MMC 2
MMC 2
MMC 2
Ar ticle
*Correspondence: [email protected]
https://doi.org/10.1016/j.stemcr.2017.10.015
SUMMARY
Alzheimer’s disease (AD) induces memory and cognitive impairment in the absence of motor and sensory deficits during its early and
middle course. A major unresolved question is the basis for this selective neuronal vulnerability. Ab, which plays a central role in AD path-
ogenesis, is generated throughout the brain, yet some regions outside of the limbic and cerebral cortices are relatively spared from Ab
plaque deposition and synapse loss. Here, we examine neurons derived from iPSCs of patients harboring an amyloid precursor protein
mutation to quantify AD-relevant phenotypes following directed differentiation to rostral fates of the brain (vulnerable) and caudal fates
(relatively spared) in AD. We find that both the generation of Ab and the responsiveness of TAU to Ab are affected by neuronal cell type,
with rostral neurons being more sensitive than caudal neurons. Thus, cell-autonomous factors may in part dictate the pattern of selective
regional vulnerability in human neurons in AD.
1868 Stem Cell Reports j Vol. 9 j 1868–1884 j December 12, 2017 j ª 2017 The Authors.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
A B
C D E
F G
H I
40 *
A /total RNA
C99 A 49 A 46 A 43 A 40
30
20
A 48 A 45 A 42 A 38
10
53 56 36 39
0
Endopeptidase Carboxypeptidase-like activity rostral caudal rostral caudal
activity
no fAD mutation fAD
C D
1.5 2.0
*** ***
A 42/40 normalized to
A 38/42 normalized to
*** **** * ** ** * ***
rostral for each line
1.0
0.5
0.5
0.0 0.0
caudal
caudal
caudal
caudal
caudal
caudal
caudal
caudal
rostral
caudal
rostral
caudal
rostral
caudal
rostral
caudal
rostral
caudal
rostral
caudal
rostral
caudal
rostral
caudal
rostral
rostral
rostral
rostral
rostral
rostral
rostral
rostral
YZ1 TZ1 YK26 BR11 BR14 fAD1A fAD2A fAD2B YZ1 TZ1 YK26 BR11 BR14 fAD1A fAD2A fAD2B
E F G
****
A 42/40 normalized to fADcorr
600 3 1.5
* **
A 38
A /total protein
A 42
400 2 1.0
A 40
200 1 0.5
12 12 12 12
12 12
0 0 0.0
fADcorr fAD fADcorr fAD fADcorr fAD
and 38:42 (G) are shown normalized to fADcorr. The ‘‘n’’ for each dataset is shown within the bar graph column. There are four independent
rounds of differentiation represented. For each comparison, a Student’s t test was performed, *p < 0.05, **p < 0.01, ***p < 0.001, ****p <
0.0001. Data are represented as mean ± SEM.
See also Figures S2–S4.
1000
1500
750
0.5 1000
500
500
250
40 38 29 28 40 38 29 28 40 38 29 28
0.0 0 0
rostral caudal rostral caudal rostral caudal rostral caudal rostral caudal rostral caudal
D E F
ADAM10
ADAM19 control control
ADAM9 3000 rostral caudal
APP-all normalized expression
ADAM17/TACE 2.0
APP-all
BACE1
** **** APP-770
BACE2 1.5
PSEN1
APP-KPI
PSEN2
2000
APLP1
1.0 APLP2
NCT
PEN2
0.5
APH1Avar1 fAD rostral fAD caudal
9 10 9 10
10
APH1Avar2 1000
caudal
rostral
caudal
APLP2
control fAD
G H
rostral caudal 200
APP normalized to GAPDH
kDa
***
150
97
(% rostral)
APP (C7)
100
50
MAP2
17 17
191
0
rostral caudal
39 GAPDH
Figure 3. APP Expression Is Elevated in Caudally Directed Neural Cultures Relative to Rostral Cultures
(A–C) Secretion of sAPPa and sAPPb were measured by duplex ELISA. Ratios of sAPPa/sAPPab are shown (normalized to rostral control)
(A), as well as sAPPa normalized to total RNA (B) and sAPPb normalized to total RNA (C). Rostral and caudal cultures are indicated by white
and teal bars, respectively. The ‘‘n’’ for each dataset is shown within the bar graph column. For each comparison, a one-way ANOVA with a
Tukey’s multiple comparisons post-test was performed, **p < 0.01, ***p < 0.001, ****p < 0.0001. Data are represented as mean ± SEM.
(D–F) RNA was harvested from day 100 cultures and analyzed using a custom NanoString CodeSet. Data shown here are derived from three
control lines and two fAD lines, over three differentiations. (D) The heatmap shows the mean expression levels of genes (n = 6–10 per
condition) encoding components of the a-, b-, and g-secretases, for both rostral and caudal wild-type and fAD cultures. (E and F) APP
expression data are shown between rostral and caudal cultures. Data in (E) are normalized to the average rostral data for each iPSC line. A
Student’s t test was performed, **p < 0.01, ****p < 0.0001. Data are represented as mean ± SEM. Heatmap of expression of APP family
members is shown, gene normalized (F). Raw NanoString data were normalized to housekeeping genes.
(G) A representative western blot (WB) from control day 100 total protein lysates.
(H) Quantification of WB data using densitometry from day 100 total protein lysates. For both rostral and caudal, n = 17, derived from four
differentiations. In (H), a Student’s t test was performed, ***p < 0.001. Data are represented as mean ± SEM.
See also Table S1.
(B) Example images of cells in nanowells that secrete high levels of Ab40 and Ab42.
