You are on page 1of 9

Research

JAMA Neurology | Original Investigation

Association of Perivascular Localization of Aquaporin-4


With Cognition and Alzheimer Disease in Aging Brains
Douglas M. Zeppenfeld, BS; Matthew Simon, BS; J. Douglas Haswell, BS; Daryl D’Abreo; Charles Murchison, MS; Joseph F. Quinn, MD;
Marjorie R. Grafe, MD, PhD; Randall L. Woltjer, MD, PhD; Jeffrey Kaye, MD; Jeffrey J. Iliff, PhD

Supplemental content
IMPORTANCE Cognitive impairment and dementia, including Alzheimer disease (AD), are
common within the aging population, yet the factors that render the aging brain vulnerable to
these processes are unknown. Perivascular localization of aquaporin-4 (AQP4) facilitates the
clearance of interstitial solutes, including amyloid-β, through the brainwide network of
perivascular pathways termed the glymphatic system, which may be compromised in the
aging brain.

OBJECTIVES To determine whether alterations in AQP4 expression or loss of perivascular


AQP4 localization are features of the aging human brain and to define their association with
AD pathology.

DESIGN, SETTING, AND PARTICIPANTS Expression of AQP4 was analyzed in postmortem


frontal cortex of cognitively healthy and histopathologically confirmed individuals with AD by
Western blot or immunofluorescence for AQP4, amyloid-β 1-42, and glial fibrillary acidic
protein. Postmortem tissue and clinical data were provided by the Oregon Health and Science
University Layton Aging and Alzheimer Disease Center and Oregon Brain Bank. Postmortem
tissue from 79 individuals was evaluated, including cognitively intact “young” individuals aged
younger than 60 years (range, 33-57 years), cognitively intact “aged” individuals aged older
than 60 years (range, 61-96 years) with no known neurological disease, and individuals older
than 60 years (range, 61-105 years) of age with a clinical history of AD confirmed by
histopathological evaluation. Forty-eight patient samples (10 young, 20 aged, and 18 with
AD) underwent histological analysis. Sixty patient samples underwent Western blot analysis
(15 young, 24 aged, and 21 with AD).

MAIN OUTCOMES AND MEASURES Expression of AQP4 protein, AQP4 immunoreactivity, and
perivascular AQP4 localization in the frontal cortex were evaluated.

RESULTS Expression of AQP4 was associated with advancing age among all individuals
(R2 = 0.17; P = .003). Perivascular AQP4 localization was significantly associated with AD
status independent of age (OR, 11.7 per 10% increase in localization; z = −2.89; P = .004) and
was preserved among eldest individuals older than 85 years of age who remained cognitively
intact. When controlling for age, loss of perivascular AQP4 localization was associated with Author Affiliations: Department of
increased amyloid-β burden (R2 = 0.15; P = .003) and increasing Braak stage (R2 = 0.14; Anesthesiology and Perioperative
P = .006). Medicine, Oregon Health and Science
University, Portland (Zeppenfeld,
Simon, Haswell, D’Abreo, Grafe, Iliff);
CONCLUSIONS AND RELEVANCE In this study, altered AQP4 expression was associated with Department of Neurology, Oregon
aging brains. Loss of perivascular AQP4 localization may be a factor that renders the aging Health and Science University,
brain vulnerable to the misaggregation of proteins, such as amyloid-β, in neurodegenerative Portland (Murchison, Quinn, Kaye);
Department of Pathology, Oregon
conditions such as AD.
Health and Science University,
Portland (Grafe, Woltjer); Knight
Cardiovascular Research Institute,
Oregon Health and Science
University, Portland (Iliff).
Corresponding Author: Jeffrey J.
Iliff, PhD, Department of
Anaesthesiology and Perioperative
Medicine, Oregon Health and Science
University, 3181 SW Sam Jackson Park
JAMA Neurol. 2017;74(1):91-99. doi:10.1001/jamaneurol.2016.4370 Rd, Mail Code L459, Portland, OR
Published online November 28, 2016. 97239 (iliffj@ohsu.edu).

(Reprinted) 91

Copyright 2017 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by b Wildberries on 02/25/2023


Research Original Investigation Aquaporin-4 Localization and Cognition in Aging Brains

