Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 May 10.
Published in final edited form as: Neuroscience. 2022 May 13;518:10–26. doi: 10.1016/j.neuroscience.2022.05.006

Molecular insights into cell type-specific roles in Alzheimer’s disease: human induced pluripotent stem cell-based disease modeling

Wenhui Qu 1,2, Peter Canoll 1, Gunnar Hargus 1,2,*
PMCID: PMC9974106  NIHMSID: NIHMS1807305  PMID: 35569647

Abstract

Alzheimer’s disease (AD) is the most common cause of dementia resulting in widespread degeneration of the central nervous system with severe cognitive impairment. Despite the devastating toll of AD, the incomplete understanding of the complex molecular mechanisms hinders the expeditious development of effective cures. Emerging evidence from animal studies has shown that different brain cell types play distinct roles in the pathogenesis of AD. Glutamatergic neurons are preferentially affected in AD and pronounced gliosis contributes to the progression of AD in both a cell-autonomous and a non-cell-autonomous manner. Much has been discovered through genetically modified animal models, yet frequently failed translational attempts to clinical applications call for better disease models. Emerging evidence supports the significance of human-induced pluripotent stem cell (iPSC) derived brain cells in modeling disease development and progression, opening new avenues for the discovery of molecular mechanisms. This review summarizes the function of different cell types in the pathogenesis of AD, such as neurons, microglia, and astrocytes, and recognizes the potential of utilizing the rapidly growing iPSC technology in modeling AD.

Introduction

Alzheimer’s disease (AD) is a fatal neurodegenerative disease characterized by progressive loss of cognitive function including learning and memory (Tarawneh and Holtzman, 2012). More than 46 million people suffer from dementia worldwide and AD accounts for at least half of these patients with expected growing numbers in future decades due to extended life expectancy. Thus, AD places a severe burden on patients and families as well as healthcare systems as society ages. Taking advantage of human genetic studies, rodent models carrying AD mutations successfully mimic the age-related neurodegenerative aspect of AD and they have provided invaluable molecular insights into cell type-specific mechanisms of AD pathogenesis (Penney et al., 2020). However, attempts to translate these insights from rodent studies to clinical trials in AD patients have failed, highlighting the need for better models (Ceyzeriat et al., 2020). The expression and cellular regulation of several key AD-related proteins differ significantly between humans and rodents, which may have contributed to negative outcomes (Maloney et al., 2007; Hernández et al., 2020; Zhou et al., 2020). Emerging evidence has suggested the importance of utilizing human cells, such as iPSC-derived brain cells, to model human neurodegenerative diseases (Penney et al., 2020). The iPSC technology was introduced more than a decade ago and allows for the generation of pluripotent stem cells from differentiated patient-derived cells, such as fibroblast from a skin biopsy or peripheral blood mononuclear cells from a blood draw, by overexpressing the reprogramming factors Oct4, Klf4, Sox2 and c-Myc in these cells (Takahashi et al., 2007). By applying robust and improved maturation protocols, these human iPSCs can be efficiently differentiated into various cell types of interest such as cortical neurons (Shi et al., 2012; Zhang et al., 2013), astrocytes (Serio et al., 2013; Hallmann et al., 2017; Zhao et al., 2017; Guttikonda et al., 2021), oligodendrocytes (Ehrlich et al., 2017) and microglia (Muffat et al., 2016; Haenseler et al., 2017; McQuade et al., 2018; Marton et al., 2019) (Figure 1). Thus, patient-derived iPSCs have been successfully used to model various neurodegenerative diseases including primary tauopathies (Lines et al., 2020; Kuhn et al., 2021) and secondary tauopathies such as AD (Yagi et al., 2011; Israel et al., 2012; Lee et al., 2020; Penney et al., 2020; Cenini et al., 2021). The application of genome editing tools, such as CRISPR/Cas9 to generate isogenic, gene-corrected control cells or to introduce AD-associated mutations in control cells, comprising an additional powerful approach to interrogate the molecular mechanisms of tauopathies in different human brain cell types in vitro (Jehuda et al., 2018). Furthermore, mixed neuron-glia co-culture systems have been established to study disease pathogenesis (Jehuda et al., 2018; Lee et al., 2020; Penney et al., 2020; Cenini et al., 2021) and rapid growing 3D/brain organoid culture techniques and human-mouse chimeric disease models provide prevailing disease modeling systems to study AD in a 3D/in vivo environment (Zhang et al., 2014; Purhonen et al., 2020; Sharma et al., 2020; Xu et al., 2020; Preman et al., 2021) (Figure 1).

Figure 1: Modeling AD with patient iPSCs.

Figure 1:

AD patient or healthy control-derived iPSCs can be differentiated into different brain cell types including neurons, microglia, astrocytes, and oligodendrocytes. In vitro and in vivo AD modeling strategies can be utilized to model AD and they can be applied for drug screening. “?” indicates available technology yet to be used in AD research.

This review summarizes key findings of the molecular mechanisms of different brain cell types in contributing to the pathogenesis of AD. Reviewing mechanistic studies in genetically modified animal models and the phenotypes of iPSC-derived brain cell types, this review provides molecular insights into the pathogenesis of AD and emphasizes the promising potential of utilizing iPSC technology for better translational approaches.

Alzheimer’s disease

AD is characterized by a progressive decline in at least two cognitive domains that commonly include episodic memory and executive function (Tarawneh and Holtzman, 2012). Pathological features of AD include extracellular deposition of amyloid plaques and formation of intracellular neurofibrillary tangles containing hyperphosphorylated tau (p-tau), accompanied by extensive gliosis, synaptic dysfunction, and neuronal loss (Serrano-Pozo et al., 2011). Amyloid plaques are formed by the Aβ peptide, which is sequentially cleaved from β-amyloid precursor protein (APP) by β- and γ-secretase that generates toxic Aβ species (Chow et al., 2010; Bernabeu-Zornoza et al., 2019). APP can also be sequentially cleaved by α- and γ-secretase, resulting in the generation of non-amyloidogenic fragments (Chow et al., 2010). Aβ is mainly produced in neurons due to the abundant expression of APP and β-secretase (Zhou et al., 2011; Das and Yan, 2017). Among all the Aβ species, Aβ42 and Aβ40 are the most common isoforms present in amyloid plaques of human AD brains and are a major focus of the research effort in the field of AD (Braak and Braak, 1991; Hardy and Allsop, 1991; Serrano-Pozo et al., 2011; Masters and Selkoe, 2012). Aβ42 has a higher rate of fibrillization and insolubility and is deposited in dense-core plaques in the brain parenchyma. The more soluble Aβ40 is the most abundantly produced Aβ species and is the major constituent of amyloid deposition in blood vessel walls leading to cerebral amyloid angiopathy (CAA). A decreased ratio of Aβ42/Aβ40 in cerebrospinal fluid (CSF) is a strong biomarker for AD, reflecting reduced Aβ clearance through CSF and increased accumulation of amyloid plaques in the brain parenchyma (Shaw et al., 2009). Emerging evidence supports the idea that soluble Aβ42 oligomers are more neurotoxic compared with Aβ deposited in amyloid plaques, interrupting glutamatergic neurotransmission, inducing synapse loss, and contributing to dysregulation of synaptic plasticity in AD (Benilova et al., 2012). In addition to the toxicity induced by Aβ, several lines of research also support mechanistic roles of altered APP metabolism and loss-of-function of γ-secretase in contributing to the pathogenesis of AD (Shen and Kelleher, 2007; Kametani and Hasegawa, 2018).

Tau pathology usually follows Aβ pathology in AD and can be induced by Aβ (Stancu et al., 2014). Tau is a microtubule-associated protein that is encoded by the MAPT gene. Under physiological conditions, tau plays important role in microtubule stabilization, in regulating the dynamics of microtubule assembly, and in assisting axonal transportation (Mietelska-Porowska et al., 2014). Alternative splicing of the exons 2, 3, and 10 of the MAPT gene produces six tau isoforms (Trabzuni et al., 2012). Splicing of exons 2 and 3 produces tau proteins with 0–2N domains and splicing of exon 10 determines the expression of tau with three or four microtubule-binding domains (3R- or 4R-tau) (Park et al., 2016). Tau pathology propagates in a prion-like manner following a stereotypical pattern during the pathogenesis of AD, striking earliest in the locus coeruleus of the brain stem and the integrity of which indicates the neuropathology and cognitive function of AD patients (Jacobs et al., 2021). From the locus coeruleus, tau pathology then emerges in the entorhinal cortex, spreading to the hippocampus and neocortex (Braak and Braak, 1991; Clavaguera et al., 2015). A recent study has identified the Wolframin-1 expressing neurons in the entorhinal cortex that are responsible for propagating toxic tau to hippocampal neurons (Delpech et al., 2021). The severity of tau pathology closely correlates with neurodegeneration and cognitive decline in AD, further highlighting the neurotoxicity of pathological tau (Jack Jr et al., 2010). Interestingly, a recent report showed that the replication rather than the spreading of toxic tau between brain regions are the main driver of tau accumulation in AD (Meisl et al., 2021). Blood-brain barrier (BBB) dysfunction is another key pathological feature of AD and emerging evidence indicated that the degeneration of pericytes of BBB is associated with neurovascular malfunction and exacerbated Aβ and tau pathology (Sagare et al., 2013; Halliday et al., 2016; Sweeney et al., 2016). Interestingly, Aβ can also signal to pericytes to restrict capillaries in AD which correlates with Aβ deposition (Nortley et al., 2019). Important work has been done to convert adult human brain pericytes into neurons, which could potentially be applied as an AD treatment strategy (Karow et al., 2018).