(C) The signal-to-noise ratio (SNR = [foreground median – background median]/[STDEV background]) of fluorescence intensity for both
Ab40 and Ab42 was measured for each well, and plotted. Each dot presents data from wells with single living cells, red dots/regression line
from fAD (n = 10,890 cells), and black dots/regression line (n = 25,603 cells) from control. The difference in the slopes of the lines is
significant, p < 0.0001, DFn = 1, DFd = 25,603, F = 571.
(D and E) Data in (C) re-graphed as scatterplots to compare Ab40 and Ab42 levels separately. While the majority of cells from both control
and fAD do not secrete detectable Ab40 or Ab42, the mean secretion of both is elevated in fAD relative to control. Mann-Whitney test
performed, ****p < 0.001.
(F) Control rostral and caudal neurons were differentiated for >100 days and then dissociated, differentially labeled with either calcein-
violet or calcein-orange, and plated into nanowell arrays at a density that favored a single cell per well. Cells in the nanowells were imaged,
and then Ab40 or Ab42 were co-detected using microengraving.
(G) Example images of cells in nanowells that secrete high levels of Ab40 or Ab42.
(H) SNR of fluorescence intensity for both Ab40 and Ab42 was measured for each well, and a relative ratio of 42:40 calculated. Shown is a
scatterplot of data from wells with single living cells, where each data point reflects data from a single cell. Only wells that had single live
cells and detectable Ab40 and Ab42 are shown. Red lines show means of the ratio for the rostral and caudal populations. Mann-Whitney
test performed, **p < 0.005.
See also Figure S5 and Tables S2 and S3.
* *
total Tau/GAPDH (% control)
125 125
100 100
75 75
50 50
25 9 16 16 16 25 9 16 16 16
0 0
rostral caudal rostral caudal rostral caudal rostral caudal
Ngn2-iN-rostral
C
fADcorr fAD
kDa
Tau
51
pTau
51 (S202)
39 GAPDH
D E
total Tau/GAPDH (% fADcorr)
****
pTau/Total Tau (% fADcorr)
150 200
150
100
100
50
50
17 17 17 17
0 0
fADcorr fAD fADcorr fAD
Ngn2-iN Ngn2-iN
rostral rostral
Figure 5. Rostral versus Caudal Cultures Respond Differentially to the fAD Environment
(A and B) Control and fAD rostral and caudal cultures at 100 days of differentiation were lysed, and total TAU (A) and phospho-TAU (B)
levels were measured by WB and densitometry. The ‘‘n’’ for each dataset is shown within the bar graph column. There are four lines rep-
resented, over four independent rounds of differentiation. One-way ANOVA with a Tukey’s multiple comparisons test performed, *p < 0.05,
****p < 0.0001. Data are represented as mean ± SEM.
(C–E) Data are from day 28 Ngn2-iN cultures. (C) A representative WB from total protein lysates. Various cell densities of 25k (n = 3), 30k
(n = 3), 35k (n = 3), and 40k (n = 1) were plated and analyzed, and protein normalized prior to gel loading. (D and E) Quantification of WB
data using densitometry from total protein lysates. Total TAU (D) and phospho-TAU (E) levels were measured by WB and densitometry. The
‘‘n’’ for each dataset is shown within the bar graph column. There are three independent rounds of differentiation represented. For each
comparison, a Student’s t test was performed, ****p < 0.0001. Data are represented as mean ± SEM.
See also Figure S6.
C D
Figure 6. Rostral versus Caudal Neural Cultures Respond Differentially to the LOAD Environment
At 80–100 days of differentiation, control iPSC-derived rostral and caudal neurons were treated for 2 days with TBS-soluble extract of
sporadic AD postmortem brain, with and without co-administration of a polyclonal Ab-neutralizing antibody (AW7). Cultures were fixed
and immunostained for total and phospho-TAU (A). Images presented here are only for visualization of the cells; relative intensities are
not acquired in a manner that allows for detection of quantitative differences. Scale bars, 100 mm. Parallel rostral and caudal cultures were
lysed and subjected to ELISA (B) and WB (C and D). Lysates were run on a phospho (Thr 231)/total TAU MSD ELISA and phospho-TAU
normalized to total TAU (B). For WBs, quantification by densitometry was performed and phospho-TAU was normalized to total TAU (C) or
else total TAU was normalized to MAP2 (D). The ‘‘n’’ for each dataset is shown within the bar graph column. For each comparison, a one-way
ANOVA with a Tukey’s multiple comparisons post-test was performed, *p < 0.05, ***p < 0.001. ns, not significant. Data are represented as
mean ± SEM, over four independent rounds of differentiation.
using in vivo microdialysis in mutant human APP trans- neurons show similar levels of spontaneous activity
genic mice, different regions of the brain showed different (Table S3). However, the precise excitatory/inhibitory bal-
levels of soluble human Ab, and higher levels of Ab were ance of activity may be different between culture types,
correlated with elevated neuronal activity (Bero et al., and future studies are warranted to more deeply interrogate
2011). Using MEAs, we find that both rostral and caudal this question.
Supplemental Information
1.0
1.5
1.0
0.5
0.5
53 56 36 39 53 56 36 39
0.0 0.0
rostral caudal rostral caudal rostral caudal rostral caudal
Figure S2. Aβ42/40 and Aβ38/42 ratio in pooled control and fAD lines. Related to Figure 2.
The data shown here is the same data from Fig. 2C and 2D, but pooled and normalized in a manner that allows the
fAD to control differences to be apparent. The Aβ42/40 ratio is shown in rostral and caudal cultures (A) and the
Aβ38/42 ratio is shown in rostral and caudal cultures (B). The ‘n’ for each line is shown in the corresponding bar.
