SUMMARY
Predisposition to sporadic Alzheimer’s disease (SAD) involves interactions between a person’s unique combination of genetic variants and the environment. The molecular effect of these variants may be subtle and difficult to analyze with standard in vitro or in vivo models. Here we used hIPSCs to examine genetic variation in the SORL1 gene and possible contributions to SAD-related phenotypes in human neurons. We found that human neurons carrying SORL1 variants associated with an increased SAD risk show a reduced response to treatment with BDNF, at the level of both SORL1 expression and APP processing. shRNA knockdown of SORL1 demonstrates that the differences in BDNF-induced APP processing between genotypes are dependent on SORL1 expression. We propose that the variation in SORL1 expression induction by BDNF is modulated by common genetic variants and can explain how genetic variation in this one locus can contribute to an individual’s risk of developing SAD.
INTRODUCTION
Late-onset, sporadic Alzheimer’s disease (SAD) is the most common cause of neurodegeneration in the elderly. SAD is a human disorder in which individual patients might develop disease differently because of a poorly understood collaboration of multiple genetic risk factors and the environment. In fact, although no genes have been identified that cause SAD, as much as 60%–80% of risk for developing SAD may be genetic (Gatz et al., 2006). Although hundreds of genetic associations with SAD have been reported (Bertram et al., 2007), many of these reported associations are based on small samples or have not been replicated independently, and large-scale meta-analyses pooling multiple genome-wide association studies (GWASs) found only 20 or so loci to be associated consistently with the risk of developing SAD (Bertram et al., 2007; Chouraki and Seshadri, 2014). Although many associated genes are seemingly random, it is possible to group some of the top hits into genes involved in specific cellular pathways, such as endocytosis, cholesterol metabolism, and immune function (Olgiati et al., 2011). At present, many attempts to measure phenotypes attributed to SAD-associated genes have used post-mortem material because of the inaccessibility of brain tissue in living subjects (Cruchaga et al., 2014; Griciuc et al., 2013; Holton et al., 2013; Karch et al., 2012). These studies have yielded mixed results, perhaps because the effects on individual cells are masked in tissues composed of multiple cell types. It has also been suggested that different variants may control gene response to damage or to normal signaling in the brain rather than controlling basal levels, highlighting the importance of studies in living human material (Holton et al., 2013).
To date, several studies have consistently generated valid human induced pluripotent stem cell (hIPSC) models of Alzheimer’s disease (AD) primarily from fibroblasts of patients with rare, early-onset familial AD (FAD) mutations (reviewed in Cao et al., 2014; Goldstein et al., 2015). In general, these models have fairly accurately represented disease phenotypes regarding increased detection of amyloid β (Aβ) peptides, increases in the Aβ42:40 ratio, increased phosphorylation of Tau protein, and the presence of enlarged endosomes. However, strikingly fewer papers have so far modeled SAD, which accounts for ≥95% of all clinical cases. Two recent studies have reported hIPSC lines from SAD patients (Israel et al., 2012; Kondo et al., 2013). Interestingly, in each case, only one of the two hIPSC-derived neuronal samples demonstrated phenotypes consistent with FAD lines. This variability speaks to the complexity of each human genetic background and its influence on disease causation and progression.
To begin to address these issues, we generated and analyzed a random cohort of 13 hIPSC lines derived from SAD patients and controls from the University of California, San Diego (UCSD) Shiley-Marcos Alzheimer’s Disease Research Center (ADRC). Using purified neurons made from these hIPSC lines, we focused on the SORL1 gene, which encodes an endocytic trafficking factor whose levels modulate the processing of amyloid precursor protein (APP) to Aβ and other proteolytic products implicated in SAD (Andersen et al., 2005). Loss of SORL1 expression has been documented in SAD cases (Dodson et al., 2006; Scherzer et al., 2004), and the SORL1 locus has been associated with SAD in both candidate gene and GWAS analyses (Bettens et al., 2008; Lambert et al., 2013; Lee et al., 2007; Reitz et al., 2011; Rogaeva et al., 2007). Despite these findings, it is currently unknown how expression of SORL1 is lost in SAD and how common variants in the SORL1 gene contribute to expression regulation.
We report that, in hIPSC-derived purified human neurons, basal levels of SORL1 expression are highly variable but that we can consistently induce SORL1 expression above the baseline and reduce levels of Aβ using neurotrophic and intracellular signaling molecules. Moreover, SORL1 expression induction and the decrease of Aβ in response to neurotrophin treatment is highly correlated with a common genotypic risk variant at the 5′ end of SORL1 (minor allele frequency = 0.42), suggesting that common variants in this region control this signaling response. Interestingly, we found that this phenotype is independent of whether neurons were derived from SAD patients or controls and that manipulation of SORL1 expression (by knockdown or overexpression) directly regulates the amount of Aβ peptides secreted by hIPSC-derived human neurons. These experiments, along with previous work in other models (Caglayan et al., 2014), confirm the importance of the SORL1/APP pathway in SAD and highlight hIPSC technology as a novel way to test the role of human genomic variants in complex disease.
RESULTS
Basal Levels of SORL1 Expression Are Variable in Human NSCs and Neurons Derived from hIPSCs
Reduced SORL1 expression has been reported in SAD patient lymphoblasts and cerebrospinal fluid and in post-mortem analyses (Dodson et al., 2006; Ma et al., 2009; Scherzer et al., 2004). We had access to 22 fibroblast samples (10 non-demented controls [NDCs] and 12 probable SAD cases clinically diagnosed by National Institute of Neurological and Communicative Disorders and Stroke-Alzheimer’s Disease and Related Disorders Association (NINCDS-ADRDA) (McKhann et al., 1984)) from the UCSD ADRC (Figure 1A and Table 1). Our laboratory has also reprogrammed fibroblasts derived from J. Craig Venter (J.C.V.) (Gore et al., 2011; Woodruff et al., 2013), whose diploid genome sequence has been published and is known to carry genomic variants in SORL1 associated with SAD (Levy et al., 2007). Cellular phenotypes of genomic risk variants may be difficult to detect in post-mortem tissue, which is often a mix of cell types taken at the end stage of a diseased brain. We hypothesized that a culture of purified human neurons containing the endogenous genome of an individual might provide a more sensitive system to detect potentially subtle phenotypes. Therefore, to ask whether SORL1 genomic variants affect basal expression at levels detectable in hIPSC-derived neurons, we first genotyped our small fibroblast cohort for variants in SORL1 documented previously to be associated with risk (R) or protection (P) from AD (Rogaeva et al., 2007). Next we reprogrammed a random subset of these fibroblasts, established hIPSC lines, differentiated neural stem cell (NSC) lines, and purified neurons from these individuals following standard protocols (Israel et al., 2012; Yuan et al., 2011; Table 1; Figure S1A). All of the hIPSC lines used in this study are euploid and can differentiate into the three germ layers: endoderm, mesoderm, and ectoderm (Table S1). Human neurons differentiated and purified using this protocol are a mix of glutamatergic, GABAergic, and cholinergic subtypes and are electrophysiologically active (Israel et al., 2012).
Figure 1. 5′ SNPs in the SORL1 Gene Comprise a Haplotype in Strong LD.
(A) Diagram of the experimental design and cell lines. See also Tables 1 and S1.
(B) Representative diagram of the SORL1 gene and LD structure of the 5′ haplotype and 3′ haplotype in SORL1: SNPs 4, 7, 8, 9, 10, and 11 and SNPs 22, 23, 24, and 25, generated from the 1000 Genomes project, Phase I. R2 correlation values are indicated on the LD plot and color-coded in the underlying squares. R2 values of 1 indicate complete linkage. See also Figure S1C.
(C) 23 patient fibroblast samples were genotyped for four SNPs (SNP4, SNP8, SNP9, and SNP10) at the 5′ end of SORL1 identified in Rogaeva et al. (2007) as being associated with AD. The percentage of individuals with homozygous risk SNPs at positions 8, 9, and 10 were identical, consistent with the previously reported strong LD of these SNPs.
(D) Fibroblast samples from the same patient group were genotyped for four SNPs (SNP22, SNP23, SNP24, and SNP25) at the 3′ end of SORL1 also identified in Rogaeva et al. (2007) as being associated with AD. In these samples, the percentage of individuals with risk SNPs at these positions was not identical, suggesting a lack of a clear haplotype at the 3′ end in this sample set.
Table 1.