The two main forms of AD include early-onset familial AD (FAD), which is caused by known mutations involved in Aβ production, and sporadic AD (SAD) (Hardy and Allsop, 1991; Chow et al., 2010; Holtzman et al., 2011). Mutations in genes involved in Aβ production leading to FAD include the APP gene and γ-secretase catalytic subunits encoding genes, PSEN1 and PSEN2, encoding presenilin 1 and presenilin 2, respectively (Goate et al., 1991; Levy-Lahad et al., 1995; Sherrington et al., 1995; Haass et al., 2012). More than 200 pathogenic mutations have been described in PSEN1, while around 30 and 20 mutations have been reported in the APP and PSEN2 loci, respectively (Lanoiselee et al., 2017). Pathological mutations of these FAD genes all increase either total Aβ42 or the ratio of Aβ42/Aβ40 (Fernandez et al., 2014). FAD consists of around 1–5% of all AD cases and the majority of AD cases are sporadic. In an attempt to elucidate the molecular pathways that contribute to the pathogenesis of AD, genome-wide association studies (GWAS) were conducted and have identified more than 40 AD risk genes, including APOE4, TREM2, ABCA7, CD33, and SORL1 which are highly expressed in glia (Kamboh et al., 2012; Tábuas-Pereira et al., 2020). These findings underscore a highly relevant role of non-cell-autonomous mechanisms, potentially involving astrocytes and microglia, that contribute to neurodegeneration in AD. Both FAD and SAD cases present with an accumulation of amyloid pathology that precedes tau pathology, followed by cognitive impairment (Holtzman et al., 2011; Selkoe and Hardy, 2016).

Neurons

Early studies have provided evidence that human iPSC-derived neurons express key components and regulators of the APP processing machinery such as β- and γ-secretase, as well as different APP isoforms and several isoforms of Aβ including Aβ37, Aβ38, Aβ39, Aβ40, Aβ42, and N-terminally truncated Aβ2–40 (Koch et al., 2012; Bergström et al., 2016). In addition, the formation of the different tau isoforms, both 3R- and 4R-tau, in human iPSC-derived neurons follows a developmental pattern (Iovino et al., 2015; Sposito et al., 2015). While the fetal 3R tau isoform appears earliest during differentiation, all 6 isoforms are expressed in mature neurons after several months of differentiation in vitro. Thus, iPSCs are suitable cell sources to study AD-associated pathologic changes in human neurons (Table 1).

Table 1:

Major AD disease phenotypes in iPSC-derived neurons

Model system Mutations/variants Phenotypes References*
2D culture FAD mutations including
APP Lon
APP Swe
APP Dup
PSEN1 M146L
PSEN1 A246E
PSEN1 E120K
PSEN2 N141I

SAD variant of APOE4

WT exposed to exogenous Aβ
  • Increased total Aβ production or increased Aβ42/Aβ40 ratio

  • Increased total tau or p-tau

(Israel et al., 2012)
(Muratore et al., 2014)
(Sproul et al., 2014)
(Yang et al., 2017)
(Ortiz-Virumbrales et al., 2017)
(Lin et al., 2018)
(Bassil et al., 2021)
APP Swe
PSEN1 ΔE9
PSEN1 M146V
  • Enhanced excitability

  • Synaptic dysfunction

(Ghatak et al., 2019)
PSEN1 A246E
PSEN1 E120K
PSEN1 R307S
  • Increased oxidative stress

  • Lysosomal dysregulation

  • DNA damage

(Martin-Maestro et al., 2017)
(Yang et al., 2017)
(Wezyk et al., 2018)
(Li et al., 2018)
3D culture Overexpression of APP Swe, APP Lon, PSEN1 ΔE9
  • Formation of Aβ plaque-like structures

  • Formation of neurofibrillary tangles

(Choi et al., 2014)
(Park et al., 2018)
(Kwak et al., 2020)
Organoid APP Swe
PSEN1 M146V
APOE4
  • Increased total Aβ production or increased Aβ42/Aβ40 ratio

  • Increased p-tau

  • Lysosomal dysregulation

  • Synaptic dysfunction

(Raja et al., 2016)
(Gonzalez et al., 2018)
(Lin et al., 2018)
(Zhao et al., 2020)
Xenotransplant in APP/PS1 mice WT
  • Amyloid plaque-associated neurite dystrophy

  • Increased p-tau and tau conformational changes

  • Decreased neuronal survival with increased plaque formation within grafts

(Espuny-Camacho et al., 2017)
Xenotransplant in APOE4 KI mice APOE4
  • Elevated production of Aβ aggregates

  • Dysregulated gene expression profiles including p53 signaling, cellular senescence pathway, and apoptosis

(Najm et al., 2020)
*

Selected references

Reports on mixed neuron-glial co-cultures and additional 3D cultures are listed in table 2.

Excitatory neurons are preferentially affected in AD. The increased rate of seizures in Alzheimer’s patients hints towards a disruption of the excitatory/inhibitory (E/I) balance in AD brains (Born, 2015). Multiple AD mouse models carrying FAD or SAD mutations/variance show dysregulation of synaptic transmission and prominent neuron hyperexcitability (Klein et al., 2014; Kazim et al., 2017; Varela et al., 2019; Qu and Li, 2020; Müller et al., 2021). Increased electrophysiological E/I balance has recently been demonstrated in the forebrain circuits of post-mortem human AD brains (Lauterborn et al., 2021). Differential gene expression analyses and protein assays also confirmed an increased expression of excitatory synaptic markers (Lauterborn et al., 2021). Interestingly, another report showed that excitatory and inhibitory neurons exhibit different vulnerabilities to tau pathology in human AD brains (Fu et al., 2019). Furthermore, single-nucleus RNA-seq (snRNA-seq) analysis of human AD brains revealed a tau homeostasis signature in excitatory neurons and identified BCL-2 associated athanogene3 (BAG3), an autophagy facilitator, as a master regulator of this tau homeostatic gene signature in these neurons (Fu et al., 2019). Increased neuronal activity enhances the propagation of tau and facilitates the development of tau pathology in a tauopathy mouse model as well as in human iPSC-derived cortical neurons (Wu et al., 2016; Lauterborn et al., 2021). Recently, the low-density lipoprotein receptor-related protein 1 (LRP1) has been identified as the receptor that controls the endocytosis and spread of tau, as also shown in human iPSC-derived neurons (Rauch et al., 2020). Consistent with these studies, ablation of tau in mice shows reduced baseline activity of excitatory neurons and enhanced excitability of inhibitory neurons, suggesting that tau plays a differential role in excitatory and inhibitory neurons (Chang et al., 2021). An abnormally enhanced electrophysiological activity can be recapitulated in human iPSC-derived cortical neurons and organoids carrying FAD mutations in PSEN1 (PS1 ΔE9 mutation and M146V mutation) and APP (KM670/671NL; Swedish mutation), when compared with their isogenic WT controls (Ghatak et al., 2019). The aberrant neuronal activity in iPSC-derived neurons correlates with ion channel dysfunction and reduced neurite length, closely mimicking the early synaptic dysfunction and hyperexcitability in human AD brains (Ghatak et al., 2019). While it may be difficult to recapitulate age-dependent neurodegenerative phenotypes in iPSC-derived neurons, a recent report showed significant overlap of gene expression and correlation of Aβ and tau species between iPSC-derived neurons and the brains of their AD iPSC donors, highlighting the suitability of using iPSC to interrogate the molecular mechanisms of AD pathogenesis in humans (Lagomarsino et al., 2021).

Characterization of iPSC-derived neurons that carry FAD mutations has provided insights into the molecular mechanisms underlying AD pathogenesis. Accumulating evidence suggests that Aβ triggers pathological tau formation and accumulation in the pathogenesis of AD in animal models (Bloom, 2014), and this pathological feature can be recapitulated in iPSC-derived neurons. iPSC-derived forebrain neurons harboring the APP London mutation (V717I) demonstrate altered APP cleavage and increased production of Aβ, leading to elevated levels of total tau and p-tau (Muratore et al., 2014). Increased levels of tau can be rescued by early Aβ antibody treatment. These findings suggested that tau pathology is downstream of Aβ raising the possibility that targeting Aβ early in the course of AD may be a promising therapeutic strategy. Similarly, increased levels of Aβ-induced p-tau have been reported in WT neurons and in several additional studies on iPSC-derived neural progenitor cells (NPCs) and neurons that carry FAD or SAD mutations/variants including FAD mutations in the APP, PSEN1, PSEN2, and APOE (APOE4) loci (Israel et al., 2012; Sproul et al., 2014; Ortiz-Virumbrales et al., 2017; Yang et al., 2017; Lin et al., 2018; Bassil et al., 2021).

AD-like pathology is observed in individuals with Down syndrome (DS) and this pathology is attributed to the supernumerary copy of the APP gene (Wisniewski et al., 1985). Interestingly, deletion of one of these copies in DS patient-derived neurons corrects Aβ pathology and rescues altered neuronal gene expression but is unable to alter levels of p-tau or apoptosis, challenging the view that Aβ-related pathology is the sole contributor to p-tau pathology in DS (Ovchinnikov et al., 2018).

