondary to the larger age-related changes in


microglia; oligodendrocyte; morphology; phagocytosis; CBA/ glial cells that have been associated with decreased
CaJ; C57BL/6J neuroprotective function and increased neurotoxicity
(Flanary et al., 2007; Luo et al., 2010; Lynch, 2010;
Streit et al., 2008; von Bernhardi et al., 2010). In partic-
ABSTRACT
ular, previously described changes in microglia include
Normal aging is often accompanied by a progressive loss of
receptor sensitivity in hearing and vision, whose consequen- altered cytokine production (Inamizu et al., 1985; Sheng
ces on cellular function in cortical sensory areas have et al., 1998; Sierra et al., 2007), increased expression of
remained largely unknown. By examining the primary activation markers (Ogura et al., 1994; Perry et al.,
auditory (A1) and visual (V1) cortices in two inbred strains of 1993; Sheffield and Berman, 1998), and the emergence
mice undergoing either age-related loss of audition (C57BL/ of dystrophic morphologies (Streit et al., 2004).
6J) or vision (CBA/CaJ), we were able to describe cellular Central changes observed during aging may be com-
and subcellular changes that were associated with normal pounded by aging-associated diseases and by other
aging (occurring in A1 and V1 of both strains) or specifically events distinct from the normal aging process, such as
with age-related sensory loss (only in A1 of C57BL/6J or V1 exposure to toxicants, injury or infections (Bilbo, 2010;
of CBA/CaJ), using immunocytochemical electron microscopy
and light microscopy. While the changes were subtle in
Zimniak, 2008). It is very likely that the deterioration of
neurons, glial cells and especially microglia were trans- sensory perception, which is a common complaint in
formed in aged animals. Microglia became more numerous older humans and is often due to the degeneration of
and irregularly distributed, displayed more variable cell the peripheral sensory organs (Congdon et al., 2004;
body and process morphologies, occupied smaller territories, Tobias et al., 1988) may also result in central histopa-
and accumulated phagocytic inclusions that often displayed thology. In particular, the loss of either hearing or sight
ultrastructural features of synaptic elements. Additionally, severely impacts the quality of life, thus contributing to
evidence of myelination defects were observed, and aged social withdrawal, anxiety and depression (Sharts-
oligodendrocytes became more numerous and were more Hopko, 2009; Tobias et al., 1988). Whether such age-
often encountered in contiguous pairs. Most of these effects
related alterations in sensory input influence the aging
were profoundly exacerbated by age-related sensory loss.
Together, our results suggest that the age-related alteration process within the brain is not well understood, but
of glial cells in sensory cortical areas can be accelerated given the large literature describing plastic changes in
by activity-driven central mechanisms that result from neuronal circuits, synapses and glial cells in sensory
an age-related loss of peripheral sensitivity. In light of cortical areas following sensory deprivation (Hensch,
our observations, these age-related changes in sensory 2005; Tremblay et al., 2010a), we hypothesized that
function should be considered when investigating cellular, age-related changes in sensory function could influence
cortical, and behavioral functions throughout the lifespan the normal process of cortical senescence in a similar
in these commonly used C57BL/6J and CBA/CaJ mouse manner.
models. V 2012 Wiley Periodicals, Inc.
C

Additional Supporting Information may be found in the online version of this


article.
Marie-Ève Tremblay and Martha L. Zettel contributed equally to this work.
INTRODUCTION
Grant sponsors: NIH, Whitehall Foundation, The Alfred P. Sloan Foundation,
and Burroughs Wellcome Fund (a Career Award in the Biomedical Sciences;
Normal aging is accompanied by a decline in neural A.K.M.); Grant number: EY019277; Grant sponsor: NIH-NIA Research; Grant
number: AG09534; Grant sponsor: NIDCD (NIH Center); Grant number: P30
function thought to be caused by cellular and molecular DC05409; Grant sponsor: NEI; Grant number: P30 EY01319; Grant sponsors:
changes in the brain that are distinct from those associ- Schmitt Program for Integrative Brain Research, Fonds de la Recherche en Sant
e
du Quebec (FRSQ), Canadian Institutes of Health Research (CIHR) (Postdoctoral
ated with neurodegenerative diseases. There is now a Fellowships; M.E.T.).
consensus that widespread central neuronal loss does Marie-Ève Tremblay is currently at Department of Psychiatry, University of
not occur during normal aging in animals (Peters, 1996; Wisconsin, 6001 Research Park Blvd., Madison, WI 53719

Rapp and Gallagher, 1996) and humans (Anderson et *Correspondence to: Ania K. Majewska, Department of Neurobiology and Anat-
omy and Center for Visual Science, University of Rochester, 601 Elmwood Ave,
al., 1983; Cragg, 1975) and that neural dysfunction is Rochester, NY 14642. E-mail: ania_majewska@URMC.rochester.edu
likely mediated by a region-specific remodeling of synap- Received 27 October 2011; Accepted 5 December 2011
ses (Burke and Barnes, 2006; Hof and Morrison, 2004; DOI 10.1002/glia.22287
Uemura, 1985). These neuronal alterations, however, Published online in Wiley Online Library (wileyonlinelibrary. com).

C 2012
V Wiley Periodicals, Inc.
2 TREMBLAY ET AL.

tested at more than one age), 61 CBA mice and 60 C57


To disentangle the cortical effects of aging and age-
mice in the acoustic experiment (25 CBA mice tested at
related loss of sensory function, we examined two com- more than one age), and 599 mice were tested with audi-
monly used strains of inbred mice, the C57BL/6J (C57) tory evoked potentials (302 CBA and 297 C57 mice). The
and CBA/CaJ (CBA), which display similar lifespans large number of auditory brainstem response (ABR)
(30.6 months versus 28.8 months, respectively; Willott et records was available because the hearing ability of all
al., 1994) but different patterns of age-related sensory of the mice in our colony was tested with the ABR before
loss. Several experiments measuring auditory evoked their entry into other experiments.
potentials have compared hearing loss across the life-
span in these strains (Henry and Chole, 1980; Hunter
and Willott, 1987; Li and Borg, 1991; Willott, 1986; and Behavioral Testing for Acoustic and
including the present research) to show that C57 mice Photic Sensitivity: The Inhibition of
begin to lose their hearing as young adults, while CBA the Acoustic Startle Response
mice maintain their hearing thresholds until near senes-
cence. In contrast, in studies of visual loss, the C57 For acoustic startle response (ASR) testing, mice were
mouse shows the least age-related retinal degeneration individually confined to a 7 3 5 3 4 cm stainless steel
(Danciger et al., 2007), and only a modest change in wire cage located in a 1 3 1 3 1 m anechoic chamber
electroretinogram recordings at 2 years of age (Caruso (Eckel) placed within a sound attenuating room (Indus-
et al., 2010; Williams and Jacobs, 2007). Up to trial Acoustics Company). The cage was mounted on a
3 months of age, the CBA mouse has normal retinal stiff acrylic shelf just above an attached accelerometer
function (Clapcote et al., 2005; Dalke et al., 2004), but in (Statham Laboratories) that was sensitive to the vertical
this study, we provide behavioral evidence for an age- force of the startle flinch response. The output of this
related loss of photic sensitivity in this strain. transducer was passed through a bridge amplifier and
Together, the comparison of C57 and CBA mice, from integrated over a 100-ms period that began with the onset
early adulthood into senescence, provided a unique of the startle-eliciting stimulus. The startle stimulus was
opportunity to study brains at the same chronological a 110-dB (SPL) noise burst, 25 ms in duration, delivered
and biological age, one with age-related reduced audi- through a high-frequency speaker (JBL Professional). For
tory input but relatively well-maintained visual input, a human observer, a noise burst at this level and duration
and the other with a converse age-related deficit in vis- approximates the sensation produced by snapping one’s
ual input. By examining the primary auditory (A1) and middle finger off the thumb into the palm, from a distance
visual (V1) cortices of the same C57 and CBA mice using of about 6 inch from the ear. The noise stimulus was cali-
immunocytochemical electron microscopy (EM) and light brated using a 1/4 inch microphone (Bruel & Kjaer)
microscopy (LM), we were able to describe changes in placed at the level of the test cage.
neurons, microglia, astrocytes, and oligodendrocytes that Two test stimuli were used to measure age-related
appeared to be driven by aging or a combination of changes in sensitivity to acoustic and photic stimuli. In
aging and sensory loss. In particular, microglia were the photic experiment, the prestimulus was a 20-ms
found to change their distribution and morphology dur- flash of light provided by a voltage-regulated fluorescent
ing aging, while accumulating phagocytic inclusions that lamp (Sylvania), which had near instantaneous rise and
displayed ultrastructural features of synaptic elements. fall times. The lamp was placed behind a double glazed
Our results propose a complex relationship between frosted window in a sound-attenuating box to eliminate
aging and age-related sensory dysfunction (arising from the slight noise coincident with light onset. The inten-
a loss of peripheral sensitivity) in effecting cortical sity of the light was measured with a Spectra Candela
senescence. Meter (PhotoResearch) held at the level of the floor of
the test cage. The intensity of the light flash was 20 lux
and its duration was 20 ms. In the acoustic experiment,
MATERIALS AND METHODS the prestimulus was a 10-ms quiet gap (with 0 ms rise
Animals and fall times) in an otherwise continuous 70-dB SPL
wideband noise delivered through a Panasonic high-fre-
Animals were treated in strict accordance with the quency speaker. In both experiments, the animals were
University of Rochester Committee on Animal Resources tested in the dark. In the photic experiment, mice were
and the National Institutes of Health standards. All group housed for 30 min in a room illuminated by red
mice were raised from birth in a separate vivarium facil- light, followed by a 10-min period of dark adaptation in
ity within the University of Rochester under a 12 h/12 h the individual test chamber. The photic experiment con-
dark-light cycle and ambient noise levels of 40 dB sound sisted of 132 trials, averaging 17 s apart (range 15–20),
pressure level (SPL) at 2,000 kHz, dropping off linearly delivered in 11 blocks of 12 trials. The noise burst that
to 25 dB SPL at 24 kHz on a log frequency scale. elicited the reflex response was presented alone (a base-
Both male and female mice were used for this study. line control trial, presented twice in each block of 12
There was a total of 257 mice tested in the behavioral trials), or preceded by the light flash, with light-onset to
experiments, 61 CBA mice and C57 mice in the photic noise-onset lead times of 10, 20, 30, 40, 50, 70, 110, 160,
experiment (with seven CBA mice and 21 C57 mice 220 ms, with one additional ‘‘no-stimulus trial’’ to assess