A
dvancing age is the strongest risk factor for the devel-
opment of neurodegenerative disorders such as Key Points
Alzheimer disease (AD).1 A common feature of these
Question Is the expression or localization of the astroglial water
diseases is the age-associated accumulation of protein aggre- channel aquaporin-4 altered in patients with advanced age or with
gates, including senile plaques comprised of amyloid-β (Aβ) Alzheimer disease?
1-42 in AD, α-synuclein within Lewy bodies in Parkinson
Findings In this postmortem analysis, aquaporin-4 protein
disease and Lewy body dementia, and hyperphosphorylated
expression and localization in the cortex of a series of aged
tau within neurofibrillary tangles in AD and chronic trau- cognitively intact individuals and patients with Alzheimer disease
matic encephalopathy, 2 yet the upstream changes that revealed statistically significant associations between aquaporin-4
render the aging brain vulnerable to the misaggregation of expression and aging. Loss of aquaporin-4 protein localization to
proteins remain unknown. perivascular astrocytic endfeet was strongly associated with
Since 2013, we have defined a brain wide perivascular path- Alzheimer disease status and pathology.
way, termed the glymphatic system, that facilitates the recir- Meaning Increasing aquaporin-4 expression is a feature of the
culation of cerebrospinal fluid (CSF) through the brain aging human brain, and mislocalization of aquaporin-4 is related to
parenchyma and supports the clearance of interstitial sol- the development of Alzheimer disease pathology.
utes including Aβ and tau.3-7 Perivascular exchange of CSF
and interstitial fluid is dependent on the astroglial water
channel aquaporin-4 (AQP4), which is localized to peri- in frozen and fixed frontal cortical tissue, respectively. De-
vascular astrocytic endfeet that ensheathe the cerebral tailed descriptions of these methods are provided in the
vasculature.7 We demonstrated that perivascular CSF recir- eMethods in the Supplement.
culation and Aβ clearance are impaired in the aging mouse
brain, impairment that was associated with the loss of peri- Statistical Evaluation
vascular AQP4 localization. Prior studies in postmortem A detailed description of statistical approaches is provided in
human tissue show that AQP4 is up regulated8,9 and that the eMethods in the Supplement. Multiple comparisons cor-
localization of AQP4 to the cerebral vasculature is disrupted rected P values, means, and standard errors of the mean are
in the AD cortex.10 This suggests that age-related mislocal- indicated in the article. All statistical analyses were carried out
ization of AQP4 may slow glymphatic function and promote using Prism 6 (GraphPad) and R 3.2.1 (R Foundation).13
protein aggregation and neurodegeneration. In this study,
we assessed AQP4 expression and perivascular localization
in brain samples from a human case series including indi-
viduals of different ages and with different cognitive and
Results
neuropathological AD profiles. Demographic and Histopathological Characteristics
Expression of AQP4 and localization were evaluated from a
cohort of 79 individuals including “young” control indi-
viduals younger than 60 years of age (range, 33-57 years)
Methods without a history of neurological disease, cognitively intact
Human Participants and Tissues (Clinical Dementia Rating14 = 0) “aged” individuals older
This study consisted of 79 patients from the Oregon Health and than 60 years of age (range, 61-96 years), and individuals
Science University Layton Aging and Alzheimer Disease Cen- with histopathologically confirmed AD older than 60 years
ter and associated postmortem tissue repository, the Oregon of age (range, 61-105 years) . Among these, 48 (10 young, 20
Brain Bank. The Oregon Health and Science University insti- aged, and 18 with AD) underwent histological analysis. Sixty
tutional review board approved the study. Volunteers signed underwent Western blot analysis (15 young, 24 aged, and 21
written informed consent. All aged participants were commu- with AD) of AQP4. Twenty-nine individuals (11 aged and 18
nity-dwelling individuals with no known neurological dis- with AD) underwent both histological and Western blot
ease (control individuals) or with a clinical history of AD as es- analysis. The Table shows sex balance and median and
tablished by neurologic evaluation in the Layton Aging and interquartile ranges of age, years of schooling, Aβ plaque
Alzheimer Disease Center as previously described11 and in ac- density, and Braak stage for all participants. Most recent
cordance with established consensus criteria.12 Brain au- Mini-Mental State Examination and Clinical Dementia Rat-
topsy was performed on all participants after consent was ob- ings are provided for a subset of aged individuals and indi-
tained from the next of kin and in accordance with Oregon viduals with AD who underwent cognitive testing at the
Health and Science University guidelines. Layton Aging and Alzheimer Disease Center. No significant
differences between groups were observed in sex balance
Neuropathological Evaluation and Evaluation (χ2 = 0.51; P = .78), years of schooling (t = 0.39; P = .70), or
of AQP4 Expression and Localization postmortem interval (F = 2.37; P = .10). Linear regression
Brains in the Oregon Brain Bank underwent neuropathologi- showed no association between postmortem interval and
cal evaluation for Aβ plaque density, neurofibrillary pathol- global AQP4-immunoreactivity (IR) or perivascular AQP4
ogy, and vascular pathology. Expression of AQP4 and localiza- localization assessed by immunofluorescence (eFigure 1A in
tion was evaluated by Western blot and immunofluorescence the Supplement, R 2 = 0.01; P = .44; eFigure 1B in the

92 JAMA Neurology January 2017 Volume 74, Number 1 (Reprinted) jamaneurology.com

Copyright 2017 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by b Wildberries on 02/25/2023


Aquaporin-4 Localization and Cognition in Aging Brains Original Investigation Research

Table. Cohort Demographic, Cognitive, and Pathological Features

Demographic Features Cognitive Features Alzheimer Pathology


Sex
Balance PostMortem Years of Amyloid Plaque Braak Stage
Age Age, (Male/ Interval, Schooling, MMSE, CDR, Density (0-3), (0-6),
Range No. Median (IQR) Female) Median (IQR) Median (IQR) No. Median (IQR) Median (IQR) No. Median (IQR) Median (IQR)
Young 25 44.0 (14/11) 17.8 NA 0 NA NA 25 0.0 0.0
(<60 y) (37.0-51.0) (10.9-32.3) (0.0-0.0) (0.0-0.0)
Aged 33 83.0 (14/19) 13.5 14.0 15 28.0 0.0 33 0.0 1.0
(>60 y) (69.0-87.6) (5.0-21.8) (12.0-18.0) (27.0-29.0) (0.0-0.0) (0.0-1.0) (0.0-3.0)a
Alzheimer 21 81.3 (10/11) 12.0 14.0 16 18.0 1.0 21 2.0 6.0
Disease (67.0-95.4) (6.0-24.0) (12.0-16.0) (9.0-22.5) (0.5-2.0) (1.5-3.0)b,c (4.0-6.0)b,c
(>60y)
b
Abbreviations: CDR, Clinical Dementia Rating; IQR, interquartile range. Padj<.001 vs young individuals.
a c
Padj<.05. Padj<.001 vs aged individuals (Kruskal-Wallis test).