In addition to Aβ and tau pathology, iPSC-derived neurons from FAD patients also show several perturbed cellular pathways including altered cellular trafficking and lysosomal degradation, increased oxidative stress, and elevated DNA damage, as similarly shown in rodent models of AD (Martin-Maestro et al., 2017; Yang et al., 2017; Li et al., 2018; Wezyk et al., 2018) (Table 1). Interestingly, an accelerated neural differentiation and an impaired proliferation of NPCs were described in iPSC neural cells derived from SAD cases without known AD-related mutations and in iPSC neural cells with introduced APOE4 (Meyer et al., 2019). This phenotype was linked to a loss of function of REST, a transcription factor, that showed impaired nuclear translocation and chromatin binding in both neural cell types, suggesting a shared phenotype in regards to epigenetic dysregulation in these cells (Meyer et al., 2019).

In conventional 2D culture, human AD neurons contain elevated levels of p-tau but they do not form amyloid plaques or neurofibrillary tangles, which are the key pathological features in AD. Using a 3D culture system, human neuronal progenitor cells overexpressing FAD APP and PSEN1 mutations show an elevated Aβ42/Aβ40 ratio that drives the formation of neurofibrillary tangles (Choi et al., 2014; Park et al., 2018; Kwak et al., 2020), indicating the importance of a 3D environment in recapitulating key AD pathological features (Table 1). A 3D environment can also be achieved by the formation of brain organoids which have been applied to model AD. As such, FAD and SAD iPSC-derived brain organoids demonstrated accumulation of β-amyloid, the elevation of p-tau, lysosomal dysregulation, and synaptic dysfunction (Raja et al., 2016; Gonzalez et al., 2018; Lin et al., 2018; Zhao et al., 2020) (Table 1). A recent report generated hippocampal spheroids from FAD patient-derived iPSC that exhibit AD pathologic changes with an increased Aβ42/Aβ40 ratio, elevated p-tau, reduced expression of synaptic makers as wells as electrophysiologically altered synaptic transmission (Pomeshchik et al., 2020). A limitation of utilizing brain organoids for disease modeling used to be the relative lack of cells that are not of ectodermal origin including microglia and vasculature (Papaspyropoulos et al., 2020). Microglia are innate immune cells of the brain that play an important role in the pathogenesis of AD and will be discussed in a later section. Vasculature delivers oxygen and endothelial cells form part of the BBB, which is dysfunctional in AD (Sweeney et al., 2018). Emerging efforts are invested in vascularizing brain organoids with the help of microfluidic devices for potential applications in vitro and in immunosuppressed mice in vivo (Daviaud et al., 2018; Cakir et al., 2019; Matsui et al., 2021; Zhang et al., 2021). Future studies may take advantage of these sophisticated 3D models to study the pathogenesis of AD.

Human-mouse chimeric transplantation models of AD have been established using human iPSC-derived neurons as single-cell suspensions (Espuny-Camacho et al., 2017; Najm et al., 2020). WT iPSC-derived human neuronal precursor cells transplanted into immunocompromised rodent brains mature in vivo and form connections and active synapses that appeared more mature compared to those in vitro-cultured cells (Gaspard et al., 2008; Espuny-Camacho et al., 2013). In a later study, human WT iPSC-derived neuronal progenitor cells were transplanted into an immunosuppressed mouse model of AD, in which injected cells differentiated into mature neurons and were exposed to Aβ plaques (Espuny-Camacho et al., 2017). Notably, grafted neurons exhibited key AD pathological features including amyloid plaque-associated neurite dystrophy, abnormal tau phosphorylation, and conformational changes, decreased neuronal survival as well as the acquisition of an AD transcriptome signature (Espuny-Camacho et al., 2017).

Neurons that carry AD-associated variants have also been explored in a human-mouse chimeric model of AD. Human iPSC-derived neurons carrying APOE4 were injected into human APOE4 knock-in (KI) mouse brains and exhibited dysregulated gene expression profiles related to p53 signaling, cellular senescence, and apoptosis (Najm et al., 2020). Interestingly, APOE4 iPSC-derived neurons produce more Aβ in vitro compared with APOE3 but neither form Aβ aggregates in vitro (Wang et al., 2018). In contrast, both transplanted APOE3 and transplanted APOE4 human neurons produced Aβ aggregates in vivo (Najm et al., 2020). Furthermore, APOE4 human neurons produced more Aβ aggregates in APOE4 KI mice than in APOE3 KI mice, which was attributed to impaired phagocytosis of Aβ by APOE4 murine microglia (Najm et al., 2020).

While it is important to continously optimize differentiation protocols to shorten the oftentimes time-intensive and laborious differentiation of iPSCs into mature neurons, these studies clearly highlight that iPSC-derived neurons are suitable to study the pathogenesis of AD. They also suggest that characterizing iPSC-derived neurons in an in vivo environment might be beneficial to study mechanisms of disease development in AD. Future studies could utilize these models to further interrogate cell-cell interactions by co-transplanting different human iPSC-derived brain cells, for instance, neurons with glial cells such as astrocytes or microglia.

Microglia

Microglia are innate immune cells of the brain that serve important roles throughout life, including facilitating neurodevelopment, modulating synaptic plasticity, and responding to injury and pathological insults to the central nervous system (Ransohoff and El Khoury, 2015). Human genetic studies have identified several risk genes for AD that are highly expressed in microglia including APOE, TREM2, and CD33, highlighting the significant contribution of microglial dysfunction in the pathogenesis of AD (Kamboh et al., 2012; Dos Santos et al., 2017; Tábuas-Pereira et al., 2020). In healthy brains, microglia maintain a homostatic state defined by their unique gene signature. In AD, microglia alter transcriptomic programs that transform them into functional activation states, including the state of disease-associated microglia (DAM) (Keren-Shaul et al., 2017; Krasemann et al., 2017). Microgliosis is prominent especially around amyloid plaques in the brain where microglia cluster around the plaques and form a barrier to prevent their expansion (Casali et al., 2020). Amyloid plaques facilitate the accumulation of p-tau in dystrophic neurites which are a component of neuritic plaques (NPs) (He et al., 2018), and at the initial tau seeding stage, microglia regulate the spread and accumulation of toxic p-tau in these NPs (Leyns et al., 2019; Delizannis et al., 2021; Gratuze et al., 2021).

Tau released from neuronal synapses can be phagocytosed by microglia and is sensed by the polyglutamine binding protein 1 (PQBP1) on microglia that triggers a microglial inflammatory response contributing to cognitive impairment (Jin et al., 2021). In addition to direct uptake of tau, microglia can phagocytose tau aggregate-bearing neurons alive (Brelstaff et al., 2018; Pampuscenko et al., 2020). Toxic tau species force microglia to enter a senescent-like, hypofunctional state that intensifies their pro-inflammatory response and results in the release of toxic tau seeds (Hopp et al., 2018; Brelstaff et al., 2021). Interestingly, microglia directly contribute to neuronal loss at a late stage of neurodegeneration, which could be prevented by genetically deleting APOE and TREM2 in mice (Leyns et al., 2017; Shi et al., 2019; Gratuze et al., 2020; Shi et al., 2021; Wang et al., 2021).

Animal studies have provided invaluable insights into the molecular mechanisms of the pathogenesis of AD but conflicting conclusions are often reported in AD mouse models carrying modified AD risk genes including APOE and TREM2 (Shi and Holtzman, 2018; Wolfe et al., 2019; Qu and Li, 2021). Notably, snRNA-seq analyses revealed that mouse DAM signatures only partially match with the microglial transcriptome in the human AD brain, indicating species-specific changes in microglial responses in AD (Mathys et al., 2019; Zhou et al., 2020).

Emerging evidence has supported the suitability of utilizing iPSC-derived microglia to study AD (Abud et al., 2017; Hasselmann and Blurton-Jones, 2020) (Table 2). However, microglia differentiation from iPSCs is challenging due to their unique embryonic origin. Microglia are derived originally from progenitors located in the yolk sac during primitive hematopoiesis and later from mesoderm that migrate into the neural tube (Ginhoux et al., 2013). Therefore, microglial cells have a different embryonic origin than neurons, astrocytes, and oligodendrocytes, which are derived from neuroectoderm and can be differentiated from NPCs (Csobonyeiova et al., 2019). Providing crucial factors to mimic the embryonic development of microglia, multiple protocols have been established to produce human iPSC-derived microglia through lineage states resembling hematopoietic precursor cells (HPC) in vitro (Muffat et al., 2016; Abud et al., 2017; Douvaras et al., 2017; Haenseler et al., 2017; Pandya et al., 2017; Takata et al., 2017; Brownjohn et al., 2018; Garcia-Reitboeck et al., 2018; Konttinen et al., 2019a; Guttikonda et al., 2021). iPSC-derived microglia resemble human primary microglia in that they express microglia-specific markers, secrete cytokines, prune synapses, and are capable of phagocytosing exogenous Aβ (Muffat et al., 2016; Abud et al., 2017; Xu et al., 2019; Guttikonda et al., 2021). These robust and reproducible HPC/microglia differentiation protocols have been utilized to assess microglial function in vitro and human-mouse chimeric models to further interrogate molecular mechanisms of AD.