Cell Lines Used in This Study
| UCSD ADRC No. | Name | SORL1 5′ Genotype | Diagnosis | Cell Type Analyzed | Reference |
|---|---|---|---|---|---|
| 8149 | SAD1 | R/R | probable SAD | fib, NSC, neuron | Israel et al., 2012 |
| 3093 | SAD2 | P/P | probable SAD | fib, NSC, neuron | Israel et al., 2012 |
| 2991 | SAD3 | R/R | probable SADa | fib, NSC, neuron | |
| 3121 | SAD4 | R/P | probable SAD | fib, NSC, neuron | |
| 3158 | SAD5 | R/P | probable SAD | fib, NSC, neuron | |
| 8097 | SAD6 | P/P | probable SAD | fib, NSC, neuron | |
| 2800 | SAD7 | R/P | probable SAD | fib, NSC, neuron | |
| 3053 | FIB3053 | P/P | probable SAD | fibb | |
| 8094 | FIB8094 | R/R | probable SAD | fibb | |
| 3131 | FIB3131 | R/R | probable SAD | fibb | |
| 8020 | FIB8020 | R/R | probable SAD | fibb | |
| 654 | FIB654 | R/R | probable SAD | fibb | |
| 3113 | FIB3113 | R/R | probable SAD | fibb | |
| 27 | NDC1 | R/P | NDC | fib, NSC, neuron | Israel et al., 2012 |
| 8011 | NDC2 | R/R | NDC | fib, NSC, neuron | Israel et al., 2012 |
| 14096 | NDC3 | R/P | NDC | fib, NSC, neuron | |
| 40 | NDC4 | R/P | NDC | fib, NSC, neuron | |
| 8078 | NDC5 | P/P | NDC | fib, NSC, neuron | |
| J.C.V. | J.C.V. | R/R | NDC | fib, NSC, neuron | Gore et al., 2011; Woodruff et al., 2013 |
| 2608 | FIB2608 | P/P | NDC | fibb | |
| 2785 | FIB2785 | P/P | NDC | fibb | |
| 8072 | FIB8072 | P/P | NDC | fibb | |
| 8150 | FIB8150 | R/P | NDC | fibb |
P/P = protective haplotype at 5′ end of SORL1 (SNPs 8, 9, and 10); R/P, heterozygous risk/protective haplotype; R/R, risk haplotype; fib, fibroblast.
Patient SAD3 was diagnosed as having probable AD during the course of this study and prior to death. This patient passed away while this manuscript was in preparation, and post-mortem pathology has revealed a diagnosis of tangle-predominant dementia with hippocampal sclerosis rather than AD. However, the statistically significant findings presented in this manuscript are only related to SORL1 genotypes and not to whether patients are diagnosed as NDC or SAD. Therefore, we still included hIPSC-derived cell lines from patient SAD3 in our results.
The fibroblast line was not reprogrammed.
Two common haplotypes in the SORL1 locus have been associated with SAD risk in multiple studies, one defined by a set of SNPs in strong linkage disequilibrium (LD) at the 5′ end of the gene and one defined by a set of SNPs in LD at the 3′ end of the gene (Rogaeva et al., 2007). Multiple studies have confirmed a strong linkage disequilibrium in these regions using various population datasets (Bettens et al., 2008; Rogaeva et al., 2007; Wen et al., 2013). We genotyped our fibroblast lines for eight representative SORL1 variants, four SNPs in the 5′ region and four SNPs in the 3′ region. For clarity, we utilized the names of these SNPs as assigned by Rogaeva et al. (2007): SNPs 4, 8, 9, and 10 at the 5′ end and SNPs 22, 23, 24, and 25 at the 3′ end of SORL1 (Figure 1B; Figure S1). We also queried publicly available data from phase I of the 1000 Genomes Project (1000 Genomes Project Consortium et al., 2012) focusing on the Centre d’Etude du Polymorphisme Humain population, which closely resembles the population of fibroblast donors from the ADRC (northern and western European populations) to confirm the extent of disequilibrium at these sites (Figure 1B). Three of the SNPs at the 5′ end of SORL1 (SNPs 8, 9, and 10) in our 23 fibroblast samples showed apparent complete LD (the SNPs were found together in all unambiguous homozygous samples, and the estimated pairwise R2 was 1.0; Figure 1C; Figure S1C). The same pattern of LD was observed in our analysis of the 1000 Genomes data (pairwise R2 = 1.0; Figure 1B). LD was not as strong at the 3′ end of SORL1 in our sample (Figure 1D; Figure S1C) or in the public data, where the pairwise R2 ranged from 0.64–0.88 (Figure 1B). There was little association between the 3′ haplotypes and cellular phenotypes in preliminary experiments (Figure S3), and, therefore, we focused our attention on the 5′ haplotype. We designated the haplotype at the 5′ end comprised by SNPs 8, 9, and 10 as either R (risk) or P (protective) based on previous association studies (Reitz et al., 2011; Rogaeva et al., 2007) and classified our patient samples as P/P, R/P, or R/R. That is, R/R patients are homozygous for the variants at SNPs 8, 9, and 10, which have been reported to be associated with an increased risk of developing SAD; P/P patients are homozygous for alleles at SNPs 8, 9, and 10, associated with a reduced SAD risk; and R/P patients are heterozygous for risk and protective SNPs. Interestingly, previous work using postmortem brain tissue has implicated the 5′ region of SORL1 in alternative splicing and reduced SORL1 expression in the temporal cortex, although the functional variants have not been identified (Grear et al., 2009; McCarthy et al., 2012). Therefore, we hypothesized that hIPSC-derived neurons with risk haplotypes in this 5′ region may show differential SORL1 expression.
We analyzed the levels of SORL1 mRNA in NSC lines and neurons differentiated and purified from NSC lines (six NDC genomes, including J.C.V., and seven SAD genomes). At the basal level, we found a highly variable expression of SORL1 in all cell types and genotypes (P/P, R/P, and R/R) with no detectable differences in SORL1 expression because of disease state (NDC versus AD) or SORL1 haplotype (Figures 2A–2D). We also analyzed the parent fibroblast lines for basal levels of SORL1 gene expression and, similarly, found no differences between genotypes or disease states in these cells (Figures S2A and S2B) We did see a significant increase in SORL1 expression upon differentiation to NSCs and neurons (Figure S2C), consistent with previous reports showing that SORL1 is highly expressed in the CNS (Mörwald et al., 1997; Motoi et al., 1999).
Figure 2. Basal Levels of SORL1 mRNA Are Variable in NSCs and Purified Human Neurons and Do Not Correlate with a Diagnosis of Probable SAD or NDC or with the Presence of SORL1 R and P Genotypes.
(A) qRT-PCR analysis of basal SORL1 expression in 13 NSC lines. No significant difference was detected related to the disease state, NDC versus AD. Statistics: Mann-Whitney test; NDC versus AD, p = 0.63; NDC, n = 6; AD, n = 7. See also Figure S2A.
(B) qRT-PCR analysis of basal SORL1 expression in purified neurons made from 13 NSC lines. No significant difference was detected related to the disease state, NDC versus AD. Statistics: Mann-Whitney test; NDC versus AD, p = 0.8; NDC, n = 6; SAD, n = 7. See also Figure S2A.
(C) qRT-PCR analysis of basal SORL1 expression in 13 NSC lines. No significant difference was detected related to the SORL1 R or P haplo-type. Statistics: Kruskal-Wallis test was used to compare P/P versus R/P versus R/R (p = 0.56; P/P, n = 3; R/P, n = 6; R/R, n = 4). See also Figure S2B.
(D) qRT-PCR analysis of basal SORL1 expression in purified neurons made from 13 NSC lines. No significant difference was detected related to the SORL1 R or P haplotype. Statistics: Kruskal-Wallis test was used to compare P/P versus R/P versus R/R (p = 0.35; P/P, n = 3; R/P, n = 6; R/R, n = 4). See also Figure S2B.
All error bars are SD.
A SORL1 Haplotype Associated with Protection from AD May Regulate the Induction of SORL1 Expression by Certain Growth and Neurotrophic Factors
Our neuronal differentiation protocol contains a mix of growth and neurotrophic factors (Yuan et al., 2011) including brain-derived neurotrophic factor (BDNF), which can induce the murine homolog of SORL1 (LR11) (Rohe et al., 2009), and cyclic AMP (cAMP), which can be elevated by BDNF treatment (Cheng et al., 2011). BDNF has been hypothesized to be protective in various animal models of AD, and loss of BDNF has been implicated in AD (Nagahara et al., 2009). Treatments that increase intracellular cAMP levels, such as liraglutide, have also been shown to be protective in AD models (Hunter and Hölscher, 2012; Parthsarathy and Hölscher, 2013). Therefore, we tested the acute effects of these two factors, BDNF and cAMP, on SORL1 expression in purified human neurons. Although several growth and neurotrophic factors were present in our neuronal media (Israel et al., 2012; Yuan et al., 2011), we induced SORL1 expression above the baseline in our system by acutely treating neurons with additional, exogenous BDNF or cAMP for 24 hr.