GLIA
EFFECT OF AGING AND SENSORY LOSS ON GLIA 3
background spontaneous activity. In the acoustic experi- PA5) in 5 dB steps between samples from 90 dB SPL
ment, the test session consisted of 121 trials, on an aver- (but 80 dB SPL maximum at 48 kHz) until the wave
age 17 s apart, delivered in 11 blocks of 11 trials. Each IV–V complex of the ABR waveform was no longer visu-
block included two baseline control trials, eight trials in ally distinguishable from background in duplicate traces
which the startle stimulus was preceded by a 10-ms gap in the judgment of a highly experienced technician. By
in noise, the gap beginning 10, 15, 20, 30, 40, 60, 110, or convention, threshold was established as the response at
160 ms before the startle stimulus, with one additional 5 dB above this final level.
‘‘no stimulus’’ trial. The order of conditions in each block
was randomized, and the first block of trials was not
used in the data analysis, in part because the startle Immunocytochemical Electron Microscopy
reflex can be unusually large on the first few trials after
confinement in the test cage. During the daytime portion of the light/dark cycle,
Prepulse inhibition (PPI) scores were calculated as the cohorts of 3-month and 20-month old C57 and CBA mice
ratio of each subject’s mean startle response amplitude (n 5 3 littermate mice from each age and strain) were
in the prepulse condition (ASRp) compared with the no- deeply anesthetized with sodium pentobarbital (80 mg/
prepulse control baseline (ASRc), PPI 5 1 – [ASRp/ASRc]. kg, i.p.) and perfused through the aortic arch with 3.5%
Because this behavioral test depends on inhibition of the acrolein (in 100 mM phosphate buffer (PB), pH 7.4) fol-
ASR by the test stimulus, and because the strength of lowed by 4% paraformaldehyde (PFA; in 100 mM PB,
the ASR can diminish with age and advanced hearing pH 7.4). Transverse sections of the brain (50 lm thick)
loss, it was necessary to exclude mice which did not have were cut with a vibratome in ice-cooled phosphate-
statistically significant ASR: a total of three CBA mice buffered saline (PBS; 0.9% NaCl in 50 mM phosphate
were excluded, two (of 29) at 19 months and one (of buffer, pH 7.4) and immunostained with a rabbit anti-
eight) at 24 months of age; a total of 28 C57 mice were ionized calcium binding adaptor molecule 1 (IBA1) anti-
excluded, four (of 33) at 4 months, three (of 30) at 7 body (1:1,000 in blocking solution; Wako Pure Chemical
months, two (of 37) at 10 months, and 19 (of 24) at 19 Industries) as previously described (Tremblay et al.,
months of age. The data of interest were the near-peak 2010a,b). Briefly, labeling was revealed with 3,30 -diami-
mean levels of PPI for each mouse at the longer intersti- nobenzidine (DAB; 0.05 mg/mL) and hydrogen peroxide
mulus intervals, for the light flash at 70, 110, and (0.03% in buffer solution; DAB Peroxidase Substrate
160 ms before the startle, and for the gap in noise at Kit; Vector Laboratories). Immunostained sections were
intervals of 60, 110, and 160 ms before the startle. postfixed flat in 1% osmium tetroxide, dehydrated in
ascending concentrations of ethanol, impregnated in
Durcupan (Electron Microscopy Sciences) overnight at
Auditory Thresholds for Tone Pips: The Auditory RT, mounted between ACLAR-embedding films (Electron
Brainstem Response Microscopy Sciences), and cured at 55°C for 48 h. Areas
of V1 and A1, at a level approximating the transverse
Mice were anaesthetized with ketamine/xylazine (120 planes A 10.16 to A 10.72 (Franklin and Paxinos,
and 10 mg/kg) then placed on a 10-cm high platform 2008), were excised from the same embedded sections,
inside a small (57L 3 41W 3 36H cm), electrically re-embedded at the tip of resin blocks, cut with an ultra-
shielded, soundproofed booth (Industrial Acoustic Com- microtome (Reichert Ultracut E) at 65–80 nm thickness,
pany) lined with 4.5-cm-thick echo-attenuating acoustic collected on bare square-mesh grids, and examined with
foam (SonexÒ; Illbruck Acoustic). The mouse was a Hitachi 7650 transmission electron microscope.
arranged in a prone position facing a broadband electro- In each animal, pictures were randomly taken in
static loudspeaker system (TDT ES1 with ED1 speaker layers II/III of both V1 and A1, at 6,0003 and 30,0003,
driver) located 10 cm from the head and was kept warm for a total surface of >10,000 lm2 acquired per region/
during the procedure with a circulating-water heating animal. Neurons, microglia, astrocytes, oligodendrocytes,
pad. Miniature subdermal needle electrodes (Nicolet) and myelinated axons were identified according to crite-
were inserted at the vertex (reference electrode), over ria previously defined (Peters and Sethares, 2004; Peters
the bulla (active electrode), and just above the hind limb et al., 1991b). Neurons with multiple features of degen-
(ground electrode). Calibrated tone bursts (5 ms dura- erating cells, such as dark nuclei and cytoplasm, ruffling
tion, 0.5-ms rise-fall time, phase alternating 90°) were of the plasma membrane, dilation of the endoplasmic
synthesized with SigGenRP' software on a TDT RP2.1 reticulum, and extensive invagination of the nuclear
real-time processor, and presented at a rate of 10/s at membrane, were referred to as ‘‘dark’’ neurons (Norman
frequencies of 3, 6, 12, 24, 32, and 48 kHz. The software et al., 2008; Oster-Granite et al., 1996; Turmaine et al.,
included an artifact detector that rejected outlier 2000; Yang et al., 2008). In addition to their immunore-
responses. Average ABR waveforms (150 repetitions 3 2 activity for the microglia-specific marker IBA1 (Ito
duplicates) were recorded at each tested frequency/am- et al., 1998), microglia were distinguished from oligoden-
plitude combination and filtered from 3 to 1,000 Hz. drocytes by their paler cytoplasm, distinctive long
Each 10 ms waveform was comprised of 500 points stretches of endoplasmic reticulm, frequent vacuoles and
(0.02-ms sample period). At each frequency, the ampli- cellular inclusions, irregular contours with obtuse
tude of the signal was automatically attenuated (TDT angles, and smaller elongated nucleus delineated by a

GLIA
4 TREMBLAY ET AL.

narrow nuclear cistern (Tremblay et al., 2010a). Quanti- and its nearest neighbor was determined and averaged
tative analysis was carried out with ImageJ software for all microglia in the image. A spacing index was cal-
(National Institutes of Health). Neurons, ‘‘dark’’ neu- culated as the square of the average nearest neighbor
rons, microglia, astrocytes, and oligodendrocytes were distance multiplied by microglial density on a per image
counted in all the pictures for a total of 818 cells basis. This value was then averaged across all images to
analyzed. In each of these cells, lysosomal inclusions determine the value per animal.
typically containing electron-dense lipopigments associ-
ated with vacuoles (Peters et al., 1991b) were counted.
In microglia, large vesicles, vacuoles, lipid droplets, and Morphological Analysis of Individual
profiles of cellular elements (Tremblay et al., 2010a) Microglia Soma
were also counted.
For the morphological analysis of individual microglia,
images were captured using a 203 objective lens
Immunocytochemical Light Microscopy (Olympus, UPlanFL 0.5 NA) and a total of 30 microglia
were analyzed for each area in each mouse of the study.
For immunocytochemistry and histochemistry, cohorts To keep imaging across subjects uniform, we focused our
of 5 CBA mice (n 5 15) from three age groups (3–4, 12– analysis in layers II/III and only choose cells that were
13, and 20–28 months) and six C57 mice (n 5 18) from perfectly in focus. Every IBA1-immunopositive microglia
three age groups (3–4, 12–14, and 24–28 months) were in a particular photomicrograph was analyzed before
used. These age groups are referred to as 3-, 12-, and moving on to the next image as to not introduce selec-
24-month-old mice in the figures. As previously tion bias. The soma area was determined by drawing a
described (Zettel et al., 2007), mice were taken during line around the cell body using the ImageJ freeform tool
the daytime portion of the light/dark cycle and deeply and allowing the program to automatically calculate the
anesthetized with sodium pentobarbital (80 mg/kg, i.p.) area. Arbor area was determined by using the trapezoid
and perfused for 25 min with 4% PFA. The brains were tool to manually connect the most distal points of the
removed, postfixed in 4% PFA overnight, and cryopro- processes of each microglia. Arbor circularity was calcu-
tected using successive solutions of 10 and 30% sucrose lated using the formula: (arbor length 2 width)/(length
in PBS. Forty-micrometer-thick frozen sections were cut 1 width). Therefore, the closer the circularity value was
and stored in cryoprotectant at 220°C until the histolog- to 0, the rounder the arbor. Furthermore, a morphologi-
ical reactions. cal index (MI) was determined using the formula: soma
Microglia were visualized by incubating free floating area/arborization area. The larger the value, the greater
sections in rabbit anti-IBA1 antibody (1:1,000; Wako the soma size was in relation to the arbor size.
Pure Chemical Industries) overnight followed by stand-
ard DAB immunocytochemistry using an anti-rabbit
peroxidase kit and a DAB peroxidase substrate kit TUNEL and Fluoro-Jade B Histochemistry
(Vector Laboratories). The reaction product was intensi-
fied using nickel sulfate to optimize the visualization of Alternate sections from the light immunocytochemis-
very fine processes. Sections from all mice of each strain try portion of this experiment were used to examine
were reacted together in sectioned dishes to ensure uni- neuronal necrosis and apoptosis. Sections containing
form experimental parameters and the analyses were both A1 and V1 from every mouse in the study were
conducted blind. Four sections were analyzed for each of mounted on a slide containing at least one section from
A1 or V1 regions in each mouse. Gray scale photomicro- each age and strain (minimum of six sections/slide) to
graphs were captured using a Spot Insight Color digital provide an appropriate internal control. Each slide
camera (Diagnostic Instruments). included as a positive control a brain section from a
mouse that received an intrahippocampal lipopolysac-
charide injection (LPS; 100 lg) 2 days before perfusion.
Microglia Cell Counts and Nearest Apoptotic neurons were visualized using a TUNEL kit
Neighbor Analysis (Millipore, Billerica, MA) following the included standar-
dized protocol. Briefly, the sections were post fixed for
Images were captured using a 103 objective lens 5 min with a mixture of 95% ethanol and 5% glacial
(Olympus UPlanFL, 0.3 NA) and processed with ImageJ acetic acid, quenched for 5 min with 3% hydrogen perox-
to determine cellular densities (number of cells/mm2) ide, incubated for 1 h in terminal deoxynucleotidyl
and provide cellular coordinates for the analysis of near- transferase enzyme, and incubated in anti-digoxigenin
est neighbor using custom algorithms in Matlab (Math- peroxidase in a humidified chamber for 30 min. Sections
works). Across all cortical layers of A1 or V1 (Franklin were then reacted using a standard DAB peroxidase
and Paxinos, 2008), the center of each microglia was substrate kit (Vector Laboratories) and counterstained
marked with a dot using the ImageJ automark function, with methyl green. The positive control had numerous
which automatically recorded cell numbers as well as purple apoptotic cells against a green background, but
spatial coordinates. Location coordinates were entered no experimental mice showed TUNEL-positive neurons.
into Matlab where the distance between each microglia No further analysis was performed. To test for necrosis,