Both ratios are normalized to the rostral control group and were calculated by paired experiments of control and
fAD samples. For this reason, BR11 and BR14 lines are not included here, since they were not paired with fAD
cultures. Rostral cultures are shown in white and caudal cultures are shown in teal. For each comparison a one-way
ANOVA with a Tukey’s multiple comparisons post-test was performed, *p<0.05, ***p<0.001, ****p<0.0001. Error
bars represent mean SEM. Nine independent rounds of differentiation are represented.
Figure S3. Forced expression of APPV717I in control rostral and caudal cultures corroborates findings with
endogenous APPV717I mutation. Related to Figure 2.
Following an embryoid aggregate differentiation, control rostral and caudal neuronal cultures were transduced with
an APPV717I virus or mock transduced at day 40 and then harvested at day 47 (A-E) or else at day 100 (F).
Conditioned media from day 40 rostral and caudal cultures were analyzed for Aβ38, 40, and 42, measured by Aβ
triplex ELISA. (A) Levels of Aβ38, 40, and 42 are shown normalized to total protein. The ratios of Aβ 42:40 (B)
and 38:42 (C) are shown. The ‘n’ for each rostral/caudal pair is listed as the following: no virus – 4/4; APPV717I –
3/3. Rostral and caudal cultures are indicated by white and teal bars, respectively. For each comparison a one-way
ANOVA with a Tukey’s multiple comparisons post-test was performed, * p<0.05, ** p<0.01, **** p<0.0001. Data
are represented as mean SEM. (D) WB for day 47 samples showing the overexpression of APP with APPV717I
lentiviral transduction, as well as levels of Tau and GAPDH. (E,F) Lysates from d47 (E) or d100 (F) were run on a
Phospho (Thr 231)/Total Tau MSD ELISA and phospho-Tau normalized to total Tau. The ‘n’ for each rostral/caudal
pair is listed as the following: no virus – 4/4; APPV717I – 3/3. For each comparison, a one-way ANOVA with
Tukey’s multiple comparison’s test was performed, * p<0.05. Data are represented as mean SEM.
Figure S4. Characterization of APPV717I (fAD) and its isogenic control (fADcorr), in NGN2-induced neuron
(iN) cultures. Related to Figure 2.
(A) Schematic outlining the targeting constructs for correcting the APPV717I mutation in iPSC line fAD 2B. The
mutation in codon 717 of APP is flanked by the gRNA targeting sites, denoted by two red arrows in exon 17. The
mutation was corrected from an isoleucine to a valine, using a CRISPR/Cas technology. Two corrected clones were
isolated and used to generate the data here. Following confirmation of correct targeting, the puromycin selection
cassette was removed following Cre-mediated excision (loxp sites represented by black arrowheads). TK=
thymidine kinase selection cassette, DTA=diphtheria toxin selection cassette. The “fAD” iPSC lines used in these
experiments are two clones of non-targeted iPSCs that were isolated in parallel to the two corrected lines. (B) fAD
and its isogenic control (fADcorr) iPSC lines were transduced with the Ngn2 virus and then differentiated using a
protocol from the Sudhof lab (Zhang et al., 2013). Scale bars = 100m. (C) qPCR for neuronal markers APP, MAPT
and synaptophysin (SYP). All targets are normalized to GAPDH and are expressed in arbitrary units (A.U.). n=3 for
each group. For each comparison a Student’s t-test was performed with no significant differences identified. Data
are represented as mean SEM. (D) Conditioned media from both fAD and fADcorr iNs were was analyzed for
Aβ38, 40, and 42 by Aβ triplex ELISA at d21 of differentiation. Levels of Aβ38, 40, and 42 are shown normalized
to total protein. Data shown are the same data from Fig. 2E, graphed separately. n=12 for each group over four
differentiations. For each comparison a Student’s t-test was performed, ** p<0.01. Data are represented as mean
SEM.
A A ND
80 40 High A40
within each group
% of total cells
60 30
40 20
20 10
* * ND * ND
0
* 0
CUX1 SATB1 FEZF2 FOXG1 RELN TBR1 HOXB13 HOXB4 HOXB6 ISL1 MNX1
cortical/forebrain cerebellar/spinal cord
100 30 80
within each group
% of total cells
% of total cells
% of total cells
80
60
20
60
40
40
10
20
20
0 0
* * 0 * *
DLG4 SYN1 MAPT SLC17A7 GRIN1 GRIK1 SLC32A1 GAD1 NKX2-1 CALB2
general neuronal excitatory inhibitory
B C
2.5 2.0
*** ***
****
2.0
1.5
*
Relative expression
Relative expression
1.5
**** ****
** ***
1.0
1.0
0.5
0.5
0.0 0.0
Cb
Cb
Cb
Cb
Temp Cx
Temp Cx
Temp Cx
Temp Cx
caudal
caudal
caudal
caudal
rostral
rostral
rostral
rostral
Figure S5. Single-cell NanoString analysis of cells following microengraving and isolation from the nanowells
and expression of excitatory and inhibitory markers in iPSC-derived cultures and post-mortem tissue.
Related to Figure 4 and Table S2.
(A) Following printing (Fig. 4), individual cells were stained with trypan blue to mark cells with intact membranes,
retrieved from nanowells, lysed, cDNA synthesized and amplified using multi-target amplification, and hybridized
to a custom NanoString codeset. Cells expressing high levels of Aβ42 (n=10), Aβ40 (n=21), and no detectable Aβ40
or 42 (n=16) were analyzed. For each cell, genes with values above 1000 counts were considered to be expressed.