We measured SORL1 expression induction by quantitative RT-PCR (qRT-PCR) and found that, averaging across neurons derived from our 13 cell lines, the absolute relative expression level for BDNF-treated neurons was not significantly different from that of untreated neurons (Figure 3A). However, we consistently noticed that neurons from 9 of the 13 patients exhibited a significant induction of SORL1 expression above the baseline in response to BDNF, whereas neurons from 4 of the 13 patients did not (Figure 3B; Table 2). When we examined the cell line response to BDNF by genotype, we found that all four of the lines in which SORL1 was not induced by BDNF were homozygous R/R, whereas all lines with a P allele did respond to BDNF treatment (Figure 3C, post hoc Fisher’s exact test, p = 0.001; Table 2). This suggests that the P haplotype is a marker for a dominant protective genetic variant. Available SAD case control data are highly consistent with a dominant protective variant (Table S2). A pooled analysis of public genotype data of 1,640 SAD cases and 1,728 controls with a similar ethnic background as our series suggests a consistent protective effect of the R/P and P/P haplotypes relative to the R/R haplotype, with an odds ratio of 0.77 (95% confidence interval [CI], 0.66–0.90) for R/P versus R/R haplotypes, an identical odds ratio (OR) of 0.77 (95% CI, 0.63–0.93) for P/P versus R/R haplotypes, and an OR of 0.99 (95% CI, 0.82–1.20) for R/P versus P/P haplotypes. The sample size was not sufficient to formally compare SORL1 expression in response to BDNF across all three haplotypes in our cell lines. However, induction in R/P cell lines was similar to induction in P/P cell lines, and mean induction in the pooled P/P and R/P cell lines was significantly higher than that observed in the R/R cell lines (Figure 3C).
Figure 3. SORL1 Expression Induction by the Neurotrophin BDNF Is Dependent on a Haplotype Associated with Protection from SAD, and SORL1 Expression Induction by cAMP Is Independent of This Haplotype.
(A) SORL1 induction was tested in response to BDNF treatment (50 ng/ml) of purified neurons differentiated from 13 NSC lines. The average relative expression across all cell lines by qRT-PCR analysis of purified neuron samples treated with BDNF from 13 patients shows no significant induction of SORL1 expression with treatment. Statistics: Mann-Whitney test, baseline versus BDNF, p = 0.16.
(B) qRT-PCR analysis of purified neurons derived from each individual cell line treated with BDNF shows that all but four cell lines induce SORL1 expression upon treatment. Statistics: t tests within each cell line, NS, not significant, *p < 0.05, **p < 0.01.
(C) Analysis of SORL1 expression levels in response to BDNF treatment when samples are grouped by variant haplotype at the 5′ end of SORL1. The fold change of SORL1 expression was calculated between non-treated (NT) and BDNF-treated samples within the genotype and reveals that neurons with protective alleles (P/P and R/P) induce SORL1 in response to BDNF, whereas R/R cells do not. Statistics: Mann-Whitney test was used to compare P/P and R/P versus R/R carriers (p = 0.003; P/P, n = 3; R/P, n = 6; R/R, n = 4. See also Figures S3A–S3E.
(D) SORL1 induction was tested in response to cAMP treatment (250 μg/ml) of purified human neurons differentiated from 13 NSC lines. The average of qRT-PCR analysis of purified neuron samples treated with cAMP shows a significant induction of SORL1 expression with treatment. Statistics: Mann-Whitney test, baseline versus cAMP, p = 0.02.
(E) qRT-PCR analysis of purified neurons derived from each individual cell line treated with cAMP shows that all but two cell lines induce SORL1 expression upon treatment. Statistics: t tests within each cell line, *p < 0.05, **p < 0.01.
(F) Analysis of SORL1 expression levels in response to cAMP treatment when samples are grouped by variant haplotype at the 5′ end of SORL1. The fold change of SORL1 expression was calculated between NT and cAMP-treated samples within the genotype and reveals no difference in SORL1 expression induction between genotypes. Statistics: Mann-Whitney test was used to compare P/P and R/P versus R/R carriers (p = 0.075; P/P, n = 3; R/P, n = 6; R/R, n = 4). All error bars are SD.
Table 2.
Summary of Findings on SORL1 Expression Induction and APP Processing after BDNF or cAMP Treatment in Human Neurons
| SORL1 Expression | Individual | Genotype | Average Fold Change in SORL1 Expression Induced by BDNF | SD | Inducibility Phenotype | Average Fold Change in SORL1 Expression Induced by cAMP | SD | Inducibility Phenotype |
|---|---|---|---|---|---|---|---|---|
| SAD2 | P/P | 1.52 | 0.007 | inducible | 1.84 | 0.29 | inducible | |
| NDC5 | P/P | 1.45 | 0.07 | inducible | 2.64 | 0.06 | inducible | |
| SAD6 | P/P | 1.49 | 0.12 | inducible | 1.53 | 0.29 | inducible | |
| NDC4 | R/P | 1.81 | 0.33 | inducible | 3.57 | 0.39 | inducible | |
| SAD4 | R/P | 1.41 | 0.14 | inducible | 2.23 | 0.57 | inducible | |
| SAD5 | R/P | 1.70 | 0.16 | inducible | 1.52 | 0.09 | inducible | |
| SAD7 | R/P | 1.51 | 0.11 | inducible | 1.72 | 0.12 | inducible | |
| NDC3 | R/P | 1.79 | 0.09 | inducible | 1.91 | 0.27 | inducible | |
| NDC1 | R/P | 2.09 | 0.13 | inducible | 2.01 | 0.57 | inducible | |
| SAD1 | R/R | 1.19 | 0.22 | non-inducible | 1.30 | 0.11 | non-Inducible | |
| CV | R/R | 1.03 | 0.41 | non-inducible | 1.17 | 0.16 | non-inducible | |
| NDC2 | R/R | 0.80 | 0.07 | non-inducible | 1.87 | 0.09 | inducible | |
| SAD3 | R/R | 1.22 | 0.09 | non-inducible | 1.57 | 0.07 | inducible |
| APP Processing | Individual | Genotype | Average Fold Change in Aβ Levels after BDNF Treatment | SD | ≥ Than 10% Decrease in Aβ Peptides | Average Fold Change in Aβ Levels after cAMP Treatment | SD | ≥ Than 10% Decrease in Aβ Peptides | Average Fold Aβ Change +BDNF +SORL1shRNA | Average Fold Aβ Change +cAMP+SORL1 shRNA |
|---|---|---|---|---|---|---|---|---|---|---|
| SAD2 | P/P | 0.64 | 0.004 | yes | 0.34 | 0.003 | yes | 0.83 | 0.41 | |
| NDC5 | P/P | 0.74 | 0.01 | yes | 0.56 | 0.002 | yes | 0.87 | 0.49 | |
| SAD6 | P/P | 0.82 | 0.12 | yes | 0.38 | 0.01 | yes | 0.99 | 0.41 | |
| NDC4 | R/P | 0.76 | 0.04 | yes | 0.49 | 0.04 | yes | 0.99 | 0.54 | |
| SAD4 | R/P | 0.91 | 0.02 | no | 0.38 | 0.03 | yes | 0.95 | 0.44 | |
| SAD5 | R/P | 0.74 | 0.003 | yes | 0.49 | 0.02 | yes | 0.87 | 0.46 | |
| SAD7 | R/P | 0.82 | 0.04 | yes | 0.58 | 0.08 | yes | 0.95 | 0.65 | |
| NDC3 | R/P | 0.74 | 0.01 | yes | 0.44 | 0.005 | yes | 0.99 | 0.34 | |
| NDC1 | R/P | 0.86 | 0.06 | yes | 0.52 | 0.003 | yes | 0.91 | 0.60 | |
| SAD1 | R/R | 0.91 | 0.03 | no | 0.44 | 0.02 | yes | 0.99 | 0.58 | |
| CV | R/R | 0.99 | 0.05 | no | 0.54 | 0.01 | yes | 0.99 | 0.62 | |
| NDC2 | R/R | 0.98 | 0.07 | no | 0.58 | 0.01 | yes | 0.99 | 0.55 | |
| SAD3 | R/R | 1.05 | 0.12 | no | 0.59 | 0.01 | yes | 0.82 | 0.47 |
13 patient hIPSC cell lines were differentiated to generate purified neurons. The following tests were performed:
(1) SORL1 mRNA expression induction with BDNF or cAMP treatment. The fold induction of SORL1 was analyzed by non-parametric Mann-Whitney test. For BDNF treatment, P/P and R/P versus R/R, p = 0.003. For cAMP treatment, P/P and R/P versus R/R, p = 0.075. To test whether the association of non-inducibility with the R/R genotype was significant for BDNF treatment, we set the phenotypic cutoff for non-inducibility at the statistically significant lowest R/P (1.41) value and performed a Fisher’s exact test. The two-tailed p value between inducible and non-inducible is 0.001. For cAMP treatment, the phenotypic cutoff for non-inducibility was set at the lowest R/P (1.52) value for the Fisher’s exact test. The two-tailed p value between inducible and non-inducible is 0.08.