GLIA
EFFECT OF AGING AND SENSORY LOSS ON GLIA 5
the sections were stained with Fluoro-Jade B (Millipore),
which labels apoptotic and necrotic neurons, using the
included standardized protocol. Briefly, sections were
incubated in 0.06% potassium permanganate for 15 min
and incubated in the dark with 0.001% Fluoro-Jade B
for 30 min. Numerous necrotic neurons from the LPS-
injected control section appeared as green fluorescent
cells around the injury site. No necrotic neurons were
found in experimental subjects.

Statistical Analysis

EM and LM analyses were performed with Prism 5


software (GraphPad). For all statistical tests, sample
size (n) represents individual animals. Significance was
set to P < 0.05 and determined using two-way ANOVA
with a Bonferroni post hoc test to compare across ages
and regions. ASR and ABR data were analyzed using an
ANOVA provided in the SPSS software (v18, IBM, Som-
ers, NY). For ASR analysis, the significance of the
observed differences in mean PPI between groups and
conditions was assessed by a mixed-design ANOVA in
which Lead Time (LT) of the prepulse was a within-
subject variable while Age (A) and Strain (S) were
between-subject variables. The degrees of freedom for
the repeated measures ANOVA were adjusted for nonho-
mogeneity of between-cell correlations by the Hunyh-
Feldt procedure.

RESULTS
Assessment of Auditory and Visual Function
in C57 and CBA Mice

We first assayed visual and auditory perception in the


two strains of mice at different ages from early adulthood
into senescence (Fig. 1; also see Supp. Info. for more
details). Behavioral testing used the phenomenon of
‘‘reflex modification’’ to assess age-related changes in sen-
sitivity to auditory or to visual stimuli, by measuring Fig. 1. Changes in sensitivity to acoustic and photic stimulation
with increasing age in C57 and CBA mice. (A) Mean (SEM) strength in
changes in the extent by which these stimuli inhibited the reflex inhibition (PPI) provided by a light flash summed over 70, 110,
expression of an acoustic startle reflex that was elicited at and 160 ms before the startle noise burst. The difference in photic sen-
brief intervals after their presentation (Ison, 2001). While sitivity between the two strains was minimal in young mice, but the
two groups separated as performance steadily decreased in CBA mice
this acoustic behavioral test has been used often for between 4 and 19 months of age. (B) Mean strength in reflex inhibition
assessing auditory processing in mice, the light flash has (PPI) provided by an acoustic stimulus, a brief gap in a background
been previously used in rats to demonstrate the develop- noise, summed over 60, 110, and 160 ms before the startle noise burst.
The greater sensitivity of the CBA mice to this acoustic stimulus was
mental course of loss of vision in young adult RCS rats present even in the youngest mice, then the difference between the
homozygous for the rd gene (Wecker and Ison, 1986), and groups increased with advancing age as performance decreased
between 4 and 10 months of age in C57 mice. (C) Age-related changes
the slower processing of photic stimuli in aging rats (Ison in hearing thresholds provided by the ABR technique. The test frequen-
et al., 1998). cies are grouped as low frequency, (Lo F, Mean SEM of 3 and 6 kHz)
These behavioral assays revealed a minimal difference and high frequency (Hi F, Mean (SEM) of 24, 32, and 48 kHz). There
was no significant difference between the strains at 2 months of age,
in photic sensitivity between the two strains during but differences emerged within the first year of life, as thresholds for
young adulthood, in agreement with previous observa- the C57 mice rapidly increased, especially at Hi F. By the end of the
tions (Caruso et al., 2010; Danciger et al., 2007; Wil- second year, the thresholds for the C57 mice were beyond the intensity
limits of our instruments (>90 dB SPL), whereas the thresholds for the
liams and Jacobs, 2007), but the two groups separated CBA mice were minimally affected by just 10–15 dB.
as performance increased with advancing age in the C57
mice and steadily decreased in the CBA mice between 4 ulus that was present even in the youngest mice, and
and 19 months of age (Fig. 1A). In contrast, the CBA the difference between the groups increased with
mice displayed a greater sensitivity to the acoustic stim- advancing age as performance was maintained in the

GLIA
6 TREMBLAY ET AL.

TABLE 1. EM Quantification and Statistics


Mean 6 SEM Two-way ANOVAs
3 months 20 months Region Age
Cell type Measure Strain A1 V1 A1 V1 3 months 20 months A1 V1
Neurons Number C57 30 61 32 64 29 6 2 23 6 3 NS NS NS NS
CBA 34 68 28 65 23 6 4 24 6 1 NS NS NS NS
Inclusions C57 0.65 6 0.1 0.67 6 0.2 1.7 6 0.3 1.8 6 0.3 NS NS <0.05 <0.05
CBA 0.71 6 0.09 0.57 6 0.06 1.3 6 0.2 1.4 6 0.1 NS NS <0.01 <0.01
Dark neurons Number C57 1.7 61 0.33 6 0.3 6.7 6 1 1.3 6 0.9 NS <0.05 <0.05 NS
CBA 0.33 6 0.3 1.7 61 0.33 6 0.3 1.7 6 1 NS NS NS NS
Inclusions C57 0.25 6 0.3 0.33 6 0.3 2.8 6 0.1 1.1 6 0.9 NS NS <0.05 NS
CBA 0 0.67 6 0.3 0 1.3 6 0.7 NS NS NS NS
Microglia Number C57 1.0 6 0 3.3 6 0.9 2.3 6 0.7 2.3 6 0.7 NS <0.05 NS <0.001
CBA 1.0 6 0.6 1.3 6 0.3 1.0 6 0 2.3 6 0.3 NS <0.05 NS <0.05
Inclusions C57 0.33 6 0.3 0 4.2 6 0.9 0.22 6 0.2 NS <0.05 <0.001 NS
CBA 0.78 6 0.4 0.67 6 0.7 0.33 6 0.3 3.7 6 0.9 NS <0.05 NS <0.05
Astrocytes Number C57 1.3 6 0.3 2.0 6 0.6 2.7 6 0.9 2.0 6 1 NS NS NS NS
CBA 1.7 6 0.3 2.0 6 0.6 3.3 6 0.3 2.3 6 0.3 NS NS <0.05 NS
Inclusions C57 0 0 2.7 6 2 0.42 6 0.4 NS NS NS NS
CBA 0 0 0 0.33 6 0.3 NS NS NS NS
Oligodendrocytes Number C57 1.0 6 0 0.33 6 0.3 3.0 6 2 0.33 6 0.3 NS NS NS NS
CBA 1.7 6 0.9 0 1.7 6 0.3 3.7 6 0.9 NS NS NS <0.01
Inclusions C57 0 0.33 6 0.3 0 0 NS NS NS NS
CBA 0.11 6 0.1 0 0.17 6 0.2 0.98 6 0.2 NS <0.05 NS <0.01

CBA mice but decreased between 4 and 10 months of oxidative stress or an early stage of neuronal degenera-
age in the C57 mice (Fig. 1B). Additionally, auditory tion (Al-Abdulla and Martin, 1998), were also observed
function as assayed using ABRs showed no significant (Fig. 2B). Additionally, cells with a condensed, electron-
difference between the strains at 2 months of age, but dense cytoplasm and nucleoplasm displaying multiple
differences emerged within the first year of life, as features of degenerating neurons described in neurodege-
thresholds for the C57 mice rapidly increased, especially nerative disease (Oster-Granite et al., 1996; Turmaine
at high frequency, but were stable for the CBA mice. By et al., 2000; Yang et al., 2008) were sometimes encoun-
the end of the second year, the thresholds for the C57 tered (see Materials and Methods section; Fig. 2C,D),
mice were beyond the intensity limits of our instruments especially in A1 of the aged and very deaf C57 mice
(>90 dB SPL) while the thresholds for the CBA mouse (Fig. 2F,G; Table 1). Consistent with previous observa-
were minimally affected by just 10–15 dB (Fig. 1C). tions, these dark neurons were not undergoing apoptosis
Together, these results reveal that the C57 mice display nor necrosis, as shown by the absence of TUNEL and
reduced auditory function but relatively well-maintained Fluoro-Jade B staining on sections from both young (3
visual function during normal aging, whereas the CBA and 12 months) and aged (24 month) sensory cortices of
mice undergo a converse age-related deficit in vision but either strain (Supp. Info. Fig. 1). Together, these observa-
well-maintained audition. tions suggest a particular form of neuronal stress or
degeneration that could be specific to the age-related loss
of sensory function.
Cellular and Subcellular Changes in Neurons