Supplement, R 2 = 0.02; P = .29; Pearson correlation) or (Figure 1B, arrowhead). Expression and localization of AQP4
postmortem interval and AQP4 expression evaluated by were dramatically altered in the aged brain. While intense AQP4
Western blot (eFigure 1C in the Supplement, R 2 = 0.01; expression was maintained near the cortical surface, AQP4
P = .39; Pearson correlation). The mean (SE) interval expression became discontinuous below cortical layer II
between age at most recent cognitive testing and death did (Figure 1C) where AQP4-IR remained intense within a sparse
not differ between aged individuals (0.9 [0.2] years) and subpopulation of astrocytes scattered throughout the deeper
individuals with AD (1.9 [0.5] years) (P = .13, independent cortical layers (Figure 1D). When compared between groups,
samples t test). Histopathological examination revealed the mean (SD) AQP4-IR was significantly increased among
greater Aβ plaque density in AD compared with young or individuals with AD (31.57 [2.84] arbitrary units) compared
aged individuals (AD vs young: Dunn’s z = 6.64; adjusted with young individuals (20.21 [0.99] arbitrary units;
P <.001; AD vs old: Dunn’s z = 5.27; adjusted P <.001; P = .004; Welch-corrected 1-way ANOVA) or aged individuals
Kruskal-Wallis test). Individuals with AD had higher Braak (23.78 [1.31] arbitrary units; adjusted P = .029; Welch-
scores compared with aged or young individuals (AD vs corrected 1-way analysis of variance [ANOVA]) (Figure 1E).
young: Dunn’s z = 7.54; adjusted P <.001; AD vs old: Dunn’s Multiple linear regression analysis showed that when correct-
z = 4.58; adjusted P <.001; Kruskal-Wallis test); however, ing for the influence of age, increasing AQP4-IR was signifi-
aged individuals exhibited significantly higher Braak scores c antly associated with increasing Aβ plaque density
than young individuals (Dunn’s z = 3.59; adjusted P = .005; (Figure 1F; unadjusted P = .006; R2 = 0.21; adjusted for age
Kruskal-Wallis test). P = .01; R 2 = 0.30; Huber-White–corrected ordinary least
General cerebrovascular pathology (VIS) was rated on the squares [OLS]). In addition, increasing AQP4-IR also tended
degree of arteriolosclerosis, perivascular hemosiderin leak- to be associated with increasing Braak stage, a relationship
age, perivascular space dilation and myelin loss from the fron- persisting even when adjusting for the effects of age
tal cortex, and cortical amyloid angiopathy in the occipital cor- (Figure 1G; unadjusted P = .002; R2 = 0.15; adjusted for age
tex according to criteria published be Deramecourt et al.15 P = .03; R2 = 0.20; Huber-White–corrected OLS).
Median and interquartile ranges for VIS and subscores are In parallel with increasing global AQP4-IR, mean (SE) peri-
shown in the eTable in the Supplement. As shown in eFigure vascular AQP4 localization was reduced in individuals with AD
2A in the Supplement, VIS was significantly greater in the aged (1.095 [0.007]) compared with young individuals (1.160
individuals and individuals with AD compared with young in- [0.019]; adjusted P = 0.01; Welch-corrected 1-way ANOVA) or
dividuals (young vs old: Dunn’s z = −2.24; adjusted P = .04; aged individuals (1.161 [0.016]; P = .003; Welch-corrected
young vs AD: Dunn’s z = −3.99; adjusted P <.001, respec- 1-way ANOVA; Figure 1H). Regression analysis showed that
tively; Kruskal-Wallis test). Evaluation of the relationship be- when correcting for the influence of age, reduced perivascu-
tween age and VIS indicated a significant linear association lar AQP4 localization was associated with increasing Aβ
(eFigure 2B in the Supplement; R2 = 0.36; P < .001, Pearson cor- plaque density (Figure 1I; unadjusted P < .001; R2 = 0.15;
relation). adjusted for age P < .001; R2 = 0.14; Huber-White–corrected
OLS) and increasing Braak stage (Figure 1J; unadjusted
Evaluation of Perivascular AQP4 Localization P = .002; R 2 = 0.14; adjusted for age P = .004; R 2 = 0.12;
by Immunofluorescence Huber-White–corrected OLS).
When AQP4 expression and localization were evaluated by im- We next evaluated whether altered AQP4 expression or lo-
munofluorescence double-labeling, we observed that in the calization are general features of the aging brain. Regression
young cortex, AQP4 expression was uniform through the cor- analysis showed a significant association between increasing
tical layers spanning from the glia limitans to the subcortical age and increasing AQP4-IR (Figure 2A; P = .001; ρ = 0.45,
white matter (Figure 1A). Although expressed in fine pro- Spearman’s correlation), while no significant association
cesses throughout the neuropil, AQP4-IR was greatest in peri- was evident between age and alterations in perivascular AQP4
vascular endfeet surrounding the cerebral vasculature localization (Figure 2B; P = .15; ρ = −0.21, Spearman’s

jamaneurology.com (Reprinted) JAMA Neurology January 2017 Volume 74, Number 1 93

Copyright 2017 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by b Wildberries on 02/25/2023


Research Original Investigation Aquaporin-4 Localization and Cognition in Aging Brains

Figure 1. Loss of Perivascular Aquaporin-4 (AQP4) Localization Is Associated With Alzheimer Disease (AD) Pathology

A Young (uniform distribution) B Young (localization) C Aged AQP4 expression D Aged AQP4 localization

DAPI DAPI
DAPI AQP4 DAPI AQP4
AQP4 500 μm 50 μm 500 μm GFAP 50 μm
GFAP AQP4

E Global AQP4 expression F Global AQP4 expression G Global AQP4 expression


a
80 80 80
b

60 60 60
AQP4-IR, AU

AQP4-IR, AU
AQP4-IR, AU

P = .03
P = .01
40 40 40

20 20 20

0 0 0
Young Aged AD 0 1 2 3 0 1 2 3 4 5 6
Amyloid Plaque Density Braak Stage

H Perivascular AQP4 localization I Perivascular AQP4 localization J Perivascular AQP4 localization


d
1.4 1.4 1.4
c Cognitively intact
Alzheimer disease
1.3 1.3
AQP4 Ratio (PV:Parenchyma)