(2) Aβ1–40 peptides were measured after treatment with BDNF or cAMP. The fold decrease in Aβ1–40 production was analyzed by Mann-Whitney test. For BDNF, P/P and R/P versus R/R, p = 0.006. For cAMP, P/P and R/P versus R/R, p = 0.14.
(3) Aβ1–40 peptides were measured after treatment with BDNF or cAMP in the presence of a SORL1 shRNA. The fold decrease in Aβ1–40 peptides after BDNF treatment was significantly elevated in P/P and R/P genotypes but not in R/R genotypes, with SORL1 knockdown as analyzed by Mann-Whitney test: P/P and R/P SCR versus KD, p = 0.008; R/R SCR versus KD, p = 0.5. The fold decrease in Aβ1–40 peptides after cAMP treatment was not significantly different in any genotype with SORL1 knockdown as analyzed by Mann-Whitney test: P/P and R/P SCR versus KD, p = 0.86; R/R SCR versus KD, p = 0.22.
Conversely, the BDNF inducibility phenotype did not correlate with the presence of putative risk variants at SNP4 or SNPs 22–25 at the 3′ end of SORL1 (Figures S3A and S3B). Two different SORL1 primer sets and two different housekeeping genes confirmed our mRNA expression results (Figure S3C). Because of the small amount of material we can routinely obtain from purified neurons, we characterized this phenotype primarily at the mRNA level. However, we analyzed SORL1 protein levels from representative fibroblast, NSC, and purified neuron genotypes and found that SORL1 protein induction from fibroblast to NSC was blunted in R/R cells compared with P/P and R/P cells, and, in purified neurons treated with BDNF, there was no apparent increase in SORL1 protein upon treatment in R/R cells (Figures S3D and S3E).
Interestingly, we also found that cAMP strongly induced SORL1 expression in neurons, and the average absolute relative expression level increase for cAMP-treated neurons was significant (Figure 3D). When each individual was examined, we found that cAMP treatment induced SORL1 expression above the baseline from most, but not all, cell lines (Figure 3E; Table 2).
When we examined cell lines treated with cAMP, we found that two R/R cell lines did not respond to treatment, whereas two R/R cell lines did (Figure 3E). cAMP treatment consistently and robustly induced SORL1 expression in all other cell lines, suggesting heterogeneity in this signaling pathway (Figure 3F; Table 2). We did not find a significant difference in induction of SORL1 expression between the P/P, R/P, or P/P genotypes after cAMP treatment (Figure 3F; Table 2).
To rule out the possibility that this induction phenotype in response to BDNF may be due to decreased pluripotency or differentiation efficiency, we compared Tra 1–81 and Tra 1–60 in hIPSC and CD24 labeling after neuronal differentiation and detected no differences in R/R cells (Figures S4A–S4C). Purified neurons of all genotypes showed significantly increased expression of Tau mRNA and exhibited an obvious neuronal morphology compared with NSC lines (Figures S1A and S4D). Therefore, we concluded that there are no differences in pluripotency, differentiation capability, or apparent neuronal phenotype in cells of different SORL1 genotypes. We also tested purified neurons for expression of the BDNF receptor TrkB and found no differences in TrkB expression in R/R cells (Figures S4E and S4F). Taken together, our data suggest that, rather than controlling basal expression levels, naturally occurring variants in humans in the 5′ region of SORL1 modulate expression induction in response to BDNF exposure.
Induction of SORL1 Expression Correlates with Decreased Aβ Peptide Production
APP undergoes two cleavage events, by β and γ secretases, to generate the potentially neurotoxic Aβ peptide and other fragments. Previous work suggests that SORL1 serves a protective function by binding full-length APP and either sorting it away from amyloidogenic β and γ secretase cleavage pathways or masking the secretase binding site (Andersen et al., 2005; Hermey, 2009; Schmidt et al., 2012). In particular, reducing SORL1 expression in mice and non-neuronal cell lines increases the production of Aβ peptides, whereas induction of SORL1 expression lowers the production of Aβ (Dodson et al., 2008; Rogaeva et al., 2007; Rohe et al., 2009). Because the amount of Aβ peptide produced in the human brain is an important SAD phenotype that is potentially related to the cause or progression of SAD, we tested whether induction of SORL1 with BDNF or cAMP in purified human neurons had an effect on Aβ production and secretion. Measurement of the average absolute levels of Aβ1–40 peptides in all cell lines revealed no significant differences between cells treated with BDNF or untreated cells (Figure 4A). However, a clear trend emerged when we evaluated the fold reduction in Aβ1–40 peptides in each individual line in response to BDNF treatment. The neurons from the four hIPSC lines carrying the R/R genomes that did not induce SORL1 expression also did not significantly reduce Aβ peptides in response to BDNF treatment, whereas all neurons from hIPSC lines carrying R/P or P/P genomes significantly reduced Aβ levels, with the exception of one line, SAD4 (Figure 4B; Table 2). When analyzed together, the reduction of Aβ1–40 in all genomes with a P allele versus R/R genomes was significant (Figure 4C). As published previously (Israel et al., 2012), in some cell lines Aβ1–42 is below the detection range of our assay because, in our system, endogenous levels of this analyte are approximately five times lower than Aβ1–40 in neuronal media. However, for some lines where we were able to consistently detect Aβ1–42 peptides, we observed a similar trend as with Aβ1–40 (Figure S5A). Although the decrease in Aβ production induced by BDNF in P/P and R/P genomes is apparently modest (20%–30%), it is in line with reasonable expectations for changes that might influence risk but not direct induction of disease. For example, aggressive, early-onset FAD can be caused by a highly penetrant APP duplication that increases APP expression and Aβ production by 50% (Israel et al., 2012; Rovelet-Lecrux et al., 2006; Shi et al., 2012). Therefore, the average 20% change in APP processing we observed in these in vitro experiments is significant in the context of SAD risk.
Figure 4. Neurons Responsive to SORL1 Expression Induction Show a Decrease in Aβ Peptides.
(A) Aβ peptide levels were measured after 24 hr of exogenous BDNF treatment. When average absolute values of Aβ in neurons from 13 cell lines were analyzed together, BDNF treatment did not significantly reduce Aβ levels. Statistics: Mann-Whitney test, baseline versus BDNF, p = 0.39.
(B) Aβ peptide levels measured from purified neurons from each individual cell line ± BDNF treatment shows that all cell lines that did induce SORL1 expression with BDNF treatment, except SAD4, have a reduction in Aβ levels but that there is no change in Aβ peptides in the four cell lines that did not induce SORL1 expression. Statistics: t tests within each cell line, *p < 0.05, **p < 0.01. See also Figure S5A.
(C). Analysis of Aβ reduction when samples are grouped by variant haplotype at the 5′ end of SORL1 in response to BDNF treatment. Neurons treated with BDNF showed a significant reduction in Aβ peptides in cells with protective alleles (P/P and R/P). Aβ peptide production in cells homozygous for the risk alleles (R/R) was not affected by BDNF treatment. Statistics: Mann-Whitney test was used to compare P/P and R/P versus R/R (p = 0.006; P/P, n = 3; R/P, n = 6; R/R n = 4).
(D) Aβ peptide levels were measured after 24 hr of exogenous cAMP treatment. When average absolute values of Aβ in neurons from 13 cell lines were analyzed together, cAMP treatment did significantly reduce Aβ levels. Statistics: Mann-Whitney test, baseline versus cAMP, p = 0.007.
(E) Aβ peptide levels from purified neurons from each individual cell line ± cAMP treatment shows that all cell lines have a significant reduction in Aβ regardless of whether SORL1 expression was induced. Statistics: t tests within each cell line, *p < 0.05, **p < 0.01.
(F) Analysis of Aβ reduction when samples are grouped by variant haplotype at the 5′ end of SORL1 in response to cAMP treatment. Neurons treated with cAMP showed a reduction in Aβ peptides in all genotypes, with no significant differences between neurons with P or R alleles. Statistics: Mann-Whitney test was used to compare P/P and R/P versus R/R Aβ response to cAMP treatment (p = 0.14; P/P, n = 3; R/P, n = 6; R//R, n = 4).
All error bars are SD.
In contrast to BDNF, cAMP treatment significantly and robustly decreased absolute Aβ levels by an average of 50%–60% in purified neurons of all genotypes (Figures 4D and 4E; Table 2). This decrease was similar among all genotypes tested, and there was no significant difference in Aβ reduction between P/P and R/P compared with R/R genomes (Figures 4E and 4F; Table 2). Taken together with our expression data, these results suggest that SAD risk associated with SORL1 genomic variants may be generated via specific signaling pathways within neurons and that the impact of SORL1 induction may vary in different genomic backgrounds.