To determine how these changes in sensory perception Cellular and Subcellular Changes in Microglia
influence neuronal function in corresponding sensory
cortices, we examined layers II/III of V1 and A1, where IBA1-labeled microglia exhibited distinct changes in
changes in neuronal and glial function have been previ- density, distribution, and morphology during aging,
ously described in younger animals during sensory depending on the strain and cortical area. In C57 mice,
deprivation (Hensch, 2005; Tremblay et al., 2010a) microglia became more numerous throughout cortical
Quantitative analysis in the same C57 and CBA mice at layers I to VI in both V1 and A1 at 24 months of age
3 and 20 months of age revealed a slight but not statisti- (Fig. 3G; Table 2). This increase in microglial density
cally significant decrease in neuronal density during was more pronounced in A1 than in V1 suggesting that
aging in both regions and strains (Table 1) in agreement loss of auditory input could exacerbate microglial prolif-
with previous stereological observations in aged neocortex eration during aging (Peters et al., 1991a; Vaughan and
and hippocampus of several species including humans Peters, 1974). We also found an age-related decrease in
(reviewed in Morrison and Hof, 1997). Nevertheless, neu- the distance between neighboring microglia in both A1
ronal cell bodies often presented signs of senescence in and V1 (reduced spacing index; Fig. 3A–F,H; Table 2),
the form of lysosomal inclusions (Fig. 2A,B; also see Riga which could not be accounted for by their increased den-
et al., 2006) which became more prevalent during aging, sity, thus suggesting that microglia are less evenly dis-
in V1 (Fig. 2E; Table 1) and A1 (Fig. 2E; Table 1) in both tributed in aged cortices irrespective of sensory input. In
strains. Accumulations of ultrastructurally intact mito- contrast with previous observations in young adult mice
chondria within neuronal cell bodies, which could indicate (Lawson et al., 1990), microglial cell bodies in aged mice

GLIA
EFFECT OF AGING AND SENSORY LOSS ON GLIA 7

Fig. 2. Ultrastructural changes in neurons during normal aging gesting an ongoing degenerative process. D, dark neuron; M, microglia;
and age-related sensory impairment in C57 and CBA mice. (A–D) EM N, neuron; O, oligodendrocyte. Scale bars 5 1 lm. (E–G) Number of
images showing aged neurons that contain multiple lysosomal inclu- lysosomal inclusions per neuron (E), number of dark neurons per
sions (asterisks) and/or mitochondria in their perikaryal cytoplasm 10,000 lm2 of ultrathin section (F), and number of lysosomal inclusions
(m; B). In (C) and (D), the neurons display condensed, electron-dense per dark neuron (G; mean 6 SEM). Grey lines and asterisks refer to
cytoplasm and nucleoplasm, more frequent ruffling of the plasma mem- statistical comparisons between A1 regions, black lines and asterisks
brane, extensive invagination of the nuclear membrane (arrow; C), and to comparisons between V1 regions, and black dotted line and asterisk
dilation of the endoplasmic reticulum (arrowheads; D), possibly sug- to comparison between A1 and V1 regions. *P < 0.05 and **P < 0.01.

GLIA
8 TREMBLAY ET AL.

Fig. 3. Changes in density and distribution of microglia in C57 and without change in density in A1 (I–K) and V1 (L–N) of the CBA. Scale
CBA mice. (A–L) Low-magnification photomicrographs showing an bars 5 100 lm. Quantification of microglial density in C57 (G) and CBA
increase in density and clumping in C57 A1 (A–F); (I–N) low-magnifica- (O), and spacing index in C57 (H) and CBA (P) mice. Statistics refer
tion photomicrographs showing an increase in density and clumping in to post hoc analyses based on a two-way ANOVA. **P < 0.01 and
A1 (A–C) and V1 (D–F) of the C57 in contrast to an increase in clumping ***P < 0.001. Statistical comparisons are illustrated as in Fig. 2.

often localized close together and sometimes in direct gressive age-related decrease in microglial nearest
contiguity (see Fig. 3C,F for examples) which could be neighbor distance was also observed at 12 and 24
suggestive of ongoing proliferation. In CBA mice, a pro- months of age to similar degrees in A1 and V1 (reduced

GLIA
EFFECT OF AGING AND SENSORY LOSS ON GLIA 9
spacing index) whereas microglial density remained

6 0.0015
6 0.0032
6 0.0017
6 0.214

6 0.003

6 0.010
6 0.008
6 27.99
6 33.23
6 15.52
6 0.24

6 8.67
6 786
6 807
relatively constant (Fig. 3I–P; Table 2), indicating a loss

6 8.5
6 6.6
of territorial organization that could be regulated
V1
independently from microglial density during aging or

8871
17,468

0.036
0.064
0.015
0.023

0.257
5.1
5.2
216
146

0.19

571
520
194
166
age-related sensory loss.
24 months

Microglia showed remarkable morphological transfor-


mation over the course of aging in cortical layers II/III

0.049 6 0.0034
0.023 6 0.0029
0.017 6 0.0012
5.3 6 0.352
(Figs. 4 and 5). In C57 mice, at 24 months of age, micro-

0.052 6 0.002

0.248 6 0.006
569 6 31.31
541 6 19.26
248 6 32.42
165 6 10.04
0.25 6 0.011
5.2 6 0.15

12,243 6 715
12,040 6 878 glial process arborizations were significantly reduced in
249 6 7.2
145 6 8.6

area, and displayed elongated geometries as evidenced


A1

by an increased circularity index (Fig. 4A–H; Table 2). A


morphological index was computed to assess the soma
area relative to the process area (soma area/arborization
area), two parameters susceptible to change during
0.038 6 0.0014
0.01 6 0.0029
0.015 6 0.0017

microglial activation (Streit et al., 1999). Aged microglia


6.3 6 0.169
27,971 6 1,919

0.023 6 0.001

0.191 6 0.009
0.223 6 0.006
605 6 25.69
533 6 21.55
142 6 14.30
6.7 6 0.07

140 6 8.91
15,289 6 397
174 6 9.0
129 6 6.4

displayed significantly increased morphological indices


V1

that were most pronounced in A1 (Fig. 4I; Table 2), sug-


gesting an association with hearing loss. As microglial
Mean 6 SEM

soma area displayed little variation during aging or age-


12 months

related sensory deficit (Fig. 4J; Table 2), these changes


in morphological index most likely resulted from the
0.021 6 0.0017
0.031 6 0.0033
0.006 6 0.0008
0.011 6 0.0023
6.3 6 0.097
30,166 6 2,536
18,043 6 1,601

0.193 6 0.013
0.202 6 0.020
581 6 22.46
502 6 21.56

reduction in the area of microglial arborizations. Never-


179 6 14.0

6.4 6 0.17

113 6 7.95
122 6 8.90
135 6 8.4

theless, microglial somas in the oldest animals were


A1
TABLE 2. LM Quantification and Statistics

more variable in size: comparisons of their standard


deviation showed a significant increase at 24 months of
age especially in A1 (Fig. 4K; Table 2). This effect in
turn translated into significant differences in the stand-
ard deviation of the morphological index with age
0.02 6 0.0008
0.024 6 0.0012
0.006 6 0.0006
0.007 6 0.0005
6.9 6 0.163
28,915 6 1,491
22,613 6 1,202

0.154 6 0.004
0.179 6 0.007
542 6 15.35
519 6 10.97

(Fig. 4L; Table 2). In the CBA mice, microglia also


6.3 6 0.15

121 6 5.58
116 6 7.34
169 6 7.0
142 6 4.8

underwent striking morphological changes as early as


V1

12 months (Fig. 5; Table 2), with a more complex profile.


The span of microglial process arborizations decreased
3 months

with age and became more elongated (Fig. 5A–H; Table


2) as in C57 animals, and there was a trend toward
smaller microglial arborizations at all ages in V1. Mor-
0.019 6 0.0009
0.021 6 0.0009
0.005 6 0.0004
0.006 6 0.0006
6.7 6 0.086
7.2 6 0.088
27,993 6 1,324
25,333 6 1,121

0.166 6 0.006

507 6 20.85
0.17 6 0.011

phological indices were also significantly increased dur-


509 6 9.08

111 6 5.35
106 6 7.86
177 6 7.2
148 6 4.3

ing aging, especially at 24 months of age (Fig. 5I; Table


A1

2), suggesting that microglial morphology could also be


modulated in an age and activity-dependent manner in
this strain. These changes in morphological indices
occurred in the absence of changes in microglial soma
areas except for an increase in standard deviations at 24
<0.0001

<0.0001
<0.0001
<0.0001
<0.0001
<0.0001
<0.0001
<0.0001
<0.0001
<0.0001
<0.0005

<0.0001
<0.0001

months of age as in the C57 mice (Fig. 5J–L; Table 2).