AQP4 Ratio (PV:Parenchyma)

AQP4 Ratio (PV:Parenchyma)

1.3
P = .004

1.2 P = .001 1.2


1.2

1.1 1.1 1.1

1.0 1.0 1.0

0.9 0.9 0.9


Young Aged AD 0 1 2 3 0 1 2 3 4 5 6
Amyloid Plaque Density Braak Stage

A, Perivascular AQP4 localization was evaluated by immunofluorescence. In the AQP4-IR in perivascular domains to global AQP4-IR) showing reduced
young cortex, AQP4 was distributed uniformly from the cortical surface to the perivascular localization in individuals with AD. I-J, Regression analysis shows
subcortical white matter boundary. B, High-power confocal imaging showing that when controlling for the effects of age, reduced perivascular AQP4 is
localization of AQP4 in perivascular astrocytic endfeet (arrowhead). C, In the significantly associated with both amyloid-β plaque density (age-adjusted
aged cortex (aged 60-85 years), AQP4 was intensely expressed in a sparse P < .001, R2 = 0.14) (I) and Braak stage (age-adjusted P = .004, R2 = 0.12;
population of cortical astrocytes, while wider AQP4 expression was reduced. Huber-White–corrected ordinary least squares) (J). DAPl indicates
D, High-magnification image showing altered AQP4 expression and localization L-diaminopimelate aminotransferase; GFAP indicates glial fibrillary acidic
in the aged cortex. E, Quantification of global AQP4-immunoreactivity (IR) protein; and PV indicates parenchyma volume.
showing increased AQP4 expression in the AD cortex compared with the a
P = .03.
cognitively intact young or aged cortex. F-G, Regression analysis shows that b
P = .004; Welch-corrected 1-way analysis of variance.
when controlling for the effects of age, increasing AQP4-IR is significantly
c
(P = .01, R2 = 0.30; Huber-White–corrected ordinary least squares) associated P = .01.
with amyloid-β plaque density (F), as is association with Braak stage (P = .029, d
P = .003; Welch-corrected 1-way analysis of variance.
R2 = 0.20) (G). H, Quantification of perivascular AQP4 localization (ratio of

94 JAMA Neurology January 2017 Volume 74, Number 1 (Reprinted) jamaneurology.com

Copyright 2017 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by b Wildberries on 02/25/2023


Aquaporin-4 Localization and Cognition in Aging Brains Original Investigation Research

Figure 2. Loss of Perivascular Aquaporin-4 (AQP4) Localization Predicts Alzheimer Disease (AD) Status

Cognitively intact (25-85 y)


A Regression analysis C Logistic regression analysis Cognitively intact − eldest (>85 y)
Alzheimer disease (>60 y)
80 80

P = .01

60 60
AQP4-IR, AU

AQP4-IR, AU
40 40

P = .001

20 20

0 0
40 60 80 100 Young Aged Eldest Aged Eldest
Age, y Cognitively Intact Alzheimer Disease

B Perivascular AQP4 localization D Perivascular AQP4 localization

1.4 1.4
P < .001

1.3 1.3
AQP4 Ratio, PV:Parenchyma
AQP4 Ratio, PV:Parenchyma

1.2 1.2

1.1 1.1

1.0 1.0
P = .02

0.9 0.9
40 60 80 100 Young Aged Eldest Aged Eldest
Age, y Cognitively Intact Alzheimer Disease

A, Regression analysis shows that among all individuals, increasing age is age (oval) diverged markedly from eldest individuals with AD. Results of
significantly associated with increasing AQP4-immunoreactivty (IR) (P = .001, White-Huber–corrected logistic regression analysis indicated that AQP4-IR (C)
ρ = 0.45, Spearman’s correlation). B, Perivascular AQP4 localization is not and perivascular AQP4 localization (D) are significant predictors of AD status
significantly associated with age across all individuals; however, among (P = .01, age-adjusted P = .29; P < .001, age-adjusted P = .02; respectively).
individuals with AD, increasing age was associated with decreasing perivascular Significance of AQP4 as a covariate predicting cognitive status shown in the
localization (P = .02, ρ = −0.57, Spearman’s correlation). Perivascular AQP4 figure. PV indicates parenchyma volume.
localization among cognitively intact eldest individuals older than 85 years of

correlation). Strikingly, although perivascular AQP4 localiza- b = −24.6; z = −3.58; Figure 2D). In contrast, increas-ing
tion values were similar between aged individuals and indi- AQP4-IR was significantly associated with AD status (P = .01;
viduals with AD between 60 and 85 years of age, there was a b = 0.13; z = 2.45) in our cohort, while age showed no associa-
marked divergence between values among cognitively intact tion (P = .29; b = 0.017; z = 1.07; Figure 2C).
individuals and individuals with AD older than 85 years of age Although both AQP4-IR and perivascular AQP4 localiza-
(referred to as “eldest” individuals; Figure 2B, oval). Among tion were associated with vascular pathology (P = .05; R2 = 0.07
individuals with AD, increasing age was significantly associ- and P = .05; R2 = 0.07, respectively), this association disap-
ated with reduced perivascular AQP4 localization (P = .02; peared when the effects of age were controlled for (P = .83;
ρ= −0.57, Spearman’s correlation). R2 = 0.17 and P = .17; R2 = 0.05, respectively; Huber-White–
The results of Huber-White–corrected logistic regression corrected OLS). These results suggest that increasing vascular
analysis modeling AD status (cognitively intact vs AD) with age pathology and increasing global AQP4-IR are general features
and perivascular AQP4 localization as independent variables of the aging brain with the preservation of perivascular AQP4
indicated that age was a significant predictor (adjusted P = .02; localization a significant predictor of preserved cognitive func-
b = 0.037; z = 2.36) and decreasing perivascular AQP4 local- tion, even in individuals who remain cognitively intact late
ization was significantly associated with AD status (P < .001; into life.