Table 2:

AD disease phenotypes in iPSC-derived glial cells

Cell type Model system Mutations/Variants Phenotype References
Microglia 2D culture APOE4
  • Reduced ramified morphology

  • Upregulated expression of inflammatory genes

  • Impaired uptake of Aβ

  • Altered metabolism

(Lin et al., 2018)
(Konttinen et al., 2019a)
PSEN1 ΔE9
APP Swe
  • Altered PSEN1 endoproteolysis and accelerated chemokinesis but limited impact overall

(Konttinen et al., 2019a)
Loss-of-function TREM2
  • Reduced phagocytosis

  • Altered inflammatory gene expression

  • Decreased metabolic capacity

(Brownjohn et al., 2018)
(Garcia-Reitboeck et al., 2018)
(Piers et al., 2020)
(Hall-Roberts et al., 2020)
(Reich et al., 2020)
2D co-culture WT microglia with WT astrocyte (Immune response induced by APP Swe AD neurons / exogenous Aβ treatment)
  • Secretion of complement components including C1q and C3

  • Internalization and exocytosis of Aβ42

(Bassil et al., 2021)
(Guttikonda et al., 2021)
3D culture (microfluidi c platform) WT microglia with neurons and astrocytes overexpressin g APP Swe, APP Lon, PSEN1 ΔE9
  • Migration towards AD neurons and astrocytes

  • Secretion of pro-inflammatory factors including NO, IL-6, TNF-α

  • Fragmentation of neurites

  • Induction of loss of astrocytes and neurons

(Park et al., 2018)
Xenotrans plant in 5xFAD mice WT
  • Phagocytosis of amyloid plaques

  • Upregulation of DAM markers near amyloid plaque, including APOE and TREM2

(Abud et al., 2017)
(Hasselmann et al., 2019)
Xenotrans plant in 5xFAD mice Loss-of-function TREM2
  • Locked in homoeostatic status

  • Reduced phagocytosis

  • Fail to cluster around amyloid plaques

  • Reduced accumulation of lipid droplets

(McQuade et al., 2020)
(Claes et al., 2021)
Astrocyte 2D culture PSEN1 M146L PSEN1 ΔE9
  • Atrophic morphology

  • Increased release of inflammatory cytokines

  • Aberrant calcium signaling

  • Increased oxidative stress

(Jones et al., 2017)
(Oksanen et al.,2017)
(Konttinen et al., 2019b)
APOE4
  • Atrophic morphology

  • Increased release of inflammatory cytokines

  • Reduced uptake of Aβ

  • Disrupted lipidomics

  • Lipid droplet accumulation

(Jones et al., 2017) (Lin et al., 2018) (Sienski et al., 2021)
2D co-culture PSEN1 L286V PSEN1 R278I (astrocytes/neurons)
  • Altered processing of APP

  • Increased oxidative stress

(Elsworthy et al., 2021)
APOE4 astrocytes/APOE3 neurons
  • Reduced neurotrophic support to APOE3 neurons

(Zhao et al., 2017)
Xenotrans plant in APP/PS1 mice WT
  • Morphological changes of astrocytes near amyloid plaques

(Preman et al., 2021)

Human iPSC-derived microglia carrying the APOE4 variant, the highest genetic risk factor for SAD, showed reduced ramified morphology, upregulation of pro-inflammatory genes, and impaired uptake of Aβ from the conditional medium of both APOE4 neurons and APOE4 brain organoids compared to their isogenic APOE3 controls (Lin et al., 2018). Interestingly, an independent report also demonstrated that APOE4 profoundly influences function in iPSC-derived microglia in regards to phagocytosis, metabolism, and inflammatory response, whereas the PSEN1 ΔE9 and APP London FAD mutations had limited impact on microglia function overall, suggesting potentially differential roles of microglia in SAD and FAD (Konttinen et al., 2019a).

Impaired microglial phagocytosis as well as altered expression of inflammatory genes and decreased metabolic capacity have been reported in iPSC-derived microglia carrying loss-of-function variants of TREM2, a microglia-specific gene that confers high-risk for AD (Brownjohn et al., 2018; Garcia-Reitboeck et al., 2018; Konttinen et al., 2019a; Hall-Roberts et al., 2020; Piers et al., 2020; Reich et al., 2020). iPSC-derived microglia have also been used to study signaling mechanisms of AD-related genes. For instance, the P522R gain-of-function variant of PLCG2 provides protection against AD (Sims et al., 2017). It was recently discovered that TREM2 and PLCG2 knockout iPSC-derived microglia show shared disease phenotypes with an increased lipid accumulation, impaired phagocytosis, and reduced cell survival (Andreone et al., 2020). In line with this observation, this study on these genetically modified iPSC-derived microglia also demonstrated that PLCG2 is required for TREM2 downstream signaling (Andreone et al., 2020). Collectively, these iPSC-based studies highlight intrinsic microglial dysfunction in AD.

Co-culture iPSC-derived microglia with other brain cell types revealed a non-cell-autonomous role of microglia in the pathogenesis of AD. For instance, human microglia were applied in a 3D AD culture model that contains iPSC-derived neurons and astrocytes overexpressing FAD mutations to recapitulate the human AD brain environment with the formation of Aβ plaque-like aggregates and p-tau in neurites and soma (Park et al., 2018) (Table 2). This AD brain-like environment recruits and activates human microglia resulting in the fragmentation of neurites of co-cultured neurons and in the secretion of pro-inflammatory factors that exacerbate neuron and astrocyte loss (Park et al., 2018). In addition, the co-culture of human iPSC-derived WT microglia and astrocytes with iPSC-derived neurons that harbor the APP London FAD mutation revealed that microglia initiate cellular cross-talk with astrocytes through complement C3 (Guttikonda et al., 2021), which is implicated in synapse loss in AD (Hong et al., 2016; Lian et al., 2016). Utilizing an automated culturing system, a recent report generated iPSC-derived neurons, astrocytes, and microglia as a triculture model of AD induced by exogenous Aβ42 oligomers that harbor key pathological features of AD including the formation of Aβ-positive plaques, induction of p-tau and neurite dystrophy, and neuroinflammation (Bassil et al., 2021). Timelapse imaging of microglia in this system revealed that microglia first internalize soluble Aβ42 oligomers and then exocytose Aβ42 to initiate plaque nucleation with the formation of Aβ plaque-like structures. Utilizing these co-culture systems, future studies could further explore AD-relevant molecular mechanisms such as tau seeding and propagation of tau.

The recent development of human-mouse chimeric models has provided unique opportunities to study human microglia in an in vivo environment. Human microglia require human colony-stimulating factor 1 (CSF1) to survive in immunodeficient mice (Svoboda et al., 2019). Human WT iPSC-derived microglia transplanted into the brain of adult mice were ramified and mobile and express microglial markers that resemble phenotypes of resting microglia (Abud et al., 2017). Also, WT microglial progenitor cells were successfully injected into neonatal mice and functionally integrated into the developing brain (Hasselmann et al., 2019; Svoboda et al., 2019; Xu et al., 2020). In fact, these microglia were capable of responding to exogenous stimuli including injury and demyelination. Single-cell RNA sequencing (scRNA-seq) analyses revealed that these transplanted iPSC-derived microglia retained their identity and displayed a heterogeneous gene expression signature that closely resembles primary human microglia (Svoboda et al., 2019). Similar microglia transplantation models using human embryonic stem cells have also been reported (Mancuso et al., 2019; Fattorelli et al., 2021).

Human microglia transplantation models have also been utilized to study AD. In the brains of immunocompromised AD mice, injected human iPSC-derived WT microglia phagocytose amyloid plaques (Abud et al., 2017). Furthermore, xenografted human WT microglia near plaques upregulate several key DAM markers including APOE and TREM2 (Hasselmann et al., 2019). Notably, comparing the gene expression signature from these grafted human microglia with the expression profile of the murine microglia in this AD mouse model revealed a high degree of discordance in differentially expressed genes, emphasizing the importance of modeling human disease using human cells.

A chimeric microglia transplantation model of AD has also been utilized to study the molecular mechanisms of microglia that harbor AD risk genes, such as TREM2. In this model, TREM2-deficient human iPSC-derived microglia displayed impaired phagocytosis of APOE and they failed to surround amyloid plaques, recapitulating key pathological features in TREM2-deficient human AD brains (McQuade et al., 2020). Furthermore, scRNA-seq analyses showed that transplanted TREM2-deficient microglia fail to upregulate human DAM genes, as similarly seen in previous TREM2 loss-of-function studies (Zhou et al., 2020). In another study, human iPSC-derived microglia from an individual carrying the TREM2 R47H mutation were injected into the brains of neonatal mice revealing a diminished response to amyloid plaques as well as reduced accumulation of lipid droplets (Claes et al., 2021), further highlighting the important role of TREM2 in the context of AD.

Collectively, these studies showed promising disease modeling approaches using iPSC-derived microglia. However, some technical limitations are associated with the application of these human microglia. For instance, microglial transcriptomic changes are sensitive to medium composition (Hasselmann and Blurton-Jones, 2020) and interaction of microglia with other brain cell types is difficult to study in a controlled culture environment. Chimeric human microglia mouse models require specific mouse strains, and the presence of innate murine microglia might complicate the interpretation of results. Thus, these technical limitations need to be addressed in future studies to further optimize disease modelling using human iPSC-derived microglia.