Decreased Aβ Peptides Caused by BDNF, but Not cAMP, Requires SORL1
To determine whether the reduction of Aβ in response to BDNF or cAMP treatment requires SORL1 expression, we designed an shRNA against the human SORL1 mRNA and documented a robust and consistent knockdown of SORL1 mRNA and protein in transduced purified neurons of all genotypes (Figures S5B and S5C). We then treated transduced neurons with BDNF and observed that the decrease in Aβ peptides in neurons with P haplotypes at the SORL1 locus was almost completely abrogated in neurons expressing the SORL1 small hairpin RNA (shRNA), whereas knockdown of SORL1 had little effect in cells with R/R genotypes (Figure 5A; Table 2), suggesting that the decrease in APP processing after BDNF treatment is dependent on SORL1. In contrast, SORL1 knockdown neurons treated with cAMP still exhibited a reduction of Aβ peptides (Figure 5B), indicating that, in purified neurons, cAMP regulates pathways in addition to SORL1 that can impact Aβ production independently.
Figure 5. SORL1 Is Required for Aβ Reduction in Neurons Treated with BDNF but Not cAMP, and Overexpression of SORL1 Decreases Aβ Production in a BDNF Non-responsive Cell Line.
(A) Neurons were transduced with a lentivirus containing either a SORL1 scrambled (SCR) shRNA (−) or a SORL1 knockdown (KD) shRNA (+) and treated with exogenous BDNF (+) for 24 hr. In BDNF-responsive neurons with P alleles, the presence of a SORL1 shRNA significantly reduced the decrease in Aβ peptides with BDNF treatment. Statistics: Mann-Whitney test was used to compare SCR versus KD for BDNF treatment (P/P and R/P SCR versus KD, p = 0.008; R/R SCR versus KD, p = 0.5; P/P, n = 3; R/P, n = 6; R/R, n = 4). Between genotypes, there was no statistical difference in Aβ peptides among all lines transduced with the SORL1 shRNA and treated with BDNF (white bars, P/P and R/P versus R/R, p = 0.19). See also Figures S5B and S5C.
(B) Neurons were transduced with a lentivirus containing either a SORL1 SCR shRNA (−) or a SORL1 KD shRNA (+) and treated with exogenous cAMP (+) for 24 hr. In all neurons there was a reduction in Aβ peptide production upon treatment with cAMP that was not significantly altered by the SORL1 shRNA. Statistics: Mann-Whitney test was used to compare SCR versus KD cAMP treatment (P/P and R/P SCR versus KD, p = 0.86; R/R SCR versus KD, p = 0.22; P/P, n = 3; R/P, n = 6; R/R, n = 4). Between genotypes, there was no statistical difference in Aβ peptides in all lines transduced with the SORL1 shRNA (P/P and R/P versus R/R, p = 0.29).
(C) J.C.V. neurons stably expressing either empty vector or a SORL1 cDNA were generated using the PiggyBac system, and qRT-PCR analysis of SORL1 expression in stable SORL1-overexpressing neurons was compared to vector alone. SORL1-overexpressing neurons show an approximately 4-fold increase in SORL1 expression.
(D) Aβ1–40&42 peptides were measured from the medium of J.C.V. neurons stably expressing either empty vector or a SORL1 cDNA after 5 days. The fold change in Aβ peptides was calculated between vector only and SORL1 cDNA-expressing neurons. Statistics: paired t test comparing vector alone with SORL1 cDNA, **p < 0.01.
(E) Summary of findings regarding SORL1 expression in human purified neurons. The schematic depicts only the region of the SORL1 gene containing the 5′ variants analyzed in this study. Neurons with P variants have increased SORL1 expression with BDNF treatment, resulting in reduced Aβ production, which may lead to reduced AD risk. Neurons with the R/R haplotype have no change in SORL1 expression or Aβ production with BDNF treatment, therefore potentially increasing the risk for developing AD. However, alternative methods to induce SORL1 expression that do not involve BDNF signal transduction pathways may be a viable strategy for individuals harboring risk variants and can be tested on various genetic backgrounds using hIPSC technology.
Overexpression of a SORL1 cDNA in BDNF Non-Responsive Cells Reduces Aβ Peptides
Our data corroborate most other work in cell and animal models, demonstrating that increased SORL1 expression reduces Aβ peptides and raises the possibility of targeting this pathway for new AD therapies (Caglayan et al., 2014). However, because SORL1 genetic risk variants may affect pathways that regulate SORL1 expression, individual patient genotypes may respond differently to various agents designed to induce SORL1. To test whether SORL1 activation can be beneficial in neurons that do not induce expression after BDNF treatment, we stably overexpressed a SORL1 cDNA in R/R neurons (derived from J.C.V. hIPSCs). We observed that, after 5 days in culture, the R/R neurons from J.C.V. containing the SORL1 cDNA showed a 4-fold increase in SORL1 mRNA, approximately the same fold induction as the highest cell line induced with cAMP (Figure 5C). These SORL1-overexpressing neurons had significantly reduced levels of both Aβ40 and 42 peptides when the cells were harvested after 5 days in culture (Figure 5D).
DISCUSSION
We used hIPSC technology to elucidate molecular phenotypes associated with naturally occurring, uniquely human variants in a gene associated with SAD risk, SORL1. In particular, the neurotrophic growth factors and intracellular signaling molecules BDNF and cAMP can induce SORL1 expression above the baseline in some genotypes of human neurons. In the case of BDNF, only neurons with P/P or R/P genotypes increased SORL1 expression and demonstrated a significant reduction in Aβ, whereas there was no effect in R/R neurons. BDNF-induced reductions in Aβ peptides in P genomes required SORL1 expression induction, as demonstrated by shRNA experiments, suggesting that the degree of change in Aβ we observed in neurons upon BDNF treatment is dependent on SORL1 action, whereas cAMP treatment activates other pathways in addition to SORL1. Finally, we noted that activation of the SORL1 pathway by overexpression of SORL1 cDNA reduces Aβ generation even in neurons whose SORL1 gene is not responsive to BDNF. The effect of BDNF on SORL1 expression and APP processing correlated only with the SORL1 R or P genotypes and not with whether the hIPSC lines were derived from SAD or control fibroblasts. This observation is consistent with previous findings showing that SORL1 haplotypes modulate SAD risk with moderate effect size and is typical of most variants associated with increased SAD risk, given that the net effects result from polygenic interactions with many other factors contributing to the overall risk of developing disease. We propose that BDNF-induced increases in SORL1 expression in patients harboring the P haplotype may reduce their risk of developing SAD by reducing amyloidogenic processing of APP in their neurons. However, the different responses of purified neurons to BDNF versus cAMP also suggest that genomic variants impact specific intracellular signaling pathways and should be considered when designing potential therapeutics, which can be tested using an hIPSC system that captures individual genetic backgrounds in a dish (Figure 5E).
As observed in our sample, the 5′ 8, 9, and 10 SNPs are almost always reported as a haplotype block in northern and western European population samples (Figure 1B; Reitz et al., 2011; Rogaeva et al., 2007). These variants, along with others in the gene, have been associated with a number of potentially relevant endophenotypes (reviewed in Reitz, 2012). SNPs 8, 9, and 10 were in complete linkage disequilibrium in our sample and were in complete disequilibrium with the cellular phenotype defined by the SORL1 response to BDNF. Although the sample size was small, we suggest that we have identified a relevant upstream phenotypic effect behind consistent findings of genotypic association with clinical SAD. Consistent with our expression-related phenotype, neighboring 5′ SORL1 SNPs predicted a 2-fold difference in the SORL1 mRNA level in the temporal cortex (McCarthy et al., 2012). Although this study did not probe the exact 8, 9, and 10 loci, it examined the same genomic interval, which is in strong LD and supports the hypothesis that genetically determined expression levels are behind the 5′ SORL1-AD association.
Recently there have been several reports of rare, causative, single-nucleotide variants in the coding region of SORL1 that are associated with early-onset familial AD (Pottier et al., 2012) and late-onset AD (Vardarajan et al., 2015). Although our study and those of others (Grear et al., 2009; McCarthy et al., 2012) point to the 5′ region of SORL1 as a potentially important region in the regulation of SORL1 gene expression, the 8, 9, 10 SNPs mark a haplotype and are not necessarily causative variants. Genome editing in hIPSCs has been used successfully to introduce dominant mutations in several neurodegenerative disease models, including AD (Li et al., 2013; Soldner et al., 2011; Woodruff et al., 2013), and is a powerful technique to elucidate molecular phenotypes caused by these variants. However, most isogenic lines derived so far harbor single point mutants, and careful dissection of large haplotype blocks is a significant technical challenge. Important future work should involve dissection of the entire haplotype block using genome editing to more precisely identify the nucleotide variant(s) that is/are important for the regulation of SORL1 expression by BDNF.