jamaneurology.com (Reprinted) JAMA Neurology January 2017 Volume 74, Number 1 95

Copyright 2017 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by b Wildberries on 02/25/2023


Research Original Investigation Aquaporin-4 Localization and Cognition in Aging Brains

Figure 3. Altered Aquaporin-4 (AQP4) Expression in Deep Layers of the Figure 4. Amyloid-β (Aβ) Plaques Are Associated With Aquaporin-4
Aging Cortex (AQP4) Mislocalization

A Young (25-45 y) B Aged (60-85 y) C Eldest (>85 y) A Young (25-45 y) B Aged (60-85 y) C Alzheimer (60-85 y)

Gyrus
Gyrus Gyrus
Cortical Gyrus

500 μm 500 μm 500 μm


AQP4

D Young (25-45 y) E Aged (60-85 y) F Eldest (>85 y) Sulcus


Sulcus
Sulcus
WM
WM WM

DAPI
DAPI DAPI AQP4
1 mm 1 mm Aβ1-42 1 mm
AQP4 AQP4
Cortical Sulcus

500 μm 500 μm 500 μm


250 μm 150 μm 250 μm

Wide field confocal montages show AQP4 immunoreactivity extending from


the cortical surface (dashed line) at the crests of gyri (A-C) and the depths of
sulci (D-F). Compared with the uniform AQP4 expression throughout different D Aβ plaque–associated AQP4 E Aβ plaque–associated AQP4
cortical layers in young cortex (A and D), AQP4 labeling in cortical layers II to IV
of cognitively intact aged (60-85 years) individuals (B and E) was reduced, with
the appearance of intensely immunoreactive astrocytes throughout these
layers. These differences were not observed among cognitively intact eldest
individuals (older than 85 years) (C and F).
DAPI DAPI
AQP4 200 μm 200 μm AQP4 50 μm
50 μm
Aβ1-42 GFAP

Regional Patterns of AQP4 Localization Large wide-field confocal montages extending from the crests of sulci to the
When evaluating the spatial pattern of AQP4 expression in the base of gyri were generated. A, AQP4-immunoreactivity (IR) in the young brain
cortex, we observed that compared with the uniform distri- is uniform through the depth of the cortex to the gray matter−white matter
(WM) boundary. Insets depict a region of the montage at higher magnification.
bution of AQP4 through layers II to VI of the young cortex B, In the cognitively intact aged cortex (60-85 years), AQP4-IR is reduced in
(Figure 3A and E), AQP4 localization was dramatically altered deeper cortical layers (blue arrowhead), while a band of intense IR runs along
in the cortex of aged individuals (Figure 3B and F). Interest- the gray matter−WM boundary (white arrowhead) within the cortex at the crest
of the gyrus but not in the cortex in the depth of the sulcus. C, In individuals
ingly, different layer-specific patterns of AQP4 expression
with Alzheimer disease, AQP4-IR in deeper cortical layers is increased compared
were evident among aged individuals at the crests of gyri vs with aged individuals, owing to the presence of AQP4-IR associated with Aβ
the depths of sulci. At the crests of gyri of aged individuals, plaques (dashed circles in inset). D, High-power confocal microscopy shows
AQP4 expression was lost between layers II and IV, while a that Aβ plaques are surrounded by foci of AQP4-IR. E, These foci are features of
Aβ plaque-associated reactive astrocytes (glial fibrillary acidic protein).
band of AQP4-IR remained in layers V to VI at the gray
matter–white matter boundary (Figure 3A-B). At the depths
of sulci of aged individuals, an intense band of AQP4-IR intense band of AQP4 immunofluorescence observed super-
remained in cortical layer II, while AQP4 expression within ficially in layer II of aged individuals was absent in the cogni-
deeper layers was reduced (Figure 3D-E). Figure 4A-B shows tively intact eldest individuals (Figure 3F). Thus, the regional
representative multifield montages depicting the effects of patterns of altered AQP4 expression and localization that are
age on AQP4 expression along the length of whole gyri from features of the aged brain are absent among the cognitively
young and aged individuals. Such regional patterns of intact eldest individuals.
AQP4-IR were observed in whole-gyrus images from 6 to 7
individuals per group. Evaluation of AQP4 Expression by Western Blot
Among cognitively intact eldest individuals (older than 85 Expression of AQP4 in the frontal cortex was evaluated by West-
years), AQP4 expression in the middle layers of the cortex was ern blot. As shown in eFigure 3A in the Supplement, no sig-
maintained (Figure 3C). Within the depths of the sulci, the nificant differences in total AQP4 expression were observed

96 JAMA Neurology January 2017 Volume 74, Number 1 (Reprinted) jamaneurology.com

Copyright 2017 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by b Wildberries on 02/25/2023