Astrocytes

Astrocytes are the most abundant glia in the brain that serve pivotal roles in supporting brain homeostasis, assisting neuronal signaling, and maintaining the blood-brain barrier (Linnerbauer et al., 2020). Reactive astrogliosis is prominent near amyloid plaques and, in response to AD pathology, astrocytes secrete inflammatory cytokines including interferon-γ (IFN-γ), interleukin-1β (IL-1β), and tumor necrosis factor α (TNF-α), all of which may contribute to neurotoxicity in AD (Hu et al., 1998; Johnstone et al., 1999; Frost and Li, 2017). Interestingly, astrocytes can also produce small levels of Aβ and due to their abundance in the brain, astrocytes contribute significantly to the Aβ burden in AD (Zhao et al., 2011). Astrocytes are the major source of APOE in the brain and they actively communicate with microglia through complement activation (Koistinaho et al., 2004; Lian et al., 2016). Toxic oligomeric tau species can induce astrocyte senescence in AD brains that contribute to neuroinflammation and cognitive impairment (Gaikwad et al., 2021). In addition, pathological tau accumulates in hilar astrocytes of the dentate gyrus of AD patients, which may directly contribute to an impairment of learning and memory (Richetin et al., 2020). Despite the limited expression of tau in astrocytes, bidirectional transmission of toxic tau species between neurons and astrocytes has been proposed as one of the mechanisms that facilitate tau propagation in AD (Maté de Gérando et al., 2021).

Astrocytes can be differentiated from human iPSCs and they have been used for studying AD-related disease mechanisms (Tchieu et al., 2019; Penney et al., 2020; Guttikonda et al., 2021) (Table 2). iPSC-derived astrocytes that harbor PSEN1 mutations show atrophy, increased secretion of Aβ, altered inflammatory response, aberrant calcium signaling, increased oxidative stress, and impaired neuronal support (Jones et al., 2017; Oksanen et al., 2017; Konttinen et al., 2019b; Elsworthy et al., 2021). Furthermore, astrocytes derived from SAD patients carrying APOE4 presented with morphological alterations with an increased release of inflammatory cytokines, reduced uptake of Aβ, disrupted lipid homeostasis, and accumulation of lipid droplets (Jones et al., 2017; Lin et al., 2018; Sienski et al., 2021). iPSC-derived astrocytes respond to exogenous Aβ treatment and form fibrous Aβ aggregates, suggesting a potential role of astrocytes in the compaction of Aβ (Bassil et al., 2021).

Cross-talk between astrocytes and other cells types has also been explored in human iPSC models. Astrocytes co-cultured with neurons show increased arborization and promote neuronal survival but this function is compromised if astrocytes harbor APOE4 (Zhao et al., 2017; Park et al., 2018; Bassil et al., 2021). Also, co-cultures of astrocytes and neurons carrying mutant PSEN1 (L286V and R278I) demonstrate altered processing of APP as well as increased oxidative stress (Elsworthy et al., 2021). iPSC-derived astrocytes can be activated by TNF-α secreted by microglia and they cross-talk with microglia through complement C3 (Guttikonda et al., 2021). In response to AD-related cues, astrocytes also secrete interleukin-3 (IL-3) that recruits and activates microglia to clear Aβ and tau (Guttikonda et al., 2021; McAlpine et al., 2021).

A few studies explored the engraftment of human astrocytes in the brains of mice and showed promising results to utilize the chimeric model to study human astrocytes. Transplantation of human glial progenitor cells into the mouse forebrain gave rise to mature human astrocytes in these mice resulting in enhanced synaptic plasticity and memory (Han et al., 2013). Furthermore, human iPSC-derived astrocytes injected into AD mouse brains showed morphological atrophy or hypertrophy in response to Aβ plaques but interestingly, this phenotype was APOE variant-independent (Preman et al., 2021). Future studies could take advantage of similar transplantation models to further determine functional and molecular changes in astrocytes in response to AD pathology.

Oligodendrocytes

Oligodendrocytes generate myelin sheets that surround axons, facilitate neuronal signaling, and provide trophic support (Simons and Nave, 2015). Myelin loss is an early pathological feature of AD that may precede Aβ and tau pathology as a result of excessive oxidative stress and neuronal dysfunction (Butt et al., 2019; Papuc and Rejdak, 2020). Myelin debris is toxic to neurons and impaired clearance of myelin debris by microglia is closely linked to cognitive decline in aging (Gabande-Rodriguez et al., 2020). Myelin loss is associated with amyloid plaques in AD, and it has been reported that seeding and spreading of tau also occur in oligodendrocytes in the mouse brain (Ferrer et al., 2019). In line with that observation, a recent study showed that the propagation of toxic tau, isolated from patients with progressive supranuclear palsy and corticobasal degeneration, two other tauopathies, in oligodendrocytes is independent of neuronal tau, suggesting that an oligodendrocyte network may be sufficient to propagate tau resulting in myelin loss (Narasimhan et al., 2020). Multiple AD risk genes are also linked to myelin pathology including APOE and TREM2 (Bartzokis et al., 2007; Qu and Li, 2021). Collectively, these studies indicate a potential contribution of oligodendrocyte dysfunction to AD pathogenesis.

Oligodendrocytes can be differentiated from human iPSCs and incorporation of human iPSC-derived oligodendroglial cells in brain organoids as well as successful cell survival after transplantation into myelin basic protein-deficient mouse brains has been reported (Hu et al., 2009; Ehrlich et al., 2017; Madhavan et al., 2018; Marton et al., 2019). However, iPSC models of AD oligodendrocytes have not been reported so far to study the function of oligodendrocytes during AD pathogenesis.

Conclusions

The recent development of generating human iPSC-derived brain cells and the creation of reproducible in vitro and in vivo AD models have provided a powerful toolkit to study the biology of different brain cell types in AD (Figure 1). The phenotypes in AD-patient-derived cells have provided insights into cell type-specific mechanisms of AD pathogenesis (Tables 12, Figure 2). In addition to determining cell-intrinsic and cell type-specific pathology in AD, a better understanding of the molecular cross-talk between different cell types will benefit future treatment strategies and may require the application of mixed cultures, generation of brain organoids or other 3D culture models as well as an analysis of grafts of different neuronal and glial cell types in vivo. The iPSC field is relatively young and some limitations still need to be addressed. For example, the overall number of patients in iPSC studies is relatively low and the in vitro environment does not resemble the microenvironment of AD patient brains with the formation of amyloid plaques and neurofibrillary tangles. Many of the iPSC differentiation protocols are time-consuming and costly to generate cultures of mature neural cells including neurons expression all six different tau isoforms or functional glial cells. Variabilities between different stem cell clones may influence the differentiation of iPSCs into various cell types. Also, microglia and oligodendrocytes are often missing from brain organoids and few studies investigated vascularizing brian organoids to model AD (Papaspyropoulos et al., 2020). In fact, BBB dysfunction is a key pathological feature of AD that should be further explored in AD iPSC models. Given the importance of pericytes in AD and the existence of iPSC-derived pericyte differentiation protocols (Kumar et al., 2017; Delsing et al., 2020; Aisenbrey et al., 2021), future research efforts may benefit from incorporating pericytes along with endothelial cells in these stem cell models of AD. Emerging efforts are underway to address these limitations including generation of isogenic stem cell lines, further improvement of differentiation protocols as well as utilization of 3D culture systems. In addition, in vivo applications of iPSC-derived brain cells in AD murine models can provide a more physiological microenvironment to study patient cells over months to years. Overall, the iPSC technology carries a very strong potential to model neurodegenerative diseases, including AD. It can be applied to drug screening in vitro and in vivo and, potentially, for the development of patient-specific treatment strategies. These models can also be used to validate key findings in rodent studies and to further explore many unsettled questions including the function of AD risk factors in different brain cell types. Combining iPSC technology with genetic and molecular studies in human AD patients and animal models should lead to important advancements in both the understanding and treatment of this devastating disease.

Figure 2: Summary of disease phenotypes in AD iPSC-derived brain cells.

Figure 2:

AD-related phenotypes have been described mainly in neurons, microglia, NPCs, and astrocytes.

Highlights.