Two-way ANOVAs

Age

NS

NS
NS

Together, these observations in both mouse strains sug-


gest that microglia are differentially sensitive to aging
and age-related sensory loss, with different populations
undergoing diverse structural and functional changes in
P<0.05

P<0.05

P<0.05
<0.01

<0.05
<0.01

<0.01
<0.001
Region

NS
NS
NS

NS

NS
NS
NS

NS

parallel.
Ultrastructural examination also revealed distinctive
features that were most pronounced in layers II/III in
V1 of CBA mice and A1 of C57 mice at 20 months of
Strain

CBA

CBA

CBA

CBA

CBA

CBA

CBA

CBA
C57

C57

C57

C57

C57

C57
C57

C57

age such as hypertrophy and rounding of the cell body


(Figs. 6B,C and 7A,C), enlargement of processes
(Fig. 7E,F) and accumulation of various types of phago-
cytic inclusions (Figs. 6A,C,D and 7). While microglial
Arbor Circularity

Soma Area SD

processes were often found in direct juxtaposition


Spacing Index

with synapse-associated dendritic spines, axon termi-


Arbor Area

Soma Area
Measure

nals, synaptic clefts and perisynaptic astrocytic proc-


Density

MI SD

esses throughout adulthood and aging (see Supp. Info.


MI

Fig. 2), as previously shown in adolescent and young

GLIA
10 TREMBLAY ET AL.

Fig. 4. Changes in the morphology of microglia in the C57 mouse. area of the process arbor (G), arbor circularity index (H), morphological
(A–F) High-magnification photomicrographs illustrating the decline in size index (I), average soma area (J), soma area standard deviation (SD; K),
and complexity of processes arborization without obvious changes in soma and morphological index standard deviation (L). Statistics refer to post hoc
size in 24 months C57 mice: A1 (A–C) and V1 (D–F). Scale bars 5 25 lm. analyses based on a two-way ANOVA. *P < 0.05, **P < 0.01, and
Arrowheads show examples of clumped microglia. Quantification of the ***P < 0.001. Statistical comparisons are illustrated as in Fig. 2.

GLIA
EFFECT OF AGING AND SENSORY LOSS ON GLIA 11

Fig. 5. Changes in the morphology of microglia in the CBA mouse. circularity index (H), morphological index (I), average soma area (J),
(A-F) High-magnification photomicrographs illustrating the decline in soma area standard deviation (SD; K), and morphological index stand-
size and complexity of processes arborization without obvious changes ard deviation (L). Statistics refer to post hoc analyses based on a two-
in soma size by 12 months in CBA mice: A1 (A–C) and V1 (D–F). Scale way ANOVA. *P < 0.05; **P < 0.01 and ***P < 0.001. Statistical com-
bars 5 25 lm. Quantification of the area of the process arbor (G), arbor parisons are illustrated as in Fig. 2.

adult mice (Tremblay et al., 2010a; Wake et al., 2009), terminals (Fig. 7A–F) or dendritic spines (Fig. 7E) some-
microglial cell bodies and large processes often contained times separated by wide electron-lucent space (Fig. 7E).
cellular inclusions that resembled profiles of axon Microglial processes extending between axon terminals

GLIA
12 TREMBLAY ET AL.

Fig. 6. Ultrastructural changes in microglia in C57 and CBA mice. bodies (see A and D, for example, of normal size, B and C, for example, of
(A–D) Aged microglia accumulating various types of noncellular inclu- enlarged size). Immunoperoxidase staining for the microglia-specific
sions, including lysosomal inclusions (asterisks), vacuoles and large marker IBA1 is shown by arrowheads. M, microglia. Scale bars 5 1 lm.
vesicles (arrow; A), and lipid droplets of various sizes (arrow; D), which (E) Number of inclusions per microglia (lysosomal inclusions, vacuoles,
could result from the phagocytic elimination of neurons or glial cells. The small to large vesicles, lipid droplets, and cellular elements such as den-
microglia in C appears almost completely filled by cellular debris, akin to dritic spines and axon terminals (see Fig. 7); mean 6 SEM). Statistical
fat granule cells or gitter cells. Also note the size diversity of microglial cell comparisons are illustrated as in Fig. 2. *P < 0.05 and ***P < 0.001.

and dendrites (Fig. 7F) in a way reminiscent of synaptic features of axon terminals and dendritic spines)
stripping during acute and inflammatory lesions (Trapp revealed an increased prevalence during aging, in V1
et al., 2007) were also observed. Furthermore, quantita- of the CBA mice and A1 of the C57 mice (Fig. 6E;
tive analysis of the total number of inclusions (lysosomal Table 1), with nearly all microglia containing inclu-
lipopigments, large vesicles, vacuoles, lipid droplets, sions. Some microglia appeared almost completely filled
and cellular elements often displaying ultrastructural with cellular debris, akin to fat granule cells or gitter

GLIA
EFFECT OF AGING AND SENSORY LOSS ON GLIA 13

Fig. 7. Changes in microglial engulfment of synaptic elements in engulfment of synaptic elements by microglia. In F, the microglia
C57 and CBA mice. (A–F) Young and aged microglia displaying cellular extends a fine process (arrows) between an axon terminal that appears
inclusions (in) in their cell body (C, inset in D), proximal process (A, completely ensheathed (‘‘t’’) and a dendrite (d), in a way reminiscent of
inset in B), or distal processes (E and F). Cellular inclusions sometimes synaptic stripping. In all pictures, microglial profiles show an electron-
resembled profiles of axon terminals (‘‘t’’; see B and D–F for examples) dense immunoperoxidase staining for IBA1. M, microglia; s, dendritic
and dendritic spines (‘‘s’’; E for example) with contained pockets of spine; t, axon terminal; v, vacuole. Scale bars 5 1 lm for A–D, 0.5 lm
electron-lucent space (arrows; D and E), suggesting the phagocytic for E, and 0.25 lm for F.

cells (nine of 45 microglia; see Fig. 6C for example; could become accelerated during age-related loss of
Vaughan and Peters, 1974), indicating that microglial sight or hearing.
phagocytosis of cellular elements including synapses

GLIA
14 TREMBLAY ET AL.

Fig. 8. Ultrastructural changes in astrocytes, oligodendrocytes, and ets in their perikaryal cytoplasm (arrowheads). (E and F) Myelinated
axonal myelination in C57 and CBA mice. (A and B) EM examples of axons (MA) with redundant (arrows; E and F) or ballooned (see the
aged astrocytes (A) with enlarged processes typically surrounding a large electron-lucent space in the sheath of the lower right MA; F)
blood vessel (BV; A) or containing large accumulations of lysosomal myelin sheaths. m, mitochondria. Scale bars 5 1 lm. (G) Number of
inclusions (B). (C and D) Aged oligodendrocytes (O) devoid of lysosomal oligodendrocytes per 10,000 lm2 of ultrathin section (mean 6 SEM).
inclusions, encountered in a pair (C) or containing electron-lucent pock- Statistical comparisons are illustrated as in Fig. 2. **P < 0.01.

Changes in Other Glial Components: Astrocytes, hypertrophic (Wilhelmsson et al., 2006), the most appa-
Oligodendrocytes, and Axonal Myelination rent change was the occasional accumulation of large
lysosomal inclusions associated with vacuoles (see
In contrast with microglia, astrocytes showed little Fig. 8B for example in an astrocytic process) in A1 of
change in number or morphology during aging or age- the very deaf C57 mice (Table 1). In contrast, oligoden-
related sensory loss (Fig. 8A,B; Table 1), in layers II/III drocytes rarely contained lysosomal inclusions
of V1 and A1, in agreement with previous observations (Fig. 8C,D) but increased in number and were more of-
in aged rat cerebral cortex (Vaughan and Peters, 1974). ten encountered in contiguous pairs during aging, par-
While some astrocytic processes ensheathing synapses ticularly in V1 of the CBA mice (Table 1; Fig. 8G), as
and blood vessels appeared enlarged during aging previously reported in aged rhesus monkeys (Peters and
(Fig. 8A), similar to reactive astrocytes which become Sethares, 2004), suggesting an ongoing proliferation