Aquaporin-4 Localization and Cognition in Aging Brains Original Investigation Research

between young individuals, aged individuals, or individuals Aβ plaque deposition.17 A similar analysis in 2015 demon-
with AD (P = .23, 1-way ANOVA). The discrepancy between strated that Aβ clearance slows with increasing age, includ-
these findings and the age-related differences in AQP4-IR ing in individuals prior to the deposition of Aβ plaques as
(Figure 2A-B) may stem from the fact that our immunofluo- detected by Pittsburgh compound B positron emission
rescence analysis specifically omitted analysis of cortical layer tomographic scans.18 The changes in the aging human brain
I and the glial limitans, which exhibit pronounced AQP4-IR in that underlie the slowing of Aβ clearance have not yet been
the aged and AD cortex (Figure 3 and Figure 4). Aquaporin-4 defined.
is expressed in the human brain as 2 alternative isoforms, Amyloid β clearance from the brain appears to involve sev-
AQP4-M1 and AQP4-M23, the relative expression of which has eral different processes, including uptake and degradation by
been proposed to drive perivascular localization.16 When we brain cells such as microglia; efflux across the blood-brain bar-
evaluated expression of AQP4-M1 and AQP4-M23 separately, rier by transporters, such as low-density lipoprotein receptor-
we did not observe any specific differences in expression of related protein-1; and clearance by bulk flow along perivascu-
either isoform between groups. However, when the ratio of lar pathways. 19-21 Because each takes place within the
AQP4-M1 to AQP4-M23 expression was evaluated, we ob- perivascular compartment, it appears likely that processes such
served that the relative expression of AQP4-M1 (the AQP4-M1 as perivascular bulk clearance and blood-brain barrier efflux
to AQP4-M23 ratio) significantly declined in aged cortex (mean interact to support Aβ clearance from the brain.4,22 The pre-
[SD], 0.10 [0.09]; adjusted P = .05, 1-way ANOVA) and AD cise nature and extent of this interaction, and how these re-
cortex (mean [SD], 0.13 [0.02]; adjusted P = .01, 1-way ANOVA) lationships may change as the brain ages, remain unknown and
(eFigure 3B in the Supplement). are an important subjects for future research.
In the rodent brain, perivascular CSF–interstitial fluid ex-
Differences in AQP4 Localization change and interstitial Aβ clearance are markedly reduced in
Associated With Senile Plaques the absence of the astroglial water channel AQP4,7 while in a
Participants with AD exhibited increased AQP4-IR in cortex transgenic mouse model of AD, deletion of the Aqp4 gene in-
(Figure 1E) in addition to reduced perivascular AQP4 local- creased Aβ plaque formation and worsened cognitive
ization (Figure 1H). Wide field immunofluorescence imaging impairment.23 In the aging mouse cortex, impairment of peri-
showed that in contrast to the pattern of AQP4-IR in the vascular CSF–interstitial fluid exchange and interstitial Aβ clear-
aged cortex (Figure 4B), AQP4-IR in the AD cortex remained ance was associated with reduced perivascular AQP4
high through all cortical depths (Figure 4C). Confocal localization.4 These experimental findings suggest that loss of
microscopy showed that this was attributable to intense perivascular localization in the aging rodent brain slows Aβ
AQP4-IR associated within glial fibrillary acidic protein– clearance and promotes Aβ deposition. Our findings here dem-
positive reactive astrocytes surrounding cortical senile onstrate that similar age-related changes to astroglial AQP4 lo-
plaques (Figure 4C-E). calization are occurring in the human as in the rodent brain,
and suggest that AQP4 mislocalization may be 1 of the factors
underlying the slowing of Aβ clearance and promoting Aβ
plaque formation in the aging human brain.
Discussion Our findings are consistent with prior studies carried out
We have evaluated the association between differences in AQP4 in human postmortem cortical tissue showing that while AQP4
expression and localization and Alzheimer pathology across expression evaluated by Western blot does not significantly
a wide range of aged human samples, including those from cog- change in the AD cortex,24 AQP4 immunoreactivity is substan-
nitively intact individuals and individuals with AD. We dis- tially increased in the AD cortex.8,9 Additionally, Wilcock et al10
covered that AQP4 expression increases in the aging brain and reported that AQP4 localization to the microvasculature is re-
that loss of perivascular AQP4 localization is associated with duced among individuals with AD. In prior studies, disrup-
worsening AD pathology. We observed that AQP4 expression tion of AQP4 expression and localization have been reported
increased with age across all individuals. Regional patterns of to be associated with both Aβ plaques and vascular Aβ
AQP4 expression and localization were dramatically altered in deposits,9,24,25 suggesting that changes in AQP4 expression and
the aged cortex (60-85 years of age) yet were preserved among localization are features of the aging brain. This study ex-
cognitively intact individuals older than 85 years. Perivascu- tends these previous findings to include what is, to our knowl-
lar AQP4 localization was reduced among individuals with AD edge, the largest clinical case series to date to assess the rela-
and reduced localization was strongly associated with in- tionship between changes in AQP4 expression and localization,
creased neurofibrillary and Aβ pathology, even when control- aging, and AD pathology. Importantly, to our knowledge, this
ling for the effect of age on AD pathology. In line with these is the first study to explicitly quantify perivascular AQP4 lo-
findings, logistic regression analysis revealed that perivascu- calization and to evaluate whether changes in this feature,
lar AQP4 localization was strongly associated with AD status which is believed to underlie perivascular Aβ clearance along
independent of age. the glymphatic pathway,4,5,7,26 are significantly associated with
Analysis of Aβ kinetics within the CSF of clinical partici- AD status or AD pathology as measured by Braak stage or Aβ
pants showed that Aβ clearance is impaired among par- plaque density.
ticipants with late-onset AD, suggesting that slowing Aβ We did not observe evidence of Aβ plaque–associated dif-
clearance rather than increasing Aβ production underpins ferences in AQP4 expression or localization in cognitively

jamaneurology.com (Reprinted) JAMA Neurology January 2017 Volume 74, Number 1 97

Copyright 2017 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by b Wildberries on 02/25/2023