Values shown are the percentage of total cells within each Aβ population expressing each listed gene. ND=Not
detectable (plain), high Aβ40 (diagonal lines) and high Aβ42 (checkered). Data is presented in Table format in
Supplemental Table S2. (B) NanoString analysis of genes expressed in excitatory and inhibitory neurons is shown
comparing rostral and caudal cultures. RNA was harvested from day 40 cultures and analyzed using a custom
NanoString codeset. Data shown here include three control lines and three fAD lines. n=10 for each group. Caudal
data were normalized to the average rostral data for each gene. (C) Publically available RNA-seq analyses (Mayo
Clinic Brain Bank) from post-mortem human temporal cortex (Temp Cx) and cerebellum (Cb) is shown for
excitatory and inhibitory markers. n=275 and 276 for Temp Cx and Cb, respectively. For each gene, a two-tailed t-
test was performed, *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. Error bars represent SEM.
150
125
100
75
50
25 4 6 6 6
0
rostral caudal rostral caudal
control fAD
Figure S6. Total Tau normalized to MAP2 to correct for relative neuron/non-neuronal cell proportions.
Related to Figure 5.
Control and fAD rostral and caudal neurons at 100 days of differentiation were lysed, and total Tau levels were
measured by Western blotting and densitometry. Here, quantification by densitometry was normalized to MAP2.
The ‘n’ for each data set is shown within the bar-graph column. There are two lines represented for both control and
fAD lines, over three independent rounds of differentiation. For each comparison a Student’s t-test was performed, *
p<0.05. Data are represented as mean SEM.
WT rostral WT rostral WT rostral WT rostral WT rostral WT rostral WT caudal WT caudal WT caudal WT caudal
APLP1 6990 6123 6842 6815 2412 3317 5794 5747 5957 5803
APLP2 7644 7198 6665 7497 2812 3550 12067 10096 10609 10049
ADAM10 833 909 787 905 681 911 788 788 798 834
ADAM19 66 102 88 168 68 104 110 91 142 116
ADAM9 1443 1311 1359 1284 1252 1828 2932 3011 2873 3105
TACE/ADA
M17 724 612 562 621 410 443 969 942 912 886
BACE1 773 747 804 852 855 1190 967 940 1015 910
BACE2 150 163 154 152 303 431 186 225 186 226
PSEN1 1201 1035 1022 1034 325 517 1093 971 1014 953
PSEN2 172 162 208 238 70 75 169 210 240 190
Nicastrin 834 787 672 782 971 1434 1022 894 952 916
PEN-2 574 654 704 770 1071 1376 880 713 867 880
aph1Avar1 597 616 494 631 941 1026 671 723 565 640
aph1Avar2 727 610 679 598 788 857 787 674 766 778
aph1Avar3 339 308 303 338 173 242 337 295 340 329
aph1Avar4 246 198 194 134 503 615 168 158 177 168
aph1Bvar1 266 267 262 252 384 455 289 250 263 270
aph1Bvar2 5 6 14 7 25 32 5 2 6 2
WT rostral WT rostral WT rostral WT rostral WT rostral WT rostral WT caudal WT caudal WT caudal WT caudal
B2M 4892 5045 4231 6441 20205 17611 15008 12549 12917 11895
GAPDH 38068 37180 39924 36092 40270 43218 33964 35895 36788 37155
Supplemental Tables
Table S1. Individual values from the NanoString data set shown in Fig. 3D. Related to Figure 3. Counts are
normalized across multiple code sets manually by using the mean of the housekeeping genes (shown in red text).
7193 6697 6280 6141 6197 5371 3849 15495 16381 15089
16470 15086 15245 15076 15401 5191 3545 4296 3158 3686
1027 876 910 860 921 701 580 984 767 859
179 133 148 143 152 57 62 37 17 26
4721 5177 5363 5318 5591 3047 2500 1265 942 1208
1128 1081 1222 1084 1113 743 521 1876 1765 1897
1356 1490 1552 1416 1446 2245 2001 2093 1632 1864
1 1 1 1 1 24 21 244 127 128
1277 1237 1245 1213 1302 843 701 2743 2607 2647
259 267 289 271 299 149 99 342 315 316
1591 1404 1379 1427 1386 1663 1096 2426 2472 2589
1097 1088 1144 1159 1245 1154 1074 2396 3134 2649
1043 898 879 844 876 596 521 2162 2819 2418
938 901 965 960 964 840 709 1357 1041 952
460 480 479 440 440 170 196 590 733 573
187 176 189 187 213 607 631 1417 1539 1291
385 324 351 319 340 361 338 929 757 821
16 1 12 1 10 29 70 178 112 137
fAD caudal fAD caudal fAD caudal fAD caudal fAD caudal fAD caudal fAD caudal fAD caudal fAD caudal fAD caudal
22098 22549 22152 22230 22349 17281 23752 5276 5809 6160
31011 32042 31821 32164 32455 48474 42952 42752 42845 44772
204 163 168 171 189 43 56 125 111 95
1216 1346 1358 1356 1391 1144 1141 1354 1915 1676
5997 5858 6125 5820 5254 6919 5189 10299 6743 7645
1599 1554 1602 1516 1536 1129 572 2649 2972 3418
1062 944 1013 952 989 1266 1093 3742 3187 2804
16812 15544 15761 15791 15836 3745 5245 13803 16419 13431
A ND High A40 High A42
CORTICAL/FOREBRAIN
CUX1 67% 71% 30%
SATB1 40% 62% 30%
FEZF2 20% 29% 40%
FOXG1 7% 19% ND
RELN 33% 48% 10%
TBR1 27% 24% 40%
CEREBELLAR/SPINAL CORD
HOXB13 7% 14% ND
HOXB4 20% 29% ND
HOXB6 ND ND ND
ISL1 7% 10% ND
MNX1 ND ND ND
EXCITATORY
SLC17A7 7% 5% 10%
GRIN1 ND 10% ND
GRIK1 27% 14% 20%
INHIBITORY
SLC32A1 13% 38% 30%
GAD1 33% 62% 40%
NKX2-1 33% 14% ND
CALB2 7% 19% ND
GENERAL NEURONAL
DLG4 73% 95% 70%
SYN1 27% 81% 40%
MAPT 47% 57% 70%
Table S2. Percentage of single-cells within each group expressing various NanoString markers. Related to
Figure 4 and Figure S5. ND = not detected.