GLIA
EFFECT OF AGING AND SENSORY LOSS ON GLIA 15
that could be exacerbated by age-related sensory loss in neuronal numbers during aging in A1 and V1 of both
both strains. Finally, our ultrastructural analysis also mouse strains that were nevertheless accompanied by
revealed alterations of myelinated axons during aging, various signs of neuronal stress. These included the prev-
also in V1 of the CBA mice and A1 of the C57 mice espe- alence of lysosomal inclusions, which were previously
cially. Some myelin sheaths appeared ballooned (Fig. 8F) associated with protease instability, lysosome and cellular
as during natural and experimental demyelination recycling dysfunction, cytoskeletal damage, amyloid
(Peters and Sethares, 2003), while other axons displayed deposits and amyloid-related neuropathology (Brunk and
redundant myelin sheaths (Fig. 8E,F) suggesting oligo- Terman, 2002; Riga et al., 2006), and the accumulation of
dendrocytic attempts at remyelination in the context of morphologically intact mitochondria within neuronal cell
cortical senescence (Peters and Sethares, 2004). bodies (Al-Abdulla and Martin, 1998). Together, our
observations thus suggest that aged neurons may have
altered metabolic function without undergoing large scale
DISCUSSION degeneration.
The most profound age-related change that we
Different mouse strains experience age-related hear- observed concerned microglia, the resident immune cells
ing and/or sight loss to different degrees. While C57 and of the brain, which are the key mediators of neuroin-
CBA mice are known to differentially develop hearing flammatory processes within the central nervous system
deficits during normal aging, here we also report recip- in various contexts including aging (Lee et al., 2000;
rocal differences in their visual sensitivity with a behav- Prinz et al., 2011; Streit, 2002; von Bernhardi et al.,
ioral assay that depends on the inhibition of the acoustic 2010). In particular, we found that microglia become
startle reflex by innocuous and even near threshold more numerous and tend to distribute less evenly, with
prestimulation (Hoffman and Ison, 1992). Whether and an increased prevalence of direct contiguities, suggest-
how these changes in sensory perception impact neuro- ing an increased proliferation of microglia and/or a
nal and glial function in corresponding sensory cortices breakdown of the mechanisms which maintain microglial
is the focus of this discussion. It is important to note territories and regulate immune surveillance (Nimmer-
that while functional changes in sensory perception jahn et al., 2005). Aged microglia also exhibited morpho-
were detected within the first year of age, the cellular logical changes that were similar to those previously
changes in A1 of the C57 and V1 of the CBA mice were described during pathological activation (Streit et al.,
first observed at 20 and 12 months of age, respectively, 1999): a general simplification of the microglial arbor
suggesting that loss of peripheral function precedes and with a decrease in the span of microglial processes com-
likely drives central changes. Nevertheless, we cannot pared to the cell body. Our findings are supported by
rule out that microglial changes in the cortex or lower recent live confocal imaging observations showing that
structures also contribute to loss of sensory function. aged microglia in retinal explants have increased density,
decreased arbor area, together with decreased total num-
ber of branch points and total process length (Damani et
Effects of Aging on Cortical Cells al., 2011). Such morphological changes may reflect age-
induced microglial activation as suggested by the
Aging is a risk factor for most neurological disorders increased expression of molecular markers of activation
(Merrill and Small, 2011), and various cellular and sub- in the aged brain (Ogura et al., 1994; Sheffield and Ber-
cellular changes that occur within the aging brain can al- man, 1998; Sierra et al., 2007), although dystrophic
ter function in the absence of pathology. Possible sources changes differing morphologically from microglial activa-
of these changes include accumulated DNA damage (Gau- tion were also reported during aging (Streit et al., 2004).
batz, 1995; Vijg and Campisi, 2008), oxidative stress Future quantitative morphological analyses, such as
(Droge and Schipper, 2007), deregulated calcium homeo- those described here, could contribute to defining the
stasis, and signaling (Disterhoft et al., 1994; O’Neill et al., exact morphological parameters that characterize differ-
1997; Zettel et al., 1997), as well as altered neuroimmune ent modes of microglial function, including activation and
function (von Bernhardi et al., 2010). The idea that brain dystrophy, and further refine our understanding of the
dysfunction during aging is mediated by a large-scale loss nature of microglial transformation during aging (Sierra
of central neurons has now been largely discredited et al., 2007). While microglial cell body size was not signif-
although a more subtle region-specific loss may occur icantly altered, we observed greater variation in the size
(West, 1993; Willott et al., 1994; Woodruff-Pak et al., of microglial somas in older animals, suggesting that pop-
2010). Similarly, biophysical properties of neurons appear ulations of aged microglia may become more heterogene-
to be largely conserved across the lifespan (Barnes and ous and that distinct types of morphological and behav-
McNaughton, 1980; Burke and Barnes, 2006) aside from ioral changes may occur in parallel. This is all the more
an increase in calcium conductance that could contribute intriguing in the context of the recent interest in defining
to altered plasticity mechanisms (Barnes et al., 2000; subpopulations of microglia (Carson et al., 2007; Lynch,
Landfield et al., 1978) and increased susceptibility to 2009) implementing different molecular signaling cas-
dendritic remodeling and synaptic elimination (Barnes cades on different timescales, with pro- and anti-inflam-
et al., 1992; Geinisman et al., 1992). In this context, our matory functions that can exacerbate or ameliorate neu-
ultrastructural analysis showed relatively unchanged ronal injury (Kigerl et al., 2009). While the expression of

GLIA
16 TREMBLAY ET AL.

both pro- and anti-inflammatory cytokines increases with of aging (Riga et al., 2006), aged microglia accumulated
aging (Sierra et al., 2007), it is currently unknown exceptionally large amounts of lysosomal inclusions,
whether such heterogeneous microglial populations exist including profiles resembling synaptic elements, in
in the aged brain and how they could impact age-related cortical areas deprived of sensory input. This striking
functional decline. In future experiments, it will be impor- behavior suggests an accumulated phagocytic activity,
tant to tie morphological alterations with an assessment supporting previous observations that microglia enlarge
of microglial expression of cytokines, chemokines, and and become filled with cellular debris during normal
other functional markers in situ, to provide further aging (Peters, 2002), a phenomenon which could eventu-
insights into the beneficial and/or detrimental effects of ally contribute to an impairment of their phagocytic clear-
microglia on age-related functional decline. ance (Neumann et al., 2009). Complete loss of vision dur-
ing adolescence also resulted in increased microglial
phagocytosis of synaptic elements, and in an overall
Effect of Sensory Deprivation on Aged decrease in microglial motility in visual cortex (Tremblay
Cortical Cells et al., 2010a), and similarly, a reduced velocity of individ-
ual microglial processes was recently observed with live
Activity-dependent plasticity in sensory subcortical confocal imaging in retinal explants from aged mice
and cortical areas has been extensively studied in young (Damani et al., 2011). Microglia were also shown to con-
animals (Rittenhouse and Majewska, 2009), but the tribute to synaptic pruning during early postnatal devel-
impact of age-related sensory loss on the aging process opment (Paolicelli et al., 2011). Together, these findings
is still not clear. The secretion of neuroprotective agents support a model in which quiescent microglia contribute
such as growth factors is activity dependent (Caleo and to restructuring neuronal circuits by eliminating synap-
Maffei, 2002), and changes in neuronal activity induce ses in an experience-dependent manner throughout the
homeostatic changes that could affect the aging process lifespan. Future experiments involving two-photon in
(Desai et al., 1999; Turrigiano and Nelson, 2004). In vivo imaging of microglia could further explore their novel
fact, many of the molecular changes observed in aged physiological role in aging.
neuronal networks can be replicated by silencing cortical Finally, our analysis also revealed ultrastructural evi-
neurons (Gleichmann et al., 2010) suggesting that sen- dence of demyelination and remyelination in deprived
sory deprivation could affect neuronal function and exac- sensory cortices of the C57 and CBA mice, suggesting
erbate age-related changes in a similar manner. that axonal dysfunction and/or myelin remodeling could
A surprising finding in our study was the increased be exacerbated by the loss of sensory function during
prevalence of ‘‘dark’’ neurons in deprived A1 of the C57 normal aging. We also found increased numbers of oligo-
mice undergoing age-related loss of hearing. These appa- dendrocytes that were often contiguous one with
rently degenerating cells were neither necrotic nor apo- another, possibly suggesting their proliferation, as previ-
ptotic and contained large numbers of lysosomal inclu- ously reported in aged rhesus monkeys (Peters and
sions suggesting metabolic dysfunction (Riga et al., 2006). Sethares, 2004). In line with these observations, myelin
Similar dark neurons have been described in Alzheimer’s remodeling was recently shown to be regulated by neu-
and Huntington’s diseases (Oster-Granite et al., 1996; ronal activity and sensory-experience dependent plastic-
Turmaine et al., 2000; Yang et al., 2008) and after chronic ity (Fields, 2010; McGee et al., 2005; Wake et al., 2011).
inhibition of long-term potentiation/cAMP-response ele- As newborn oligodendrocytes are derived in the mature
ment-binding protein (CREB) function (Valor et al., 2010). brain from oligodendrocytic progenitors sharing few ul-
Interestingly, CREB is downregulated in various neurode- trastructural features with oligodendrocytes (Peters and
generative diseases including Alzheimer and Huntington, Sethares, 2004) but specifically expressing the NG2
as well as during normal aging (Bach et al., 1999; Porte et chondroitin sulfate proteoglycan (Trotter et al., 2010), it
al., 2008; Shimohata et al., 2000; Tomobe et al., 2007; will be important in subsequent experiments to examine
Vitolo et al., 2002). As this cellular ‘‘darkening’’ can be re- these oligodendrocytic progenitors and their differential
versible in certain conditions (Auer et al., 1985; Ingvar et changes in terms of morphology and behavior during
al., 1988), it presently remains unclear whether the aging and age-related sensory loss.
‘‘dark’’ neurons that we observed were undergoing cellu-
lar stress or degeneration in the aging brain. ACKNOWLEDGMENTS
Our results also showed consistent effects of age-
related sensory loss on microglia in both C57 and CBA The authors are grateful to John A. Olschowka and
mice. Recent findings have shown that neuroimmune his laboratory for providing LPS-injected brain samples,
molecular pathways contribute to synaptic plasticity Robert D. Frisina for mice, and Kathy Barsz for statisti-
(Boulanger and Shatz, 2004), and changes in microglial cal advice.
morphology and behavior have been described during
visual deprivation in adolescent and adult mice (Trem- REFERENCES
blay et al., 2010a; Wake et al., 2009) suggesting they are
a general consequence of sensory loss. While lysosomal Al-Abdulla NA, Martin LJ. 1998. Apoptosis of retrogradely degenerat-
ing neurons occurs in association with the accumulation of perikaryal
inclusions were observed in all types of cells except oli- mitochondria and oxidative damage to the nucleus. Am J Pathol
godendrocytes, and appear to be a normal consequence 153:447–456.