Research Original Investigation Aquaporin-4 Localization and Cognition in Aging Brains

intact young or aged individuals. The relationship between in- Limitations


creasing AQP4-IR and advancing age and between differ- In experimental studies in rodents, the role of AQP4 in glym-
ences in AQP4 localization and increasing Aβ plaque deposi- phatic system function has been best characterized in the cor-
tion and neurofibrillary pathology observed even among tex, while the spread of amyloid and neurofibrillary pathol-
cognitively intact individuals argues that the loss of perivas- ogy throughout the frontal cortex is believed to play a key role
cular AQP4 localization is not the result of advancing AD pa- in the development of cognitive decline in the progression of
thology. However, we cannot rule out the possibility that sub- AD. Therefore, this initial study was limited to analysis of the
clinical Aβ plaque burden or neurofibrillary pathology, which expression and localization of AQP4 in the frontal cortex.
is present both among cognitively intact individuals and those Whether these age-associated effects also occur in other brain
with mild cognitive impairment,27 contributed to differences areas relevant to cognitive decline, such as the hippocampus
in AQP4 expression or localization in this study. However, the and entorhinal cortex, is an important consideration. Be-
absence of a significant association between cerebrovascular cause this study is reliant on postmortem tissue, it is impos-
pathology and differences in AQP4 expression or localization sible from these data to verify whether AQP4 mislocalization
when controlling for effects of age suggest that these effects is itself the causative feature rendering the aging brain sus-
are likely not driven by vascular injury. ceptible to neurodegenerative processes or merely a parallel
Age-related cognitive decline in the absence of frank de- consequence of an independent upstream pathological pro-
mentia is commonly viewed as an inherent concomitant of cess. Additionally, whether preservation of perivascular AQP4
normal aging.28 Yet among the nondemented aging popula- localization throughout the aging process provides protec-
tion, an important subset harbor a high prevalence of the neu- tion or a reserve against development of AD pathology or
ropathologies that are commonly associated with frank whether other factors contribute independently to both re-
dementia.29 The substrate of this resistance to age-related pa- duced AD pathology and increased perivascular AQP4 local-
thologies and cognitive decline is not well understood but ization cannot be ascertained from these data. Further in-
has been attributed to aspects of greater brain reserve or quiry into these questions is warranted.
resilience.30 In this study, we observed loss of perivascular
AQP4 localization with increasing age among individuals with
AD, while perivascular AQP4 localization was preserved in in-
dividuals older than 85 years of age. Similarly, regional differ-
Conclusions
ences in AQP4 expression and localization observed in aging These histological findings are consistent with the notion that
individuals 60 to 85 years of age were generally absent among loss of perivascular AQP4 localization is one of the factors that
these cognitively intact individuals older than 85 years of age. renders the aging brain vulnerable to Aβ aggregation and neu-
Whether preservation of perivascular AQP4 localization rodegeneration. If substantiated, this would potentially place
throughout the aging process provides protection or a re- age-related changes in perivascular AQP4 localization mecha-
serve against the development of AD pathology or whether nistically upstream of processes of protein misaggregation and
other factors contribute independently to both reduced AD pa- would suggest that targeting AQP4 localization may provide
thology and increased perivascular AQP4 localization cannot a therapeutic approach to intervene in the AD pathogenesis up-
be ascertained from this data. stream of Aβ plaque or neurofibrillary tangle formation.

ARTICLE INFORMATION Funding/Support: This work was supported by 2. Ross CA, Poirier MA. Protein aggregation and
Accepted for Publication: September 9, 2016. funding from the American Heart Association grant neurodegenerative disease. Nat Med. 2004;10
12SDG11820014 (Dr Iliff), the Oregon Partnership (suppl):S10-S17.
Published Online: November 28, 2016. for Alzheimer’s Research (Dr Iliff), grants from the
doi:10.1001/jamaneurol.2016.4370 3. Plog BA, Moll KM, Kang H, et al. A novel
Research and Development Office of the technique for morphometric quantification of
Author Contributions: Dr Iliff had full access to all Department of Veterans Affairs (Merit Review subarachnoid hemorrhage-induced microglia
the data in the study and takes responsibility for the Grant, Dr Kaye), and grant NS089709 from the activation. J Neurosci Methods. 2014;229:44-52.
integrity of the data and the accuracy of the data National Institutes of Health (Dr Iliff), including
analysis. Alzheimer’s Disease Center grant AG08017 from 4. Kress BT, Iliff JJ, Xia M, et al. Impairment of
Concept and design: Zeppenfeld, Haswell, Iliff. the National Institute on Aging that supported the paravascular clearance pathways in the aging brain.
Acquisition, analysis, or interpretation of data: All longitudinal follow-up and subsequent brain Ann Neurol. 2014;76(6):845-861.
Authors. autopsies providing the human brain samples used 5. Xie L, Kang H, Xu Q, et al. Sleep drives
Drafting of the manuscript: Zeppenfeld, Haswell, in this study. metabolite clearance from the adult brain. Science.
D'Abreo, Murchison, Grafe, Iliff. Role of the Funder/Sponsor: The funding sources 2013;342(6156):373-377.
Critical revision of the manuscript for important had no role in the design and conduct of the study; 6. Iliff JJ, Lee H, Yu M, et al. Brain-wide pathway for
intellectual content: Zeppenfeld, Simon, Murchison, collection, management, analysis, and waste clearance captured by contrast-enhanced
Quinn, Grafe, Woltjer, Kaye, Iliff. interpretation of the data; and preparation, review, MRI. J Clin Invest. 2013;123(3):1299-1309.
Statistical analysis: D'Abreo, Murchison, Iliff. or approval of the manuscript; and decision to
Administrative, technical, or material support: 7. Iliff JJ, Wang M, Liao Y, et al. A paravascular
submit the manuscript for publication. pathway facilitates CSF flow through the brain
Zeppenfeld, Simon, Grafe, Woltjer, Kaye, Iliff.
Study supervision: Zeppenfeld, Grafe, Iliff. parenchyma and the clearance of interstitial
REFERENCES solutes, including amyloid β. Sci Transl Med. 2012;4
Conflict of Interest Disclosures: Dr Iliff reports 1. Alzheimer’s Association. 2015 Alzheimer’s (147):147ra111.
serving as a consultant for Shire and receiving disease facts and figures. Alzheimers Dement. 2015;
honoraria from Shire Pharmaceuticals and 8. Moftakhar P, Lynch MD, Pomakian JL, Vinters
11(3):332-384. HV. Aquaporin expression in the brains of patients
Genentech. No other disclosures are reported.