  • Patient-derived induced pluripotent stem cells (iPSCs) are suitable to model Alzheimer’s disease (AD)

  • Neurons, microglia and astrocytes can be efficiently differentiated from human iPSCs

  • iPSC technology provides a tool to study AD pathology in 2D and 3D culture systems and in human-mouse transplantation models

  • AD patient iPSC neurons, microglia and astrocytes show pathologic changes in these culture and transplantation models

  • iPSC technology allows to study cell autonomy and non-cell-autonomous mechanisms of disease development in AD

Acknowledgments

Figures 1 and 2 were created with BioRender.com. This work was supported by grants to GH from the NIH including R25NS070697, R03NS112785, R21AG070414, K08NS116166 and ADRC Development Project Award P30AG066462 as well as from the Thompson Family Foundation (TAME-AD grant GT006988-19) and from the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain (TIGER grant).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Abud EM, Ramirez RN, Martinez ES, Healy LM, Nguyen CHH, Newman SA, Yeromin AV, Scarfone VM, Marsh SE, Fimbres C, et al. (2017). iPSC-Derived Human Microglia-like Cells to Study Neurological Diseases. Neuron 94, 278–293 e279. 10.1016/j.neuron.2017.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Aisenbrey EA, Torr E, Johnson H, Soref C, Daly W, and Murphy WL (2021). A protocol for rapid pericyte differentiation of human induced pluripotent stem cells. STAR protocols 2, 100261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Andreone BJ, Przybyla L, Llapashtica C, Rana A, Davis SS, van Lengerich B, Lin K, Shi J, Mei Y, and Astarita G (2020). Alzheimer’s-associated PLCγ2 is a signaling node required for both TREM2 function and the inflammatory response in human microglia. Nature Neuroscience 23, 927–938. [DOI] [PubMed] [Google Scholar]
  4. Bartzokis G, Lu PH, Geschwind DH, Tingus K, Huang D, Mendez MF, Edwards N, and Mintz J (2007). Apolipoprotein E affects both myelin breakdown and cognition: implications for age-related trajectories of decline into dementia. Biol Psychiatry 62, 1380–1387. 10.1016/j.biopsych.2007.03.024. [DOI] [PubMed] [Google Scholar]
  5. Bassil R, Shields K, Granger K, Zein I, Ng S, and Chih B (2021). Improved modeling of human AD with an automated culturing platform for iPSC neurons, astrocytes and microglia. Nat Commun 12, 5220. 10.1038/s41467-021-25344-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Benilova I, Karran E, and De Strooper B (2012). The toxic Abeta oligomer and Alzheimer’s disease: an emperor in need of clothes. Nat Neurosci 15, 349–357. 10.1038/nn.3028. [DOI] [PubMed] [Google Scholar]
  7. Bergström P, Agholme L, Nazir FH, Satir TM, Toombs J, Wellington H, Strandberg J, Bontell TO, Kvartsberg H, and Holmström M (2016). Amyloid precursor protein expression and processing are differentially regulated during cortical neuron differentiation. Scientific reports 6, 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bernabeu-Zornoza A, Coronel R, Palmer C, Monteagudo M, Zambrano A, and Liste I (2019). Physiological and pathological effects of amyloid-beta species in neural stem cell biology. Neural Regen Res 14, 2035–2042. 10.4103/1673-5374.262571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bloom GS (2014). Amyloid-beta and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol 71, 505–508. 10.1001/jamaneurol.2013.5847. [DOI] [PubMed] [Google Scholar]
  10. Born HA (2015). Seizures in Alzheimer’s disease. Neuroscience 286, 251–263. 10.1016/j.neuroscience.2014.11.051. [DOI] [PubMed] [Google Scholar]
  11. Braak H, and Braak E (1991). Neuropathological stageing of Alzheimer-related changes. Acta neuropathologica 82, 239–259. [DOI] [PubMed] [Google Scholar]
  12. Brelstaff J, Tolkovsky AM, Ghetti B, Goedert M, and Spillantini MG (2018). Living Neurons with Tau Filaments Aberrantly Expose Phosphatidylserine and Are Phagocytosed by Microglia. Cell Rep 24, 1939–1948 e1934. 10.1016/j.celrep.2018.07.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Brelstaff JH, Mason M, Katsinelos T, McEwan WA, Ghetti B, Tolkovsky AM, and Spillantini MG (2021). Microglia become hypofunctional and release metalloproteases and tau seeds when phagocytosing live neurons with P301S tau aggregates. Sci Adv 7, eabg4980. 10.1126/sciadv.abg4980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Brownjohn PW, Smith J, Solanki R, Lohmann E, Houlden H, Hardy J, Dietmann S, and Livesey FJ (2018). Functional Studies of Missense TREM2 Mutations in Human Stem Cell-Derived Microglia. Stem Cell Reports 10, 1294–1307. 10.1016/j.stemcr.2018.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Butt AM, De La Rocha IC, and Rivera A (2019). Oligodendroglial Cells in Alzheimer’s Disease. Adv Exp Med Biol 1175, 325–333. 10.1007/978-981-13-9913-8_12. [DOI] [PubMed] [Google Scholar]
  16. Cakir B, Xiang Y, Tanaka Y, Kural MH, Parent M, Kang YJ, Chapeton K, Patterson B, Yuan Y, He CS, et al. (2019). Engineering of human brain organoids with a functional vascular-like system. Nat Methods 16, 1169–1175. 10.1038/s41592-019-0586-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Casali BT, MacPherson KP, Reed-Geaghan EG, and Landreth GE (2020). Microglia depletion rapidly and reversibly alters amyloid pathology by modification of plaque compaction and morphologies. Neurobiol Dis 142, 104956. 10.1016/j.nbd.2020.104956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cenini G, Hebisch M, Iefremova V, Flitsch LJ, Breitkreuz Y, Tanzi RE, Kim DY, Peitz M, and Brustle O (2021). Dissecting Alzheimer’s disease pathogenesis in human 2D and 3D models. Mol Cell Neurosci 110, 103568. 10.1016/j.mcn.2020.103568. [DOI] [PubMed] [Google Scholar]
  19. Ceyzeriat K, Zilli T, Millet P, Frisoni GB, Garibotto V, and Tournier BB (2020). Learning from the Past: A Review of Clinical Trials Targeting Amyloid, Tau and Neuroinflammation in Alzheimer’s Disease. Curr Alzheimer Res 17, 112–125. 10.2174/1567205017666200304085513. [DOI] [PubMed] [Google Scholar]
  20. Chang CW, Evans MD, Yu X, Yu GQ, and Mucke L (2021). Tau reduction affects excitatory and inhibitory neurons differently, reduces excitation/inhibition ratios, and counteracts network hypersynchrony. Cell Rep 37, 109855. 10.1016/j.celrep.2021.109855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Choi SH, Kim YH, Hebisch M, Sliwinski C, Lee S, D’Avanzo C, Chen H, Hooli B, Asselin C, Muffat J, et al. (2014). A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 515, 274–278. 10.1038/nature13800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chow VW, Mattson MP, Wong PC, and Gleichmann M (2010). An overview of APP processing enzymes and products. Neuromolecular Med 12, 1–12. 10.1007/s12017-009-8104-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Claes C, Danhash EP, Hasselmann J, Chadarevian JP, Shabestari SK, England WE, Lim TE, Hidalgo JLS, Spitale RC, Davtyan H, and Blurton-Jones M (2021). Plaque-associated human microglia accumulate lipid droplets in a chimeric model of Alzheimer’s disease. Mol Neurodegener 16, 50. 10.1186/s13024-021-00473-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Clavaguera F, Hench J, Goedert M, and Tolnay M (2015). Invited review: Prion-like transmission and spreading of tau pathology. Neuropathol Appl Neurobiol 41, 47–58. 10.1111/nan.12197. [DOI] [PubMed] [Google Scholar]
  25. Csobonyeiova M, Polak S, Zamborsky R, and Danisovic L (2019). Recent Progress in the Regeneration of Spinal Cord Injuries by Induced Pluripotent Stem Cells. Int J Mol Sci 20, 3838. 10.3390/ijms20153838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Das B, and Yan R (2017). Role of BACE1 in Alzheimer’s synaptic function. Transl Neurodegener 6, 23. 10.1186/s40035-017-0093-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Daviaud N, Friedel RH, and Zou H (2018). Vascularization and Engraftment of Transplanted Human Cerebral Organoids in Mouse Cortex. eNeuro 5. 10.1523/ENEURO.0219-18.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Delizannis AT, Nonneman A, Tsering W, De Bondt A, Van den Wyngaert I, Zhang B, Meymand E, Olufemi MF, Koivula P, Maimaiti S, et al. (2021). Effects of microglial depletion and TREM2 deficiency on Abeta plaque burden and neuritic plaque tau pathology in 5XFAD mice. Acta Neuropathol Commun 9, 150. 10.1186/s40478-021-01251-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Delpech JC, Pathak D, Varghese M, Kalavai SV, Hays EC, Hof PR, Johnson WE, Ikezu S, Medalla M, Luebke JI, and Ikezu T (2021). Wolframin-1-expressing neurons in the entorhinal cortex propagate tau to CA1 neurons and impair hippocampal memory in mice. Sci Transl Med 13, eabe8455. 10.1126/scitranslmed.abe8455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Delsing L, Herland A, Falk A, Hicks R, Synnergren J, and Zetterberg H (2020). Models of the blood-brain barrier using iPSC-derived cells. Molecular and Cellular Neuroscience 107, 103533. [DOI] [PubMed] [Google Scholar]