GLIA
EFFECT OF AGING AND SENSORY LOSS ON GLIA 17
Anderson JM, Hubbard BM, Coghill GR, Slidders W. 1983. The effect of tems in the rat dentate gyrus as revealed by the unbiased stereologi-
advanced old age on the neurone content of the cerebral cortex. cal dissector technique. Hippocampus 2:437–444.
Observations with an automatic image analyser point counting Gleichmann M, Zhang Y, Wood WH III, Becker KG, Mughal MR, Pazin
method. J Neurol Sci 58:235–246. MJ, van Praag H, Kobilo T, Zonderman AB, Troncoso JC, Markesbery
Auer RN, Kalimo H, Olsson Y, Siesjo BK. 1985. The temporal evolution WR, Mattson MP. 2010. Molecular changes in brain aging and Alzhei-
of hypoglycemic brain damage. I. Light- and electron-microscopic mer’s disease are mirrored in experimentally silenced cortical neuron
findings in the rat cerebral cortex. Acta Neuropathol 67:13–24. networks. Neurobiol Aging. 33:205.e1-205.e18.
Bach ME, Barad M, Son H, Zhuo M, Lu YF, Shih R, Mansuy I, Haw- Henry KR, Chole RA. 1980. Genotypic differences in behavioral, physio-
kins RD, Kandel ER. 1999. Age-related defects in spatial memory logical and anatomical expressions of age-related hearing loss in the
are correlated with defects in the late phase of hippocampal long- laboratory mouse. Audiology 19:369–383.
term potentiation in vitro and are attenuated by drugs that Hensch TK. 2005. Critical period plasticity in local cortical circuits. Nat
enhance the cAMP signaling pathway. Proc Natl Acad Sci USA Rev Neurosci 6:877–888.
96:5280–5285. Hof PR, Morrison JH. 2004. The aging brain: Morphomolecular senes-
Barnes CA, McNaughton BL. 1980. Physiological compensation for loss cence of cortical circuits. Trends Neurosci 27:607–613.
of afferent synapses in rat hippocampal granule cells during senes- Hoffman HS, Ison JR. 1992. Reflex modification and the anlaysis of sen-
cence. J Physiol 309:473–485. sory processing in developmental and comparative research. In: Camp-
Barnes CA, Rao G, Foster TC, McNaughton BL. 1992. Region-specific bell BA, Hayne H, Richardson R, editors. Attention and information
age effects on AMPA sensitivity: Electrophysiological evidence for processing in infants and adults: Perspectives from human and animal
loss of synaptic contacts in hippocampal field CA1. Hippocampus research. Hillsdale: Lawrence Erlbaum Associates. pp. 83–111.
2:457–468. Hunter KP, Willott JF. 1987. Aging and the auditory brainstem response
Barnes CA, Rao G, Houston FP. 2000. LTP induction threshold change in mice with severe or minimal presbycusis. Hear Res 30:207–218.
in old rats at the perforant path-granule cell synapse. Neurobiol Inamizu T, Chang MP, Makinodan T. 1985. Influence of age on the produc-
Aging 21:613–620. tion and regulation of interleukin-1 in mice. Immunology 55:447–455.
Bilbo SD. 2010. Early-life infection is a vulnerability factor for aging- Ingvar M, Morgan PF, Auer RN. 1988. The nature and timing of excito-
related glial alterations and cognitive decline. Neurobiol Learn Mem toxic neuronal necrosis in the cerebral cortex, hippocampus and thal-
94:57–64. amus due to flurothyl-induced status epilepticus. Acta Neuropathol
Boulanger LM, Shatz CJ. 2004 Immune signalling in neural develop- 75:362–369.
ment, synaptic plasticity and disease. Nat Rev Neurosci 5:521–531. Ison JR. 2001. The acoustic startle response in the mouse: Reflex elici-
Brunk UT, Terman A. 2002. The mitochondrial-lysosomal axis theory of tation and reflex modification by preliminary stimuli. In: Willott JF,
aging: Accumulation of damaged mitochondria as a result of imper- editor. Handbook of mouse auditory research: From behavior to mo-
fect autophagocytosis. Eur J Biochem 269:1996–2002. lecular biology. Boca Raton: CRC Press. pp. 59–82.
Burke SN, Barnes CA. 2006. Neural plasticity in the ageing brain. Nat Ison J, Bowen GP, del Cerro M. 1998. A behavioral study of temporal
Rev Neurosci 7:30–40. processing and visual persistence in young and aged rats. Beh Neuro-
Caleo M, Maffei L. 2002. Neurotrophins and plasticity in the visual sci 112:1273–1279.
cortex. Neuroscientist 8:52–61. Ito D, Imai Y, Ohsawa K, Nakajima K, Fukuuchi Y, Kohsaka S. 1998.
Carson MJ, Bilousova TV, Puntambekar SS, Melchior B, Doose JM, Microglia-specific localisation of a novel calcium binding protein,
Ethell IM. 2007. A rose by any other name? The potential consequen- Iba1. Brain Res Mol Brain Res 57:1–9.
ces of microglial heterogeneity during CNS health and disease. Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popo-
Neurotherapeutics 4:571–579. vich PG. 2009. Identification of two distinct macrophage subsets with
Caruso RC, Aleman TS, Cideciyan AV, Roman AJ, Sumaroka A, Mullins divergent effects causing either neurotoxicity or regeneration in the
CL, Boye SL, Hauswirth WW, Jacobson SG. 2010. Retinal disease in injured mouse spinal cord. J Neurosci 29:13435–13444.
Rpe65-deficient mice: Comparison to human leber congenital amaurosis Landfield PW, McGaugh JL, Lynch G. 1978. Impaired synaptic poten-
due to RPE65 mutations. Invest Ophthalmol Vis Sci 51:5304–5313. tiation processes in the hippocampus of aged, memory-deficient rats.
Clapcote SJ, Lazar NL, Bechard AR, Roder JC. 2005. Effects of the rd1 Brain Res 150:85–101.
mutation and host strain on hippocampal learning in mice. Behav Lawson LJ, Perry VH, Dri P, Gordon S. 1990. Heterogeneity in the dis-
Genet 35:591–601. tribution and morphology of microglia in the normal adult mouse
Congdon N, O’Colmain B, Klaver CC, Klein R, Munoz B, Friedman DS, brain. Neuroscience 39:151–170.
Kempen J, Taylor HR, Mitchell P. 2004. Causes and prevalence of Lee CK, Weindruch R, Prolla TA. 2000. Gene-expression profile of the
visual impairment among adults in the United States. Arch Ophthal- ageing brain in mice. Nat Genet 25:294–297.
mol 122:477–485. Li HS, Borg E. 1991. Age-related loss of auditory sensitivity in two
Cragg BG. 1975. The density of synapses and neurons in normal, men- mouse genotypes. Acta Otolaryngol 111:827–834.
tally defective ageing human brains. Brain 98:81–90. Luo XG, Ding JQ, Chen SD. 2010. Microglia in the aging brain: Rele-
Dalke C, Loster J, Fuchs H, Gailus-Durner V, Soewarto D, Favor J, Neu- vance to neurodegeneration. Mol Neurodegener 5:12.
hauser-Klaus A, Pretsch W, Gekeler F, Shinoda K, Zrenner E, Mei- Lynch MA. 2009. The multifaceted profile of activated microglia. Mol
tinger T, Hrab e de Angelis M, Graw J. 2004. Electroretinography as a Neurobiol 40:139–156.
screening method for mutations causing retinal dysfunction in mice. Lynch MA. 2010. Age-related neuroinflammatory changes negatively
Invest Ophthalmol Vis Sci 45:601–609. impact on neuronal function. Front Aging Neurosci 1:6.
Damani MR, Zhao L, Fontainhas AM, Amaral J, Fariss RN, Wong WT. McGee AW, Yang Y, Fischer QS, Daw NW, Strittmatter SM. 2005. Expe-
2011. Age-related alterations in the dynamic behavior of microglia. rience-driven plasticity of visual cortex limited by myelin and Nogo
Aging Cell 10:263–276. receptor. Science 309:2222–2226.
Danciger M, Yang H, Ralston R, Liu Y, Matthes MT, Peirce J, Lavail Merrill DA, Small GW. 2011. Prevention in psychiatry: Effects of
MM. 2007. Quantitative genetics of age-related retinal degeneration: healthy lifestyle on cognition. Psychiatr Clin North Am 34:249–261.
A second F1 intercross between the A/J and C57BL/6 strains. Mol Vis Morrison JH, Hof PR. 1997. Life and death of neurons in the aging
13:79–85. brain. Science 278:412–419.
Desai NS, Rutherford LC, Turrigiano GG. 1999. BDNF regulates the Neumann H, Kotter MR, Franklin RJ. 2009. Debris clearance by micro-
intrinsic excitability of cortical neurons. Learn Mem 6:284–291. glia: An essential link between degeneration and regeneration. Brain
Disterhoft JF, Gispen WH, Traber J, Khachaturian ZF. 1994. Calcium 132:288–295.
hypothesis of aging and dementia. Ann NY Acad Sci 747. Nimmerjahn A, Kirchhoff F, Helmchen F. 2005. Resting microglial cells
Droge W, Schipper HM. 2007. Oxidative stress and aberrant signaling are highly dynamic surveillants of brain parenchyma in vivo. Science
in aging and cognitive decline. Aging Cell 6:361–370. 308:1314–1318.
Fields RD. 2010. Neuroscience. Change in the brain’s white matter. Norman JP, Perry SW, Reynolds HM, Kiebala M, De Mesy Bentley KL,
Science 330:768–769. Trejo M, Volsky DJ, Maggirwar SB, Dewhurst S, Masliah E, Gelbard
Flanary BE, Sammons NW, Nguyen C, Walker D, Streit WJ. 2007. HA. 2008. HIV-1 Tat activates neuronal ryanodine receptors with
Evidence that aging and amyloid promote microglial cell senescence. rapid induction of the unfolded protein response and mitochondrial
Rejuvenation Res 10:61–74. hyperpolarization. PLoS One 3:e3731.
Franklin KBJ, Paxinos G. 2008. The Mouse brain in stereotaxic coordi- O’Neill WE, Zettel ML, Whittemore KR, Frisina RD. 1997. Calbindin
nates. San Diego, CA: Academic Press. 256 p. D-28k immunoreactivity in the medial nucleus of the trapezoid body
Gaubatz JW. 1995. Genomic instability during aging of postmitotic declines with age in C57BL/6, but not CBA/CaJ, mice. Hear Res
mammalian cells. In: Macieira-Coelho A, editor. Molecular basis of 112:158–166.
aging. Boca Raton: CRC Press. pp. 71–182. Ogura K, Ogawa M, Yoshida M. 1994. Effects of ageing on microglia in
Geinisman Y, deToledo-Morrell L, Morrell F, Persina IS, Rossi M. 1992. the normal rat brain: Immunohistochemical observations. Neurore-
Age-related loss of axospinous synapses formed by two afferent sys- port 5:1224–1226.

GLIA
18 TREMBLAY ET AL.