Spike properties
WT rostral WT caudal
n n
Mean 95%CI/SEM (wells) Mean 95%CI/SEM (wells)
Firing
0.10 0.02-0.16 19 0.12 0.06-0.19 21
frequency (Hz)
Burst count
84.5 40.9 19 113.8 43.1 21
per well
Spike properties
fAD rostral fAD caudal
n n
Mean 95%CI/SEM (wells) Mean 95%CI/SEM (wells)
Firing
0.14 0.07-0.20 29 0.39 0.11-0.68 42
frequency (Hz)
Burst count
30.9 13.7 29 277.6 117.1 42
per well
Table S3. Multi-electrode array data of rostral and caudal cultures reveals no detectable differences in
spontaneous activity between rostral and caudal cultures. Related to Figure 4.
Spike frequency and spike voltage waveform parameters derived from spontaneous firing of cultures. Metrics
calculated from integration of 10, 30 min recordings of cultures performed over a 20 day period (d37 to d57). Firing
frequency (Hz) determined by total number of spike events per second of recording; burst count represents total
number of ‘bursts’ determined by poisson surprise algorithm (Legéndy and Salcman, 1985). n represents replicate
wells (8 electrodes per well) of each culture. Waveform properties determined from reconstruction of momentary
voltage recordings sampled at 12.5kHz, followed by unsupervised clustering of waveform shapes of all spike events
recorded by an individual electrode. Statistics calculated from the waveforms representing the centroid of each
cluster. n represents individual clusters identified. Individual cluster sizes range from 10 - 6228 spike events.
Supplemental Experimental Procedures:
Study Design
The primary research objective of this study was to differentiate control and fAD iPSC lines to rostral and caudal
neural cultures and examine AD-relevant phenotypes such as APP processing and Tau alteration, in order to study
the differential vulnerability observed in AD subjects. Sample size was selected based on availability of lines and
differentiation capacity and efficiency (Fig. S1). Typically, 10 wells of a 96 well-plate, for both rostral and caudal
neurons, were plated per differentiation experiment. Multiple differentiations (as noted in figure legends, over 100
across the entire study) were conducted in order to assess 6 control and 3 fAD lines. Samples were excluded based
on the metrics outlined in Fig. S1. At 40 days of differentiation, qPCR was performed in order to confirm expression
of neuronal markers on a well-to-well basis. Samples that met the selection criteria were then subjected to a second
metric of either rostral or caudal marker expression. Only samples that met these selection criteria were included in
subsequent analyses. Replicates, experimental ‘n’ and iPSC lines used are noted in the figure legends.
Cell Lines
fAD iPSCs were generated in collaboration with the Harvard Stem Cell Institute as previously reported (Muratore et
al., 2014a). For WT lines the following previously published and commercially available lines were differentiated:
HDFa-YK26 (Martins-Taylor et al., 2011) and IMR90-TZ1 and IMR90-YZ1 (Zeng et al., 2010) (UCONN Stem
Cell Core). BR01, BR11 and BR14 were generated and characterized in collaboration with the New York Stem Cell
Foundation (NYSCF) using previously described methods (Paull et al., 2015). BR01, BR11 and BR14 were all
derived from Caucasian female donors, BR01 was not cognitively impaired at death at age 89, BR11 was not
cognitively impaired at death at age 79, BR14 had a diagnosis of late-onset AD at death at age 90.
Medias
MEF Medium- 435 mL DMEM (Invitrogen), 5 mL 100x Penicillin/Streptomycin (Invitrogen), 5 mL 100x L-
glutamine (Invitrogen), 50 mL FBS (Invitrogen)
iPSC Medium- 390 mL DMEM/F12 (Invitrogen), 100 mL KOSR (Invitrogen), 5 mL 100x
Penicillin/Streptomycin/Glutamine (Invitrogen), 5 mL 100x MEM-NEAA (Invitrogen), 50 μM b-mercaptoethanol
(Invitrogen), with the addition of fresh FGF2 (Millipore, 10 ng/mL) to the medium
N2 Neural Induction Medium- 490 mL DMEM/F12, 5 mL N2 supplement (Invitrogen), 5 mL 100x MEM-NEAA,
and Heparin (Sigma-Aldrich, 2 μg/mL)
N2/B27 Neural Induction Medium- 480 mL DMEM/F12, 5 mL N2 supplement, 10 mL B27 supplement
(Invitrogen), 5 mL MEM-NEAA and 2 μg/ml Heparin (Sigma-Aldrich), with the addition of fresh cAMP (1 μM)
(Sigma) and IGF1 (PeproTech, 10 ng/mL) to the medium
Neural Differentiation Medium- 490 mL Neurobasal medium (Invitrogen), 5 mL N2 supplement (Invitrogen), 5 mL
100x MEM-NEAA, and 10 mL B27 supplement (Invitrogen), with the addition of fresh cAMP (1 μM), BDNF,
GDNF, and IGF-1 (all 10 ng/mL) to the medium.