Our work on SORL1 corroborates most previous studies in cell and animal models (Andersen et al., 2005; Caglayan et al., 2014; Rogaeva et al., 2007; Rohe et al., 2009) and reiterates that SORL1 is an important player in SAD biology. However, our data highlight some important differences between mouse and human systems and underscores the unique and novel applications of hIPSC technology to investigate disease-associated genes. First, by capturing an individual patient’s unique genetic background in a hIPSC line, we are able to examine genetic variants that are either not present or do not have the same effect in the mouse genome. Second, our data reveal that, in living cell types relevant to human neurodegenerative disease, expression, and response of SORL1 to stimulation is highly variable, and the phenotypic readout is different depending on the genetic backgrounds. Our study is distinct from other hIPSC models, demonstrating phenotypes of highly penetrant genetic variants that deterministically cause disease and in which the described phenotype is correlated with the disease state. Instead, we suggest that we have demonstrated the potential effects of a risk factor, not a penetrant, hereditary mutation. The fact that the genetic variant and associated cellular phenotype we have described is not correlated with the clinical disease state is, in fact, important when thinking about SAD. Each individual human harbors factors in his or her genome that predispose to or protect from SAD. How these factors interact with each other and the environment is unknown. Interestingly, two of the R/R individuals in this study are classified as NDC and are over the age of 65 with no clinically apparent signs of cognitive impairment. In the case of these individuals, we speculate that other protective factors in their genome may overcome possible gene expression defects in SORL1 and prevent disease. Similarly, two of the P/P individuals, whose hIPSC-derived neurons responded significantly to BDNF-induced SORL1 expression and reduction of Aβ, were diagnosed with probable SAD, indicating that these individuals may harbor variants at other loci that enhance the SAD risk by other mechanisms. These observations in our data are consistent with the pattern of low clinical penetrance expected for most SAD risk alleles (Bertram et al., 2007; Chouraki and Seshadri, 2014)
Because the hIPSC field progresses rapidly and more lines are generated from patients, we propose that an important future step in the field will be to use hIPSCs to understand the biology in human neurons and glia driven by the genes identified as risk factors. In addition to elucidating important biological pathways behind hits identified in GWASs, these studies will also drive the development of individualized treatments because risk factors may identify which patients are viable candidates for a specific drug or would preferentially participate in, or be excluded from, certain clinical trials. Progress in this area is vital to developing SAD treatments in particular because SAD may well be a function of a variety of phenotypes driven by different genetic factors and primary phenotypic lesions. Our work provides an important first advance in understanding how individual human genomes and specific genomic haplotypes might generate neuronal phenotypes and which factors might contribute to SAD in individual patients and provide concrete insights into how stem cell technology can drive an understanding of personalized medicine and clinical trial design in AD and beyond.
EXPERIMENTAL PROCEDURES
hIPSC, NSCs, and Neuron Generation and Characterization
All hIPSC lines were generated by four-factor reprogramming (Gore et al., 2011; Israel et al., 2012). hIPSC lines were maintained on a mouse embryonic fibroblast (MEF) feeder layer in HUES medium (Israel et al., 2012) and were routinely tested for mycoplasma. For RNA/DNA analysis, cells were transferred from MEFs to Matrigel, cultured in MEF-conditioned HUES medium, and maintained for several passages before harvesting. For germ layer differentiation analyses, embryoid bodies (EBs) were generated by growing hIPSC in low-adherence plates for 14 days without fibroblast growth factor (FGF) with a medium change every other day. Previously published hIPSC lines were cytogenetically karyotyped (Cell Line Genetics), whereas unpublished hIPSC lines were karyotyped digitally by hybridization to the Illumina Infinium Human-CoreExomeBeadChip module version 1.9.4. Pluripotency gene expression in published lines has been described previously (Gore et al., 2011; Israel et al., 2012). Unpublished lines were analyzed for pluripotency by fluorescence-activated cell sorting (FACS) for the cell surface antigens Tra 1–81 and Tra 1–60 (BD Biosciences). The differentiation capability of published lines has been described previously (Israel et al., 2012). For unpublished lines, RNA from EBs and parent hIPSC lines was harvested and converted to cDNA (Superscript, Invitrogen). qRT-PCR was performed with a primer specific for endoderm (AFP), mesoderm (DCN), and ectoderm/neuroectoderm (COL1A1, NEST). NSCs and neurons were generated using protocols described previously (Israel et al., 2012; Yuan et al., 2011) and purified by FACS. NSCs were stained for Nestin (Millipore), and neurons were stained for MAP2 (Millipore). For each genotype, neurons derived from at least two independent hIPSC lines were examined.
Cell Culture, Experimental Design, and Sample Collection
NSCs were maintained in DMEMF12 medium supplemented with B27, N2, and Fgf (20 ng/ml) and were routinely tested for mycoplasma. NSCs were differentiated to neurons by Fgf withdrawal and cultured in the presence of BDNF, glial cell line-derived neurotrophic factor, and dibutyryl cAMP (dbcAMP) for 3 weeks as described previously (Israel et al., 2012; Yuan et al., 2011). Purified neurons were generated by FACS using a published cell surface signature (Yuan et al., 2011). Neurons were plated in biological replicates treated as described in the figure legends and harvested for RNA or protein. All qRT-PCR samples were run in triplicate. SORL1 expression experiments and analyses in purified neurons are reported in the figure legends in terms of the 13 individual patient genomes analyzed. However, for each genotype (P/P, R/P, or R/R), neurons derived from at least two independent hIPSC lines were examined. SORL1 expression experiments and analyses were repeated in all genotypes for a minimum of two independent differentiations and, for some genotype representative cell lines (SAD1 [P/P], NDC1 [R/P], and CV [R/R]), four or more times.
SORL1 SNP Genotyping
Genotyped SNPs were identified from Rogaeva et al. (2007) and are listed in Figure 1.
Fibroblast cultures from each patient were lysed, and genomic DNA was isolated using the DNeasy kit (QIAGEN). Allele-specific primers for the SNPs analyzed were designed using the Web-based allele-specific primer-designing tool (http://bioinfo.biotec.or.th/WASP). PCR were run using Hot-Star Polymerase (QIAGEN) and analyzed by agarose gel electrophoresis. A list of all allele-specific primers used in this study is provided in the Supplemental Experimental Procedures.
Expression Analyses
SORL1 expression was analyzed by measuring mRNA and protein. For mRNA, total RNA from triplicate cell culture experiments was isolated using the RNeasy kit (QIAGEN) and DNase-treated (Ambion). cDNA was transcribed from total RNA using the Superscript first-strand system (Invitrogen). qRT-PCR was performed using FastStart SyBr Green (Roche) and samples were run on an Applied Biosystems 7300 real-time PCR system. RT-PCR primers for all genes in this study were designed using Primer3 and spanned intron-exon boundaries. A list of all primers is provided in the Supplemental Experimental Procedures. qRT-PCR data were analyzed using the ΔΔCt method and normalized to housekeeping genes TBP or RPL27, as noted in the figure legends. For protein, cells were lysed in 1% NP40 or radioimmunoprecipitation assay (RIPA) buffer. Protein concentration was determined using either Bradford (Bio-Rad) or bicinchoninic acid (BCA) (Pierce) assays. Protein lysates were run on NuPAGE 4%–12% Bis-Tris gels (Invitrogen), transferred to nitrocellulose membranes, and probed with antibodies to either SORL1 (BD Biosciences, clone 48/LR11) or Actin (Millipore). Quantification of the western blots was performed using ImageJ software.
BDNF and cAMP Treatment
For purified neurons, triplicate cell culture experiments were treated with BDNF (PeproTech, 50 ng/ml) or dbcAMP (Sigma, 250 μg/ml) for 24 hr. After 24 hr, cells were harvested for RNA using the RNeasy kit (QIAGEN) or protein using either 1% NP40 or RIPA buffer. Data represent at least two independent differentiation experiments.
Aβ Measurements
Purified neurons were seeded at 200,000 cells/well of a 96-well plate. After 5 days in culture, medium was harvested from triplicate wells and run on an Aβ Triplex ELISA plate (Meso Scale Discovery). Corresponding wells were lysed with RIPA buffer, and protein concentration was determined by BCA assay (Pierce). Aβ levels were normalized to the amount of total protein.