Oster-Granite ML, McPhie DL, Greenan J, Neve RL. 1996. Age-depend- Trapp BD, Wujek JR, Criste GA, Jalabi W, Yin X, Kidd GJ, Stohlman
ent neuronal and synaptic degeneration in mice transgenic for the C S, Ransohoff R. 2007. Evidence for synaptic stripping by cortical
terminus of the amyloid precursor protein. J Neurosci 16:6732–6741. microglia. Glia 55:360–368.
Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Tremblay ME, Lowery RL, Majewska AK. 2010a. Microglial interac-
Giustetto M, Ferreira TA, Guiducci E, Dumas L, Ragozzino D, Gross tions with synapses are modulated by visual experience. PLoS Biol
CT. 2011. Synaptic pruning by microglia is necessary for normal 8:e1000527.
brain development. Science 333:1456–1458. Tremblay ME, Riad M, Majewska A. 2010b. Preparation of mouse brain
Perry VH, Matyszak MK, Fearn S. 1993. Altered antigen expression of tissue for immunoelectron microscopy. J Vis Exp 41:2021.
microglia in the aged rodent CNS. Glia 7:60–67. Trotter J, Karram K, Nishiyama A. 2010. NG2 cells: Properties, prog-
Peters A. 1996. Age-related changes in oligodendrocytes in monkey cer- eny and origin. Brain Res Rev 63:72–82.
ebral cortex. J Comp Neurol 371:153–163. Turmaine M, Raza A, Mahal A, Mangiarini L, Bates GP, Davies SW.
Peters A. 2002. Structural changes that occur during normal aging of 2000. Nonapoptotic neurodegeneration in a transgenic mouse model
primate cerebral hemispheres. Neurosci Biobehav Rev 26:733–741. of Huntington’s disease. Proc Natl Acad Sci USA 97:8093–8097.
Peters A, Josephson K, Vincent SL. 1991a. Effects of aging on the neu- Turrigiano GG, Nelson SB. 2004. Homeostatic plasticity in the develop-
roglial cells and pericytes within area 17 of the rhesus monkey cere- ing nervous system. Nat Rev Neurosci 5:97–107.
bral cortex. Anat Rec 229:384–398. Uemura E. 1985. Age-related changes in the subiculum of Macaca
Peters A, Palay SL, Webster H. 1991b. The fine structure of the nerv- mulatta: Dendritic branching pattern. Exp Neurol 87:412–427.
ous system: The neurons and supporting cells. Philadelphia: W.B. Valor LM, Jancic D, Lujan R, Barco A. 2010. Ultrastructural and tran-
Saunders. 528 p. scriptional profiling of neuropathological misregulation of CREB
Peters A, Sethares C. 2003. Is there remyelination during aging of the function. Cell Death Differ 17:1636–1644.
primate central nervous system? J Comp Neurol 460:238–254. Vaughan DW, Peters A. 1974. Neuroglial cells in the cerebral cortex of
Peters A, Sethares C. 2004. Oligodendrocytes, their progenitors and rats from young adulthood to old age: An electron microscope study. J
other neuroglial cells in the aging primate cerebral cortex. Cereb Cor- Neurocytol 3:405–429.
tex 14:995–1007. Vijg J, Campisi J. 2008. Puzzles, promises and a cure for ageing. Na-
Porte Y, Buhot MC, Mons N. 2008. Alteration of CREB phosphorylation ture 454:1065–1071.
and spatial memory deficits in aged 129T2/Sv mice. Neurobiol Aging Vitolo OV, Sant’Angelo A, Costanzo V, Battaglia F, Arancio O, Shelanski
29:1533–1546. M. 2002. Amyloid beta-peptide inhibition of the PKA/CREB pathway
Prinz M, Priller J, Sisodia SS, Ransohoff RM. 2011. Heterogeneity of and long-term potentiation: Reversibility by drugs that enhance
CNS myeloid cells and their roles in neurodegeneration. Nat Neuro- cAMP signaling. Proc Natl Acad Sci USA 99:13217–13221.
sci 13:1227–1235. von Bernhardi R, Tichauer JE, Eugenin J. 2010. Aging-dependent
Rapp PR, Gallagher M. 1996. Preserved neuron number in the hippo- changes of microglial cells and their relevance for neurodegenerative
campus of aged rats with spatial learning deficits. Proc Natl Acad Sci disorders. J Neurochem 112:1099–1114.
USA 93:9926–9930.
Wake H, Lee PR, Fields RD. 2011. Control of local protein synthesis and
Riga D, Riga S, Halalau F, Schneider F. 2006. Brain lipopigment accu-
initial events in myelination by action potentials. Science 333:1647–
mulation in normal and pathological aging. Ann N Y Acad Sci
1651.
1067:158–163.
Rittenhouse CD, Majewska AK. 2009. Synaptic mechanisms of activity- Wake H, Moorhouse AJ, Jinno S, Kohsaka S, Nabekura J. 2009. Rest-
dependent remodeling in visual cortex. J Exp Neurosci 2:23–41. ing microglia directly monitor the functional state of synapses in vivo
Sharts-Hopko N. 2009. Low vision and blindness among midlife and and determine the fate of ischemic terminals. J Neurosci 29:3974–
older adults: A review of the nursing research literature. Holist Nurs 3980.
Pract 23:94–100. Wecker JR, Ison JR. 1986. Visual function measured by reflex modifica-
Sheffield LG, Berman NE. 1998. Microglial expression of MHC class II tion in rats with inherited retinal dystrophy. Behav Neurosci 100:679–
increases in normal aging of nonhuman primates. Neurobiol Aging 684.
19:47–55. West MJ. 1993. Regionally specific loss of neurons in the aging human
Sheng JG, Mrak RE, Griffin WS. 1998. Enlarged and phagocytic, but hippocampus. Neurobiol Aging 14:287–293.
not primed, interleukin-1 alpha-immunoreactive microglia increase Wilhelmsson U, Bushong EA, Price DL, Smarr BL, Phung V, Terada M,
with age in normal human brain. Acta Neuropathol 95:229–234. Ellisman MH, Pekny M. 2006. Redefining the concept of reactive
Shimohata T, Nakajima T, Yamada M, Uchida C, Onodera O, Naruse S, astrocytes as cells that remain within their unique domains upon
Kimura T, Koide R, Nozaki K, Sano Y, Ishiguro H, Sakoe K, Ooshima reaction to injury. Proc Natl Acad Sci USA 103:17513–17518.
T, Sato A, Ikeuchi T, Oyake M, Sato T, Aoyagi Y, Hozumi I, Nagatsu Williams GA, Jacobs GH. 2007. Cone-based vision in the aging mouse.
T, Takiyama Y, Nishizawa M, Goto J, Kanazawa I, Davidson I, Vision Res 47:2037–2046.
Tanese N, Takahashi H, Tsuji S. 2000. Expanded polyglutamine Willott JF. 1986. Effects of aging, hearing loss, and anatomical location
stretches interact with TAFII130, interfering with CREB-dependent on thresholds of inferior colliculus neurons in C57BL/6 and CBA
transcription. Nat Genet 26:29–36. mice. J Neurophysiol 56:391–408.
Sierra A, Gottfried-Blackmore AC, McEwen BS, Bulloch K. 2007. Willott JF, Bross LS, McFadden SL. 1994. Morphology of the inferior
Microglia derived from aging mice exhibit an altered inflammatory colliculus in C57BL/6J and CBA/J mice across the life span. Neuro-
profile. Glia 55:412–424. biol Aging 15:175–183.
Streit WJ. 2002. Microglia as neuroprotective, immunocompetent cells Woodruff-Pak DS, Foy MR, Akopian GG, Lee KH, Zach J, Nguyen KP,
of the CNS. Glia 40:133–139. Comalli DM, Kennard JA, Agelan A, Thompson RF. 2010. Differential
Streit WJ, Miller KR, Lopes KO, Njie E. 2008. Microglial degeneration effects and rates of normal aging in cerebellum and hippocampus.
in the aging brain—bad news for neurons? Front Biosci 13:3423– Proc Natl Acad Sci USA 107:1624–1629.
3438. Yang DS, Kumar A, Stavrides P, Peterson J, Peterhoff CM, Pawlik M,
Streit WJ, Sammons NW, Kuhns AJ, Sparks DL. 2004. Dystrophic Levy E, Cataldo AM, Nixon RA. 2008. Neuronal apoptosis and
microglia in the aging human brain. Glia 45:208–212. autophagy cross talk in aging PS/APP mice, a model of Alzheimer’s
Streit WJ, Walter SA, Pennell NA. 1999. Reactive microgliosis. Prog disease. Am J Pathol 173:665–681.
Neurobiol 57:563–581. Zettel ML, Frisina RD, Haider SE, O’Neill WE. 1997. Age-related
Tobias JV, Bilger RC, Brody H, Gates GA, Haskell G, Howard D, Mar- changes in calbindin D-28k and calretinin immunoreactivity in the
shall LA, Nerbonne MA, Pickett JA. 1988. Speech understanding and inferior colliculus of CBA/CaJ and C57Bl/6 mice. J Comp Neurol
aging. Working Group on Speech Understanding and Aging. Commit- 386:92–110.
tee on Hearing, Bioacoustics, and Biomechanics, Commission on Zettel ML, Zhu X, O’Neill WE, Frisina RD. 2007. Age-related decline in
Behavioral and Social Sciences and Education, National Research Kv3.1b expression in the mouse auditory brainstem correlates with
Council. J Acoust Soc Am 83:859–895. functional deficits in the medial olivocochlear efferent system.
Tomobe K, Okuma Y, Nomura Y. 2007. Impairment of CREB phospho- J Assoc Res Otolaryngol 8:280–293.
rylation in the hippocampal CA1 region of the senescence-accelerated Zimniak P. 2008. Detoxification reactions: Relevance to aging. Ageing
mouse (SAM) P8. Brain Res 1141:214–217. Res Rev 7:281–300.

GLIA

You might also like