  31. Dos Santos LR, Pimassoni LHS, Sena GGS, Camporez D, Belcavello L, Trancozo M, Morelato RL, Errera FIV, Bueno MRP, and de Paula F (2017). Validating GWAS variants from microglial genes implicated in Alzheimer’s disease. Journal of Molecular Neuroscience 62, 215–221. [DOI] [PubMed] [Google Scholar]
  32. Douvaras P, Sun B, Wang M, Kruglikov I, Lallos G, Zimmer M, Terrenoire C, Zhang B, Gandy S, Schadt E, et al. (2017). Directed Differentiation of Human Pluripotent Stem Cells to Microglia. Stem Cell Reports 8, 1516–1524. 10.1016/j.stemcr.2017.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Ehrlich M, Mozafari S, Glatza M, Starost L, Velychko S, Hallmann AL, Cui QL, Schambach A, Kim KP, Bachelin C, et al. (2017). Rapid and efficient generation of oligodendrocytes from human induced pluripotent stem cells using transcription factors. Proc Natl Acad Sci U S A 114, E2243–E2252. 10.1073/pnas.1614412114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Elsworthy RJ, King MC, Grainger A, Fisher E, Crowe JA, Alqattan S, Ludlam A, Hill DEJ, and Aldred S (2021). Amyloid-beta precursor protein processing and oxidative stress are altered in human iPSC-derived neuron and astrocyte co-cultures carrying presenillin-1 gene mutations following spontaneous differentiation. Mol Cell Neurosci 114, 103631. 10.1016/j.mcn.2021.103631. [DOI] [PubMed] [Google Scholar]
  35. Espuny-Camacho I, Arranz AM, Fiers M, Snellinx A, Ando K, Munck S, Bonnefont J, Lambot L, Corthout N, and Omodho L (2017). Hallmarks of Alzheimer’s disease in stem-cell-derived human neurons transplanted into mouse brain. Neuron 93, 1066–1081. e1068. [DOI] [PubMed] [Google Scholar]
  36. Espuny-Camacho I, Michelsen KA, Gall D, Linaro D, Hasche A, Bonnefont J, Bali C, Orduz D, Bilheu A, Herpoel A, et al. (2013). Pyramidal neurons derived from human pluripotent stem cells integrate efficiently into mouse brain circuits in vivo. Neuron 77, 440–456. 10.1016/j.neuron.2012.12.011. [DOI] [PubMed] [Google Scholar]
  37. Fattorelli N, Martinez-Muriana A, Wolfs L, Geric I, De Strooper B, and Mancuso R (2021). Stem-cell-derived human microglia transplanted into mouse brain to study human disease. Nat Protoc 16, 1013–1033. 10.1038/s41596-020-00447-4. [DOI] [PubMed] [Google Scholar]
  38. Fernandez MA, Klutkowski JA, Freret T, and Wolfe MS (2014). Alzheimer presenilin-1 mutations dramatically reduce trimming of long amyloid beta-peptides (Abeta) by gamma-secretase to increase 42-to-40-residue Abeta. The Journal of biological chemistry 289, 31043–31052. 10.1074/jbc.M114.581165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Ferrer I, Aguilo Garcia M, Carmona M, Andres-Benito P, Torrejon-Escribano B, Garcia-Esparcia P, and Del Rio JA (2019). Involvement of Oligodendrocytes in Tau Seeding and Spreading in Tauopathies. Front Aging Neurosci 11, 112. 10.3389/fnagi.2019.00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Frost GR, and Li YM (2017). The role of astrocytes in amyloid production and Alzheimer’s disease. Open Biol 7, 170228. 10.1098/rsob.170228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Fu H, Possenti A, Freer R, Nakano Y, Hernandez Villegas NC, Tang M, Cauhy PVM, Lassus BA, Chen S, Fowler SL, et al. (2019). A tau homeostasis signature is linked with the cellular and regional vulnerability of excitatory neurons to tau pathology. Nat Neurosci 22, 47–56. 10.1038/s41593-018-0298-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Gabande-Rodriguez E, Keane L, and Capasso M (2020). Microglial phagocytosis in aging and Alzheimer’s disease. J Neurosci Res 98, 284–298. 10.1002/jnr.24419. [DOI] [PubMed] [Google Scholar]
  43. Gaikwad S, Puangmalai N, Bittar A, Montalbano M, Garcia S, McAllen S, Bhatt N, Sonawane M, Sengupta U, and Kayed R (2021). Tau oligomer induced HMGB1 release contributes to cellular senescence and neuropathology linked to Alzheimer’s disease and frontotemporal dementia. Cell Rep 36, 109419. 10.1016/j.celrep.2021.109419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Garcia-Reitboeck P, Phillips A, Piers TM, Villegas-Llerena C, Butler M, Mallach A, Rodrigues C, Arber CE, Heslegrave A, Zetterberg H, et al. (2018). Human Induced Pluripotent Stem Cell-Derived Microglia-Like Cells Harboring TREM2 Missense Mutations Show Specific Deficits in Phagocytosis. Cell Rep 24, 2300–2311. 10.1016/j.celrep.2018.07.094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Gaspard N, Bouschet T, Hourez R, Dimidschstein J, Naeije G, van den Ameele J, Espuny-Camacho I, Herpoel A, Passante L, Schiffmann SN, et al. (2008). An intrinsic mechanism of corticogenesis from embryonic stem cells. Nature 455, 351–357. 10.1038/nature07287. [DOI] [PubMed] [Google Scholar]
  46. Ghatak S, Dolatabadi N, Trudler D, Zhang X, Wu Y, Mohata M, Ambasudhan R, Talantova M, and Lipton SA (2019). Mechanisms of hyperexcitability in Alzheimer’s disease hiPSC-derived neurons and cerebral organoids vs isogenic controls. Elife 8, e50333. 10.7554/eLife.50333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Ginhoux F, Lim S, Hoeffel G, Low D, and Huber T (2013). Origin and differentiation of microglia. Front Cell Neurosci 7, 45. 10.3389/fncel.2013.00045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Goate A, Chartier-Harlin M-C, Mullan M, Brown J, Crawford F, Fidani L, Giuffra L, Haynes A, Irving N, and James L (1991). Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer’s disease. Nature 349, 704–706. [DOI] [PubMed] [Google Scholar]
  49. Gonzalez C, Armijo E, Bravo-Alegria J, Becerra-Calixto A, Mays CE, and Soto C (2018). Modeling amyloid beta and tau pathology in human cerebral organoids. Mol Psychiatry 23, 2363–2374. 10.1038/s41380-018-0229-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Gratuze M, Chen Y, Parhizkar S, Jain N, Strickland MR, Serrano JR, Colonna M, Ulrich JD, and Holtzman DM (2021). Activated microglia mitigate Abeta-associated tau seeding and spreading. J Exp Med 218, e20210542. 10.1084/jem.20210542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Gratuze M, Leyns CE, Sauerbeck AD, St-Pierre M-K, Xiong M, Kim N, Serrano JR, Tremblay M-È, Kummer TT, and Colonna M (2020). Impact of TREM2 R47H variant on tau pathology–induced gliosis and neurodegeneration. The Journal of clinical investigation 130, 4954–4968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Guttikonda SR, Sikkema L, Tchieu J, Saurat N, Walsh RM, Harschnitz O, Ciceri G, Sneeboer M, Mazutis L, Setty M, et al. (2021). Fully defined human pluripotent stem cell-derived microglia and tri-culture system model C3 production in Alzheimer’s disease. Nat Neurosci 24, 343–354. 10.1038/s41593-020-00796-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Haass C, Kaether C, Thinakaran G, and Sisodia S (2012). Trafficking and proteolytic processing of APP. Cold Spring Harbor perspectives in medicine 2, a006270. 10.1101/cshperspect.a006270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Haenseler W, Sansom SN, Buchrieser J, Newey SE, Moore CS, Nicholls FJ, Chintawar S, Schnell C, Antel JP, Allen ND, et al. (2017). A Highly Efficient Human Pluripotent Stem Cell Microglia Model Displays a Neuronal-Co-culture-Specific Expression Profile and Inflammatory Response. Stem Cell Reports 8, 1727–1742. 10.1016/j.stemcr.2017.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Hall-Roberts H, Agarwal D, Obst J, Smith TB, Monzon-Sandoval J, Di Daniel E, Webber C, James WS, Mead E, Davis JB, and Cowley SA (2020). TREM2 Alzheimer’s variant R47H causes similar transcriptional dysregulation to knockout, yet only subtle functional phenotypes in human iPSC-derived macrophages. Alzheimers Res Ther 12, 151. 10.1186/s13195-020-00709-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Halliday MR, Rege SV, Ma Q, Zhao Z, Miller CA, Winkler EA, and Zlokovic BV (2016). Accelerated pericyte degeneration and blood–brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease. Journal of Cerebral Blood Flow & Metabolism 36, 216–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hallmann AL, Arauzo-Bravo MJ, Mavrommatis L, Ehrlich M, Ropke A, Brockhaus J, Missler M, Sterneckert J, Scholer HR, Kuhlmann T, et al. (2017). Astrocyte pathology in a human neural stem cell model of frontotemporal dementia caused by mutant TAU protein. Sci Rep 7, 42991. 10.1038/srep42991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Han X, Chen M, Wang F, Windrem M, Wang S, Shanz S, Xu Q, Oberheim NA, Bekar L, Betstadt S, et al. (2013). Forebrain engraftment by human glial progenitor cells enhances synaptic plasticity and learning in adult mice. Cell Stem Cell 12, 342–353. 10.1016/j.stem.2012.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hardy J, and Allsop D (1991). Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends in pharmacological sciences 12, 383–388. [DOI] [PubMed] [Google Scholar]