Human KSR media (day 1 and 2)- 415 mL KO DMEM (Invitrogen), 75 mL KOSR, 5 mL, MEM-NEAA, 0.5 mL
beta-mercaptoethanol (Invitrogen), 5 mL Glutamax (Gibco)
N2 media (day 2)- 500 mL DMEM/F12, 5mL Glutamax, 7.5mL 20% Dextrose (Stem Cell Technologies), 5 mL
N2B supplement (Stem Cell Technologies)
N2 + B27 media (day 3)- N2 media + 1:100 B27 (50X) (B27), B27 should be added just prior to use
Neurobasal Medium (day 4) - 485 mL NBM (Invitrogen), 5 mL Glutamax, 7.5 mL 20% Dextrose, 2.5mL MEM-
NEAA, B27 should be added just prior to use, with the addition of fresh BDNF, GDNF, and IGF-1 (all 10 ng/mL) to
the medium.
Western Blots
Lysates were prepared with standard lysis buffer containing 1% NP40, 0.5M EDTA, 5M NaCl, 1M Tris and
cOmplete protease inhibitors and phosSTOP (Roche). Lysates were protein normalized, and samples were loaded
onto 4–12% Bis-Tris gels using MES or MOPS running buffer (Invitrogen), transferred to nitrocellulose
membranes, and probed for various proteins using standard WB methods. The resultant blots were detected with
either an infrared imaging system (Odyssey; LI-COR Biosciences) or by ECL and exposure to film.
Antibodies
Antibody Vendor Concentration
APP (C7) Selkoe Lab 1:1000
EN1 DSHB (#4G11) 1ug/mL
GAPDH Millipore (MAB374) 1ug/mL
GFAP abcam (#ab4674) 33ug/mL
HoxB1 R&D Systems (#AF6318) 5ug/mL
PCRP-HOXB2 DSHB (#1C9) 5ug/mL
Anti-HOXB4 DSHB (#I12) 5ug/mL
PCRP-HOXB6 DSHB (#1A4) 5ug/mL
PCRP-HOXB9 DSHB (#1E6) 5ug/mL
MAP2 abcam (#ab5392) 1.9ug/mL
NeuN (A60) Millipore (#MAB377) 1ug/mL
Olig2 (C-17) Santa Cruz (#sc-19969) 4ug/mL
pTau (AT8) Thermo Fisher Scientific/Pierce (#MN1020) ICC: 5ug/mL, WB: 20ug/mL
Tau (K9JA) DAKO (#A-0024) 55ug/mL
Tbr1 abcam (#ab31940) 5ug/mL
TuJ1 Sigma (#T2200) 1ug/mL
RNA Analyses
RNA Sequencing: Total RNA from rostral and caudal cultures (4 per condition) were assayed for quality on an
Agilent TapeStation 4200. Samples with an electronic RNA Integrity Number (eRIN) of at least 8.0 were polyA
selected using oligo-dT Dynabeads (Thermo Fisher). Double stranded cDNA was synthesized using the SuperScript
III reverse transcriptase protocol using random hexamers on the polyA-RNA eluate. After purifying the cDNA with
AMPure magnetic beads (Agencourt) and quantitation on the TapeStation, libraries were generated by processing
1ng of the double stranded cDNA product through the Illumina Nextera Tagmentation library protocol to fragment
the DNA and add overhangs for barcode (i5 and i7) annealing. Each of the 8 amplified and barcoded libraries was
quality controlled for concentration and integrity (Agilent TapeStation), normalized to 1ng/ul and combined in equal
concentrations into a single 8x multiplexed library. Multiplexed libraries were sequenced on an Illumina NextSeq
500 to a depth of at 433 million paired-end reads (75 bases per read) total. RNAseq reads were run through an
analysis pipeline that included the following steps: fastQC (v0.10.1) to assay the quality of reads; trimmomatic
(v0.33) to remove end bases with phred33 quality scores below 25 and to eliminate resulting reads that are shorter
than 30 bases (Bolger et al., 2014); kallisto (v0.42.4) for pseudoalignment and quantification program (Bray et al.,
2016) running 100 bootstraps against a kallisto index generated from ChGR38p10 with a k-mer length of 31. Hox
gene-level expression data (Transcripts Per Kilobase Million, TPM) were extracted from the expression table and
the mean plotted as a heat map. Quantitative PCR (qPCR): Each sample was assayed in 3 technical replicates. Data
were analyzed using the ΔΔCT method and expression was normalized to GAPDH expression (Livak and
Schmittgen, 2001). Primer efficiency was calculated for each pair of primers and the slope of the dilution line was
found to be within the appropriate range. Dissociation curves also showed single peak traces, indicating template-
specific products. NanoString: Hybridization reactions were carried out with 100-200 ng RNA for pooled culture
samples (single cell data was derived from amplified cDNA). Post-hybridization, samples were processed with the
nCounter Prep-station. Following run completion, the cartridge was scanned using the nCounter Digital Analyzer, at
max resolution (~1000 images/sample). Data were analyzed using the nSolver Analysis Software as follows: 1)
background subtracted by negative control subtraction (mean of negative controls + 2 STDEVs), 2) normalized to
mean of positive controls and 3) normalized to a set of 8 house-keeping genes (B2M, GAPDH, GUSB, HPRT1,
POLR2A, RPL13a, RPL27 and MAP2) or to the total gene set, as noted.
Microengraving/ Printing
The nanowell array is fabricated on a biocompatible PDMS (polydimethylsioxane) affixed to a glass microscope
slide. Each array has 84,672 wells with 50x50x50µm dimensions. Prior to cell plating, PDMS arrays were incubated
in a vaccuum plasma cleaner at a high radio frequency for 2 min in order to make the surface hydrophilic for later
cell plating, and then placed into PBS. Aged differentiated control rostral and caudal neurons were dissociated with
accutase and then stained with live markers-calcein violet or calcein orange, respectively. Cells were quickly spun
down to wash out the extra live maker before mixing together and loading on PDMS. Cells are loaded from
suspension (1-2x105 cells/ml) by gravity, which favors 0-2 cells per well (examined in real-time under a
microscope). Non-adherent cells are removed through subsequent washing. The cells on PDMS were imaged using
an automated inverted epifluorescence microscope (Zeiss). Cell viability and numbers per well were analyzed with
Enumerator software (custom software, available upon request to JCL (Han et al., 2010; Ogunniyi et al., 2009)).