98 JAMA Neurology January 2017 Volume 74, Number 1 (Reprinted) jamaneurology.com

Copyright 2017 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by b Wildberries on 02/25/2023


Aquaporin-4 Localization and Cognition in Aging Brains Original Investigation Research

with or without cerebral amyloid angiopathy. 16. Crane JM, Tajima M, Verkman AS. Live-cell accumulation and memory deficits. Mol
J Neuropathol Exp Neurol. 2010;69(12):1201-1209. imaging of aquaporin-4 diffusion and interactions in Neurodegener. 2015;10(1):58.
9. Hoshi A, Yamamoto T, Shimizu K, et al. orthogonal arrays of particles. Neuroscience. 2010; 24. Pérez E, Barrachina M, Rodríguez A, et al.
Characteristics of aquaporin expression 168(4):892-902. Aquaporin expression in the cerebral cortex is
surrounding senile plaques and cerebral amyloid 17. Mawuenyega KG, Sigurdson W, Ovod V, et al. increased at early stages of Alzheimer disease.
angiopathy in Alzheimer disease. J Neuropathol Exp Decreased clearance of CNS beta-amyloid in Brain Res. 2007;1128(1):164-174.
Neurol. 2012;71(8):750-759. Alzheimer’s disease. Science. 2010;330(6012):1774. 25. Yang J, Lunde LK, Nuntagij P, et al. Loss of
10. Wilcock DM, Vitek MP, Colton CA. Vascular 18. Patterson BW, Elbert DL, Mawuenyega KG, astrocyte polarization in the tg-ArcSwe mouse
amyloid alters astrocytic water and potassium et al. Age and amyloid effects on human central model of Alzheimer’s disease. J Alzheimers Dis.
channels in mouse models and humans with nervous system amyloid-beta kinetics. Ann Neurol. 2011;27(4):711-722.
Alzheimer’s disease. Neuroscience. 2009;159(3): 2015;78(3):439-453. 26. Iliff JJ, Goldman SA, Nedergaard M.
1055-1069. 19. Tarasoff-Conway JM, Carare RO, Osorio RS, Implications of the discovery of brain lymphatic
11. Nelson JW, Young JM, Borkar RN, et al. Role of et al. Clearance systems in the brain-implications pathways. Lancet Neurol. 2015;14(10):977-979.
soluble epoxide hydrolase in age-related vascular for Alzheimer disease. Nat Rev Neurol. 2015;11(8): 27. Chételat G, La Joie R, Villain N, et al. Amyloid
cognitive decline. Prostaglandins Other Lipid Mediat. 457-470. imaging in cognitively normal individuals, at-risk
2014;113-115:30-37. 20. Zlokovic BV, Deane R, Sagare AP, Bell RD, populations and preclinical Alzheimer’s disease.
12. Tierney MC, Fisher RH, Lewis AJ, et al. The Winkler EA. Low-density lipoprotein Neuroimage Clin. 2013;2:356-365.
NINCDS-ADRDA Work Group criteria for the clinical receptor-related protein-1: a serial clearance 28. Institute of Medicine (US), Committee on the
diagnosis of probable Alzheimer’s disease: homeostatic mechanism controlling Alzheimer’s Public Health Dimensions of Cognitive Aging, Blazer
a clinicopathologic study of 57 cases. Neurology. amyloid β-peptide elimination from the brain. DG, Yaffe K, Liverman CT, eds. Cognitive Aging:
1988;38(3):359-364. J Neurochem. 2010;115(5):1077-1089. Progress in Understanding and Opportunities For
13. R: A Language and Environment for Statistical 21. Simon MJ, Iliff JJ. Regulation of cerebrospinal Action. Washington, DC: The National Academies
Computing [computer program]. Vienna, Austria: R fluid (CSF) flow in neurodegenerative, Press; 2015.
Foundation for Statistical Computing; 2013. neurovascular and neuroinflammatory disease. 29. Sonnen JA, Santa Cruz K, Hemmy LS, et al.
14. Morris JC. The Clinical Dementia Rating (CDR): Biochim Biophys Acta. 2016;1862(3):442-451. Ecology of the aging human brain. Arch Neurol.
current version and scoring rules. Neurology. 1993; 22. Shibata M, Yamada S, Kumar SR, et al. 2011;68(8):1049-1056.
43(11):2412-2414. Clearance of Alzheimer’s amyloid-ss (1-40) peptide 30. Erten-Lyons D, Woltjer RL, Dodge H, et al.
15. Deramecourt V, Slade JY, Oakley AE, et al. from brain by LDL receptor-related protein-1 at the Factors associated with resistance to dementia
Staging and natural history of cerebrovascular blood-brain barrier. J Clin Invest. 2000;106(12): despite high Alzheimer disease pathology. Neurology.
pathology in dementia. Neurology. 2012;78(14): 1489-1499. 2009;72(4):354-360.
1043-1050. 23. Xu Z, Xiao N, Chen Y, et al. Deletion of
aquaporin-4 in APP/PS1 mice exacerbates brain Aβ

jamaneurology.com (Reprinted) JAMA Neurology January 2017 Volume 74, Number 1 99

Copyright 2017 American Medical Association. All rights reserved.

Downloaded From: https://jamanetwork.com/ by b Wildberries on 02/25/2023

You might also like