Statistics
Based on examination of distribution assumptions, t tests were used for within-cell line comparisons of expression levels before and after neurotrophin treatment, but otherwise all analyses were performed using non-parametric statistical tests. These tests are more conservative and less powerful that parametric ANOVA and t tests but are robust to normality assumptions and, therefore, not prone to spurious type I errors. Between-genotype differences in continuous variables were analyzed by Kruskal-Wallis test, as stated in the figure legends. Two group comparisons of pooled P/P and R/P cells lines to R/R cell lines were tested using non-parametric Mann-Whitney tests, as stated in the figure legends. The distribution of categorical outcomes across genotype groups was compared using Fisher’s exact test. All testing was two-sided and was performed using the R statistical programming language (R Development Core Team, 2008). All statistical results were verified independently using the VassarStats computational website (http://vassarstats.net/).
Supplementary Material
Highlights.
hIPSC-derived neurons were made from six control individuals and seven SAD patients
Neurons have SAD risk (R) or protective (P) variants in the SORL1 gene
P variant neurons increase SORL1 expression and reduce Aβ after BDNF treatment
The reduction of Aβ in response to BDNF is dependent on SORL1 expression
Acknowledgments
J.E.Y. is funded by postdoctoral fellowships from the California Institute of Regenerative Medicine, the A.P. Gianinni Foundation for Medical Research, and the BrightFocus Foundation. L.S.B.G. is funded by the California Institute of Regenerative Medicine (CIRM Tools and Technologies II, RT2-01927), CIRM Basic Biology V, (RR5-07011), and the NIH (NIH-NIA 2P50AG005131-31). We thank the donors of fibroblast samples from the UCSD Shiley-Marcos ADRC.
Footnotes
Supplemental Information includes Supplemental Experimental Procedures, five figures, and two tables and can be found with this article online at http://dx.doi.org/10.1016/j.stem.2015.02.004.
AUTHOR CONTRIBUTIONS
J.E.Y. and L.S.B.G conceived the project. J.E.Y, J.B.W., D.A.W, G.W., F.B., and A.C.R performed the experiments. G.W. and C.H. reprogrammed the fibroblast lines. J.E.Y, L.S.B.G., and S.D.E. analyzed the data. M.A.I. and S.H.Y. provided cell lines and commented on the manuscript. J.E.Y. and L.S.B.G. wrote the manuscript. S.D.E. edited the manuscript.
References
- 1000 Genomes Project Consortium. Abecasis GR, Auton A, Brooks LD, DePristo MA, Durbin RM, Handsaker RE, Kang HM, Marth GT, McVean GA. An integrated map of genetic variation from 1,092 human genomes. Nature. 2012;491:56–65. doi: 10.1038/nature11632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andersen OM, Reiche J, Schmidt V, Gotthardt M, Spoelgen R, Behlke J, von Arnim CA, Breiderhoff T, Jansen P, Wu X, et al. Neuronal sorting protein-related receptor sorLA/LR11 regulates processing of the amyloid precursor protein. Proc Natl Acad Sci USA. 2005;102:13461–13466. doi: 10.1073/pnas.0503689102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bertram L, McQueen MB, Mullin K, Blacker D, Tanzi RE. Systematic meta-analyses of Alzheimer disease genetic association studies: the AlzGene database. Nat Genet. 2007;39:17–23. doi: 10.1038/ng1934. [DOI] [PubMed] [Google Scholar]
- Bettens K, Brouwers N, Engelborghs S, De Deyn PP, Van Broeckhoven C, Sleegers K. SORL1 is genetically associated with increased risk for late-onsetAlzheimer diseaseinthe Belgian population. HumMutat. 2008;29:769–770. doi: 10.1002/humu.20725. [DOI] [PubMed] [Google Scholar]
- Caglayan S, Takagi-Niidome S, Liao F, Carlo AS, Schmidt V, Burgert T, Kitago Y, Fuchtbauer EM, Fuchtbauer A, Holtzman DM, et al. Lysosomal sorting of amyloid-β by the SORLA receptor is impaired by a familial Alzheimer’s disease mutation. Sci Transl Med. 2014;6:223ra220. doi: 10.1126/scitranslmed.3007747. [DOI] [PubMed] [Google Scholar]
- Cao L, Tan L, Jiang T, Zhu XC, Yu JT. Induced Pluripotent Stem Cells for Disease Modeling and Drug Discovery in Neurodegenerative Diseases. Mol Neurobiol. 2014 doi: 10.1007/s12035-014-8867-6. Published online August 23 2014 http://dx.doi.org/10.1007/s12035-014-8867-6. [DOI] [PubMed]
- Cheng PL, Song AH, Wong YH, Wang S, Zhang X, Poo MM. Self-amplifying autocrine actions of BDNF in axon development. Proc Natl Acad Sci USA. 2011;108:18430–18435. doi: 10.1073/pnas.1115907108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chouraki V, Seshadri S. Genetics of Alzheimer’s disease. Adv Genet. 2014;87:245–294. doi: 10.1016/B978-0-12-800149-3.00005-6. [DOI] [PubMed] [Google Scholar]
- Cruchaga C, Karch CM, Jin SC, Benitez BA, Cai Y, Guerreiro R, Harari O, Norton J, Budde J, Bertelsen S, et al. Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer’s disease. Nature. 2014;505:550–554. doi: 10.1038/nature12825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dodson SE, Gearing M, Lippa CF, Montine TJ, Levey AI, Lah JJ. LR11/SorLA expression is reduced in sporadic Alzheimer disease but not in familial Alzheimer disease. J Neuropathol Exp Neurol. 2006;65:866–872. doi: 10.1097/01.jnen.0000228205.19915.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dodson SE, Andersen OM, Karmali V, Fritz JJ, Cheng D, Peng J, Levey AI, Willnow TE, Lah JJ. Loss of LR11/SORLA enhances early pathology in a mouse model of amyloidosis: evidence for a proximal role in Alzheimer’s disease. J Neurosci. 2008;28:12877–12886. doi: 10.1523/JNEUROSCI.4582-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gatz M, Reynolds CA, Fratiglioni L, Johansson B, Mortimer JA, Berg S, Fiske A, Pedersen NL. Role of genes and environments for explaining Alzheimer disease. Arch Gen Psychiatry. 2006;63:168–174. doi: 10.1001/archpsyc.63.2.168. [DOI] [PubMed] [Google Scholar]
- Goldstein LS, Reyna S, Woodruff G. Probing the Secrets of Alzheimer’s Disease Using Human-induced Pluripotent Stem Cell Technology. Neurotherapeutics. 2015;12:121–125. doi: 10.1007/s13311-014-0326-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gore A, Li Z, Fung HL, Young JE, Agarwal S, Antosiewicz-Bourget J, Canto I, Giorgetti A, Israel MA, Kiskinis E, et al. Somatic coding mutations in human induced pluripotent stem cells. Nature. 2011;471:63–67. doi: 10.1038/nature09805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grear KE, Ling IF, Simpson JF, Furman JL, Simmons CR, Peterson SL, Schmitt FA, Markesbery WR, Liu Q, Crook JE, et al. Expression of SORL1 and a novel SORL1 splice variant in normal and Alzheimers disease brain. Mol Neurodegener. 2009;4:46. doi: 10.1186/1750-1326-4-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griciuc A, Serrano-Pozo A, Parrado AR, Lesinski AN, Asselin CN, Mullin K, Hooli B, Choi SH, Hyman BT, Tanzi RE. Alzheimer’s disease risk gene CD33 inhibits microglial uptake of amyloid beta. Neuron. 2013;78:631–643. doi: 10.1016/j.neuron.2013.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hermey G. The Vps10p-domain receptor family. Cell Mol Life Sci. 2009;66:2677–2689. doi: 10.1007/s00018-009-0043-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holton P, Ryten M, Nalls M, Trabzuni D, Weale ME, Hernandez D, Crehan H, Gibbs JR, Mayeux R, Haines JL, et al. Alzheimer’s Disease Genetics Consortium . Initial assessment of the pathogenic mechanisms of the recently identified Alzheimer risk Loci. Ann Hum Genet. 2013;77:85–105. doi: 10.1111/ahg.12000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hunter K, Hölscher C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 2012;13:33. doi: 10.1186/1471-2202-13-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, Herrera C, Hefferan MP, Van Gorp S, Nazor KL, Boscolo FS, et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature. 2012;482:216–220. doi: 10.1038/nature10821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karch CM, Jeng AT, Nowotny P, Cady J, Cruchaga C, Goate AM. Expression of novel Alzheimer’s disease risk genes in control and Alzheimer’s disease brains. PLoS ONE. 2012;7:e50976. doi: 10.1371/journal.pone.0050976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kondo T, Asai M, Tsukita K, Kutoku Y, Ohsawa Y, Sunada Y, Imamura K, Egawa N, Yahata N, Okita K, et al. Modeling Alzheimer’s disease with iPSCs reveals stress phenotypes associated with intracellular Aβ and differential drug responsiveness. Cell Stem Cell. 2013;12:487–496. doi: 10.1016/j.stem.2013.01.009. [DOI] [PubMed] [Google Scholar]
- Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, DeStafano AL, Bis JC, Beecham GW, Grenier-Boley B, et al. European Alzheimer’s Disease Initiative (EADI); Genetic, Environmental Risk in Alzheimer’s Disease; Alzheimer’s Disease Genetic Consortium; Cohorts for Heart, Aging Research in Genomic Epidemiology . Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet. 2013;45:1452–1458. doi: 10.1038/ng.2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JH, Cheng R, Schupf N, Manly J, Lantigua R, Stern Y, Rogaeva E, Wakutani Y, Farrer L, St George-Hyslop P, Mayeux R. The association between genetic variants in SORL1 and Alzheimer disease in an urban, multiethnic, community-based cohort. Arch Neurol. 2007;64:501–506. doi: 10.1001/archneur.64.4.501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levy S, Sutton G, Ng PC, Feuk L, Halpern AL, Walenz BP, Axelrod N, Huang J, Kirkness EF, Denisov G, et al. The diploid genome sequence of an individual human. PLoS Biol. 2007;5:e254. doi: 10.1371/journal.pbio.0050254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Wang H, Muffat J, Cheng AW, Orlando DA, Lovén J, Kwok SM, Feldman DA, Bateup HS, Gao Q, et al. Global transcriptional and translational repression in human-embryonic-stem-cell-derived Rett syndrome neurons. Cell Stem Cell. 2013;13:446–458. doi: 10.1016/j.stem.2013.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma QL, Galasko DR, Ringman JM, Vinters HV, Edland SD, Pomakian J, Ubeda OJ, Rosario ER, Teter B, Frautschy SA, Cole GM. Reduction of SorLA/LR11, a sorting protein limiting beta-amyloid production, in Alzheimer disease cerebrospinal fluid. Arch Neurol. 2009;66:448–457. doi: 10.1001/archneurol.2009.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McCarthy JJ, Saith S, Linnertz C, Burke JR, Hulette CM, Welsh-Bohmer KA, Chiba-Falek O. The Alzheimer’s associated 5′ region of the SORL1 gene cis regulates SORL1 transcripts expression. Neurobiol Aging. 2012;33:e1–e8. doi: 10.1016/j.neurobiolaging.2010.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM. Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology. 1984;34:939–944. doi: 10.1212/wnl.34.7.939. [DOI] [PubMed] [Google Scholar]
- Mörwald S, Yamazaki H, Bujo H, Kusunoki J, Kanaki T, Seimiya K, Morisaki N, Nimpf J, Schneider WJ, Saito Y. A novel mosaic protein containing LDL receptor elements is highly conserved in humans and chickens. Arterioscler Thromb Vasc Biol. 1997;17:996–1002. doi: 10.1161/01.atv.17.5.996. [DOI] [PubMed] [Google Scholar]
- Motoi Y, Aizawa T, Haga S, Nakamura S, Namba Y, Ikeda K. Neuronal localization of a novel mosaic apolipoprotein E receptor, LR11, in rat and human brain. Brain Res. 1999;833:209–215. doi: 10.1016/s0006-8993(99)01542-5. [DOI] [PubMed] [Google Scholar]
- Nagahara AH, Merrill DA, Coppola G, Tsukada S, Schroeder BE, Shaked GM, Wang L, Blesch A, Kim A, Conner JM, et al. Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat Med. 2009;15:331–337. doi: 10.1038/nm.1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olgiati P, Politis AM, Papadimitriou GN, De Ronchi D, Serretti A. Genetics of late-onset Alzheimer’s disease: update from the alzgene database and analysis of shared pathways. Int J Alzheimers Dis. 2011;2011:832379. doi: 10.4061/2011/832379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parthsarathy V, Hölscher C. Chronic treatment with the GLP1 analogue liraglutide increases cell proliferation and differentiation into neurons in an AD mouse model. PLoS ONE. 2013;8:e58784. doi: 10.1371/journal.pone.0058784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pottier C, Hannequin D, Coutant S, Rovelet-Lecrux A, Wallon D, Rousseau S, Legallic S, Paquet C, Bombois S, Pariente J, et al. High frequency of potentially pathogenic SORL1 mutations in autosomal dominant early-onset Alzheimer disease. Mol Psychiatry. 2012;17:875–879. doi: 10.1038/mp.2012.15. [DOI] [PubMed] [Google Scholar]
- R Development Core Team. R: A language and environment for statistical computing. Vienna: R Foundation for Statistical Computing; 2008. [Google Scholar]
- Reitz C. The role of intracellular trafficking and the VPS10d receptors in Alzheimer’s disease. Future Neurol. 2012;7:423–431. doi: 10.2217/fnl.12.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reitz C, Cheng R, Rogaeva E, Lee JH, Tokuhiro S, Zou F, Bettens K, Sleegers K, Tan EK, Kimura R, et al. Genetic Environmental Risk in Alzheimer Disease 1 Consortium . Meta-analysis of the association between variants in SORL1 and Alzheimer disease. Arch Neurol. 2011;68:99–106. doi: 10.1001/archneurol.2010.346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rogaeva E, Meng Y, Lee JH, Gu Y, Kawarai T, Zou F, Katayama T, Baldwin CT, Cheng R, Hasegawa H, et al. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat Genet. 2007;39:168–177. doi: 10.1038/ng1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rohe M, Synowitz M, Glass R, Paul SM, Nykjaer A, Willnow TE. Brain-derived neurotrophic factor reduces amyloidogenic processing through control of SORLA gene expression. J Neurosci. 2009;29:15472–15478. doi: 10.1523/JNEUROSCI.3960-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rovelet-Lecrux A, Hannequin D, Raux G, Le Meur N, Laquerrière A, Vital A, Dumanchin C, Feuillette S, Brice A, Vercelletto M, et al. APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nat Genet. 2006;38:24–26. doi: 10.1038/ng1718. [DOI] [PubMed] [Google Scholar]
- Scherzer CR, Offe K, Gearing M, Rees HD, Fang G, Heilman CJ, Schaller C, Bujo H, Levey AI, Lah JJ. Loss of apolipoprotein E receptor LR11 in Alzheimer disease. Arch Neurol. 2004;61:1200–1205. doi: 10.1001/archneur.61.8.1200. [DOI] [PubMed] [Google Scholar]
- Schmidt V, Baum K, Lao A, Rateitschak K, Schmitz Y, Teichmann A, Wiesner B, Petersen CM, Nykjaer A, Wolf J, et al. Quantitative modelling of amyloidogenic processing and its influence by SORLA in Alzheimer’s disease. EMBO J. 2012;31:187–200. doi: 10.1038/emboj.2011.352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shi Y, Kirwan P, Smith J, MacLean G, Orkin SH, Livesey FJ. A human stem cell model of early Alzheimer’s disease pathology in Down syndrome. Sci Transl Med. 2012;4:124ra129. doi: 10.1126/scitranslmed.3003771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soldner F, Laganière J, Cheng AW, Hockemeyer D, Gao Q, Alagappan R, Khurana V, Golbe LI, Myers RH, Lindquist S, et al. Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations. Cell. 2011;146:318–331. doi: 10.1016/j.cell.2011.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vardarajan BN, Zhang Y, Lee JH, Cheng R, Bohm C, Ghani M, Reitz C, Reyes-Dumeyer D, Shen Y, Rogaeva E, et al. Coding mutations in SORL1 and Alzheimer’s disease. Ann Neurol. 2015;77:215–227. doi: 10.1002/ana.24305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen Y, Miyashita A, Kitamura N, Tsukie T, Saito Y, Hatsuta H, Murayama S, Kakita A, Takahashi H, Akatsu H, et al. SORL1 is genetically associated with neuropathologically characterized late-onset Alzheimer’s disease. J Alzheimers Dis. 2013;35:387–394. doi: 10.3233/JAD-122395. [DOI] [PubMed] [Google Scholar]
- Woodruff G, Young JE, Martinez FJ, Buen F, Gore A, Kinaga J, Li Z, Yuan SH, Zhang K, Goldstein LS. The presenilin-1 ΔE9 mutation results in reduced γ-secretase activity, but not total loss of PS1 function, in isogenic human stem cells. Cell Rep. 2013;5:974–985. doi: 10.1016/j.celrep.2013.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan SH, Martin J, Elia J, Flippin J, Paramban RI, Hefferan MP, Vidal JG, Mu Y, Killian RL, Israel MA, et al. Cell-surface marker signatures for the isolation of neural stem cells, glia and neurons derived from human pluripotent stem cells. PLoS ONE. 2011;6:e17540. doi: 10.1371/journal.pone.0017540. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.