Capture antibody (6E10) (35 μg/ml) and human IgG (15 ug/ml) were coated onto a poly-lysine coated glass slide at
room temperature for 1 hr, and blocked with 5% BSA in PBS for 30 minutes. Before printing, the glass slide is
briefly incubated with human serum. Detection of human serum bound to human IgG allows for software alignment
of wells. The coated slide is used to seal the nanowell array plated with cells (clamped between plates using a
microarray hybridization chamber). The sealed PDMS array with antibody-coated slide was incubated at 37C for
overnight, followed by removal of the glass slide. Captured Aβ40 and Aβ42 on the glass slide were detected by
incubation with HRP-conjugated 2G3 antibody and Biotin-conjugated HJ7.4 antibody, respectively for 1 hr at RT,
followed by a fluorescently-labeled secondary antibody at RT for 1 hr. Fluorescent signals are detected using a
Genepix Pro slide scanner and aligned with Crossword (Gierahn et al., 2014). Secretion data from Crossword and
cell information from Enumerator are combined according the unique ID for each well. Wells were deemed analyte-
positive if the signal was one standard deviation above the mean and the intensity of >30% of the pixels within the
well area was greater than two standard deviations above the local mean background intensity.
AD TBS treatments
AD brain was obtained and used in accordance with the Partners IRB assurance. Informed consent was obtained
from the subject. The AD brain sample was from an 82-year-old male with numerous amyloid-beta plaques and
neurofibrillary tangles in multiple brain regions (cortex, hippocampus and amygdala). The subject also had moderate
brain atrophy in the frontal and parietal lobes. Brain homogenate was prepared as specified in (Shankar et al., 2008).
Frozen cerebral cortices were provided by Dr. M. Frosch (Massachusetts ADRC Neuropathology Core, MGH,
Boston, MA, USA) under institutional review board-approved protocols. Frozen samples of temporal cortex (1 g)
were allowed to thaw on ice, chopped into small pieces with a razor blade, and then homogenized with 25 strokes of
a Dounce homogenizer (Fisher) in 4 mL ice-cold 20 mM Tris HCl (pH 7.4) containing 150 mM NaCl and protease
inhibitors [called Tris-buffered saline (TBS)]. TBS-soluble Aβ was separated from membrane-bound and plaque Aβ
by centrifuging the homogenates at 175,000 g at 4°C in a TLA 100 rotor (Beckman Coulter) for 30 min, and the
supernatant (referred to as the TBS extract) was aliquoted and stored at −80°C.
Lentivirus Preparation
The coding sequences for mRFP and APPV717I were introduced into the pcDH-CB vector by Infusion cloning
(Clontech). HEK293FT cells were co-transfected with psPax2, pMD2.G, and pcDH-CB plasmids using
Lipofectamine 2000 (Invitrogen). Lentiviral conditioned media was harvested 24 and 36 hours post-transfection,
pooled, and concentrated 100-fold by centrifugation over 20% sucrose at 100,000 x g for 2 hours followed by
resuspension in fresh media. pCDH-CB (Addgene plasmid # 72267) was a gift from Kazuhiro Oka, and psPAX2
(Addgene plasmid # 12260) and pMD2.G (Addgene plasmid #12259) were gifts from Didier Trono.
MEA Recordings
Spontaneous firing is measured using a multi-well MEA recorder (Maestro, Axion Biosystems) with 768 electrodes
(8 electrodes per well in a 96 well plate). All recordings were performed for 30min under environmentally
controlled conditions (37°C and 5% CO2). Potentials are recorded at a frequency of 12.5kHz with ‘spikes’ called
every time the voltage exceeds 5.5 standard deviations away from the root mean squared (RMS) of the background
potential calculated over a 10 ms moving window (“crossing-threshold”). Momentary voltage recordings are saved
for a 3 ms interval around each spiking event, as well as the value of the crossing-threshold at the time of the spike.
The total number of spiking events detected across all electrodes in the well over the duration of recording is used to
calculate the firing frequency for each well.
Here, iPSC-derived rostral and caudal cultures were plated on 96-well MEA plates at day 28 of
differentiation at a density of 500,000/well. Recordings were acquired every 2-3 days from d37 to d57. Data from all
recordings were integrated. Recording data was filtered in order to remove spike events detected by ‘high-noise’
electrodes, RMS voltage >= 3 µV at time of detection, and additionally data from wells failing to meet a minimum
activity level of 0.01Hz during a given recording were also excluded. Mean firing rate (MFR, Hz) was calculated as
the number of spikes per total duration of recording (in seconds). Bursting data was extracted from Axion software.
In order to reconstruct voltage waveforms, momentary voltage measurements were extracted from recording data
using MATLAB functions (MATLAB, The MathWorks Inc) provided by Axion Biosystems, while custom scripts in
the R programming language (R Core Team, 2017) were developed to calculate a series of waveform parameters
and to perform shape-based clustering analysis. Shape-based clustering analysis was performed on all spike events
detected by a given electrode. Thus, each electrode of the arrays may have multiple clusters assigned to it, each of
which may represent a different neuron spatially adjacent to the electrode. Shape-based clustering consisted of first
performing principal components analysis on the calculated waveform parameters followed by an unsupervised
mean-shift clustering (Ciollaro, Mattia and Wang, Daren, 2016) of spike events projected along the first two
principle components. Minor clusters containing few than 10 spike events were excluded from further analysis.
Waveform metrics of individual clusters were reported for the spike events representing the centroid of each cluster.
Metrics calculated from clustered waveform data include: amplitude form maximum hyperpolarization to maximum
depolarization (µV), Peak-Valley Interval (PVI), the time (µs) between the maximum and minimum potential (µV),
and Non-Linear Energy (NLE) of the waveform, a parameter that measure the ‘sharpness’ of waveform (Kim and
Kim, 2000)
Supplemental References
Bolger, A.M., Lohse, M., and Usadel, B. (2014). Trimmomatic: a flexible trimmer for Illumina sequence data.
Bioinformatics 30, 2114–2120.
Bray, N.L., Pimentel, H., Melsted, P., and Pachter, L. (2016). Near-optimal probabilistic RNA-seq quantification.
Nat. Biotechnol. 34, 525–527.
Ciollaro, Mattia, and Wang, Daren (2016). MeanShift: Clustering via the Mean Shift Algorithm. R package version
1.1-1. https://CRAN.R-project.org/package=MeanShift.
Gierahn, T.M., Loginov, D., and Love, J.C. (2014). Crossword: a fully automated algorithm for the segmentation
and quality control of protein microarray images. J. Proteome Res. 13, 362–371.
Han, Q., Bradshaw, E.M., Nilsson, B., Hafler, D.A., and Love, J.C. (2010). Multidimensional analysis of the
frequencies and rates of cytokine secretion from single cells by quantitative microengraving. Lab. Chip 10, 1391–
1400.
Hu, B.-Y., and Zhang, S.-C. (2009). Differentiation of spinal motor neurons from pluripotent human stem cells. Nat.
Protoc. 4, 1295–1304.
Kim, K.H., and Kim, S.J. (2000). Neural spike sorting under nearly 0-dB signal-to-noise ratio using nonlinear
energy operator and artificial neural-network classifier. IEEE Trans. Biomed. Eng. 47, 1406–1411.
Legéndy, C.R., and Salcman, M. (1985). Bursts and recurrences of bursts in the spike trains of spontaneously active
striate cortex neurons. J. Neurophysiol. 53, 926–939.
Livak, K.J., and Schmittgen, T.D. (2001). Analysis of relative gene expression data using real-time quantitative PCR
and the 2(-Delta Delta C(T)) Method. Methods 25, 402–408.
Maherali, N., Ahfeldt, T., Rigamonti, A., Utikal, J., Cowan, C., and Hochedlinger, K. (2008). A high-efficiency
system for the generation and study of human induced pluripotent stem cells. Cell Stem Cell 3, 340–345.
Martins-Taylor, K., Nisler, B.S., Taapken, S.M., Compton, T., Crandall, L., Montgomery, K.D., Lalande, M., and
Xu, R.-H. (2011). Recurrent copy number variations in human induced pluripotent stem cells. Nat. Biotechnol. 29,
488–491.
Muratore, C.R., Rice, H.C., Srikanth, P., Callahan, D.G., Shin, T., Benjamin, L.N.P., Walsh, D.M., Selkoe, D.J., and
Young-Pearse, T.L. (2014a). The familial Alzheimer’s disease APPV717I mutation alters APP processing and Tau
expression in iPSC-derived neurons. Hum. Mol. Genet. 13, 3523-3536.
Muratore, C.R., Srikanth, P., Callahan, D.G., and Young-Pearse, T.L. (2014b). Comparison and Optimization of
hiPSC Forebrain Cortical Differentiation Protocols. PLoS ONE 9, e105807.
Ogunniyi, A.O., Story, C.M., Papa, E., Guillen, E., and Love, J.C. (2009). Screening individual hybridomas by
microengraving to discover monoclonal antibodies. Nat. Protoc. 4, 767–782.
Paull, D., Sevilla, A., Zhou, H., Hahn, A.K., Kim, H., Napolitano, C., Tsankov, A., Shang, L., Krumholz, K.,
Jagadeesan, P., et al. (2015). Automated, high-throughput derivation, characterization and differentiation of induced
pluripotent stem cells. Nat. Methods 12, 885–892.
R Core Team (2017). R: A language and environment for statistical computing. R Foundation for Statistical
Computing, Vienna, Austria. https://www.R-project.org/.
Shankar, G.M., Li, S., Mehta, T.H., Garcia-Munoz, A., Shepardson, N.E., Smith, I., Brett, F.M., Farrell, M.A.,
Rowan, M.J., Lemere, C.A., et al. (2008). Amyloid β-Protein Dimers Isolated Directly from Alzheimer Brains
Impair Synaptic Plasticity and Memory. Nat. Med. 14, 837–842.
Vierbuchen, T., Ostermeier, A., Pang, Z.P., Kokubu, Y., Sudhof, T.C., and Wernig, M. (2010). Direct conversion of
fibroblasts to functional neurons by defined factors. Nature 463, 1035–1041.
Zeng, H., Guo, M., Martins-Taylor, K., Wang, X., Zhang, Z., Park, J.W., Zhan, S., Kronenberg, M.S., Lichtler, A.,
Liu, H.-X., et al. (2010). Specification of Region-Specific Neurons Including Forebrain Glutamatergic Neurons from
Human Induced Pluripotent Stem Cells. PLoS ONE 5, e11853.
Zhang, Y., Pak, C., Han, Y., Ahlenius, H., Zhang, Z., Chanda, S., Marro, S., Patzke, C., Acuna, C., Covy, J., et al.
(2013). Rapid Single-Step Induction of Functional Neurons from Human Pluripotent Stem Cells. Neuron 78, 785–
798.