  60. Hasselmann J, and Blurton-Jones M (2020). Human iPSC-derived microglia: A growing toolset to study the brain’s innate immune cells. Glia 68, 721–739. 10.1002/glia.23781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Hasselmann J, Coburn MA, England W, Figueroa Velez DX, Kiani Shabestari S, Tu CH, McQuade A, Kolahdouzan M, Echeverria K, Claes C, et al. (2019). Development of a Chimeric Model to Study and Manipulate Human Microglia In Vivo. Neuron 103, 1016–1033 e1010. 10.1016/j.neuron.2019.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. He Z, Guo JL, McBride JD, Narasimhan S, Kim H, Changolkar L, Zhang B, Gathagan RJ, Yue C, Dengler C, et al. (2018). Amyloid-beta plaques enhance Alzheimer’s brain tau-seeded pathologies by facilitating neuritic plaque tau aggregation. Nat Med 24, 29–38. 10.1038/nm.4443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Hernández F, Merchán-Rubira J, Vallés-Saiz L, Rodríguez-Matellán A, and Avila J (2020). Differences between human and murine tau at the N-terminal end. Frontiers in aging neuroscience 12, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Holtzman DM, Morris JC, and Goate AM (2011). Alzheimer’s disease: the challenge of the second century. Sci Transl Med 3, 77sr71. 10.1126/scitranslmed.3002369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hong S, Beja-Glasser VF, Nfonoyim BM, Frouin A, Li S, Ramakrishnan S, Merry KM, Shi Q, Rosenthal A, Barres BA, et al. (2016). Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352, 712–716. 10.1126/science.aad8373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Hopp SC, Lin Y, Oakley D, Roe AD, DeVos SL, Hanlon D, and Hyman BT (2018). The role of microglia in processing and spreading of bioactive tau seeds in Alzheimer’s disease. J Neuroinflammation 15, 269. 10.1186/s12974-018-1309-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hu BY, Du ZW, and Zhang SC (2009). Differentiation of human oligodendrocytes from pluripotent stem cells. Nat Protoc 4, 1614–1622. 10.1038/nprot.2009.186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Hu J, Akama KT, Krafft GA, Chromy BA, and Van Eldik LJ (1998). Amyloid-beta peptide activates cultured astrocytes: morphological alterations, cytokine induction and nitric oxide release. Brain Res 785, 195–206. 10.1016/s0006-8993(97)01318-8. [DOI] [PubMed] [Google Scholar]
  69. Iovino M, Agathou S, Gonzalez-Rueda A, Del Castillo Velasco-Herrera M, Borroni B, Alberici A, Lynch T, O’Dowd S, Geti I, Gaffney D, et al. (2015). Early maturation and distinct tau pathology in induced pluripotent stem cell-derived neurons from patients with MAPT mutations. Brain 138, 3345–3359. 10.1093/brain/awv222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, Herrera C, Hefferan MP, Van Gorp S, Nazor KL, and Boscolo FS (2012). Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 482, 216–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Jack CR Jr, Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, Petersen RC, and Trojanowski JQ (2010). Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. The Lancet Neurology 9, 119–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Jacobs HIL, Becker JA, Kwong K, Engels-Dominguez N, Prokopiou PC, Papp KV, Properzi M, Hampton OL, d’Oleire Uquillas F, Sanchez JS, et al. (2021). In vivo and neuropathology data support locus coeruleus integrity as indicator of Alzheimer’s disease pathology and cognitive decline. Sci Transl Med 13, eabj2511. 10.1126/scitranslmed.abj2511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Jehuda RB, Shemer Y, and Binah O (2018). Genome editing in induced pluripotent stem cells using CRISPR/Cas9. Stem Cell Reviews and Reports 14, 323–336. [DOI] [PubMed] [Google Scholar]
  74. Jin M, Shiwaku H, Tanaka H, Obita T, Ohuchi S, Yoshioka Y, Jin X, Kondo K, Fujita K, Homma H, et al. (2021). Tau activates microglia via the PQBP1-cGAS-STING pathway to promote brain inflammation. Nat Commun 12, 6565. 10.1038/s41467-021-26851-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Johnstone M, Gearing AJ, and Miller KM (1999). A central role for astrocytes in the inflammatory response to beta-amyloid; chemokines, cytokines and reactive oxygen species are produced. J Neuroimmunol 93, 182–193. 10.1016/s0165-5728(98)00226-4. [DOI] [PubMed] [Google Scholar]
  76. Jones VC, Atkinson-Dell R, Verkhratsky A, and Mohamet L (2017). Aberrant iPSC-derived human astrocytes in Alzheimer’s disease. Cell Death Dis 8, e2696. 10.1038/cddis.2017.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Kamboh MI, Demirci FY, Wang X, Minster RL, Carrasquillo MM, Pankratz VS, Younkin SG, Saykin AJ, Alzheimer’s Disease Neuroimaging I, Jun G, et al. (2012). Genome-wide association study of Alzheimer’s disease. Transl Psychiatry 2, e117. 10.1038/tp.2012.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Kametani F, and Hasegawa M (2018). Reconsideration of Amyloid Hypothesis and Tau Hypothesis in Alzheimer’s Disease. Front Neurosci 12, 25. 10.3389/fnins.2018.00025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Karow M, Camp JG, Falk S, Gerber T, Pataskar A, Gac-Santel M, Kageyama J, Brazovskaja A, Garding A, and Fan W (2018). Direct pericyte-to-neuron reprogramming via unfolding of a neural stem cell-like program. Nature neuroscience 21, 932–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Kazim SF, Chuang S-C, Zhao W, Wong RK, Bianchi R, and Iqbal K (2017). Early-onset network hyperexcitability in presymptomatic Alzheimer’s disease transgenic mice is suppressed by passive immunization with anti-human APP/Aβ antibody and by mGluR5 blockade. Frontiers in aging neuroscience 9, 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK, David E, Baruch K, Lara-Astaiso D, Toth B, et al. (2017). A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease. Cell 169, 1276–1290 e1217. 10.1016/j.cell.2017.05.018. [DOI] [PubMed] [Google Scholar]
  82. Klein RC, Acheson SK, Mace BE, Sullivan PM, and Moore SD (2014). Altered neurotransmission in the lateral amygdala in aged human apoE4 targeted replacement mice. Neurobiology of aging 35, 2046–2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Koch P, Tamboli IY, Mertens J, Wunderlich P, Ladewig J, Stuber K, Esselmann H, Wiltfang J, Brustle O, and Walter J (2012). Presenilin-1 L166P mutant human pluripotent stem cell-derived neurons exhibit partial loss of gamma-secretase activity in endogenous amyloid-beta generation. Am J Pathol 180, 2404–2416. 10.1016/j.ajpath.2012.02.012. [DOI] [PubMed] [Google Scholar]
  84. Koistinaho M, Lin S, Wu X, Esterman M, Koger D, Hanson J, Higgs R, Liu F, Malkani S, Bales KR, and Paul SM (2004). Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat Med 10, 719–726. 10.1038/nm1058. [DOI] [PubMed] [Google Scholar]
  85. Konttinen H, Cabral-da-Silva MEC, Ohtonen S, Wojciechowski S, Shakirzyanova A, Caligola S, Giugno R, Ishchenko Y, Hernandez D, Fazaludeen MF, et al. (2019a). PSEN1DeltaE9, APPswe, and APOE4 Confer Disparate Phenotypes in Human iPSC-Derived Microglia. Stem Cell Reports 13, 669–683. 10.1016/j.stemcr.2019.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Konttinen H, Gureviciene I, Oksanen M, Grubman A, Loppi S, Huuskonen MT, Korhonen P, Lampinen R, Keuters M, Belaya I, et al. (2019b). PPARbeta/delta-agonist GW0742 ameliorates dysfunction in fatty acid oxidation in PSEN1DeltaE9 astrocytes. Glia 67, 146–159. 10.1002/glia.23534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Krasemann S, Madore C, Cialic R, Baufeld C, Calcagno N, El Fatimy R, Beckers L, O’Loughlin E, Xu Y, Fanek Z, et al. (2017). The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity 47, 566–581 e569. 10.1016/j.immuni.2017.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Kuhn R, Mahajan A, Canoll P, and Hargus G (2021). Human Induced Pluripotent Stem Cell Models of Frontotemporal Dementia With Tau Pathology. Front Cell Dev Biol 9, 766773. 10.3389/fcell.2021.766773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Kumar A, D’Souza SS, Moskvin OV, Toh H, Wang B, Zhang J, Swanson S, Guo L-W, Thomson JA, and Slukvin II (2017). Specification and diversification of pericytes and smooth muscle cells from mesenchymoangioblasts. Cell reports 19, 1902–1916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Kwak SS, Washicosky KJ, Brand E, von Maydell D, Aronson J, Kim S, Capen DE, Cetinbas M, Sadreyev R, Ning S, et al. (2020). Amyloid-beta42/40 ratio drives tau pathology in 3D human neural cell culture models of Alzheimer’s disease. Nat Commun 11, 1377. 10.1038/s41467-020-15120-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Lagomarsino VN, Pearse RV 2nd, Liu L, Hsieh YC, Fernandez MA, Vinton EA, Paull D, Felsky D, Tasaki S, Gaiteri C, et al. (2021). Stem cell-derived neurons reflect features of protein networks, neuropathology, and cognitive outcome of their aged human donors. Neuron 109, 3402–3420 e3409. 10.1016/j.neuron.2021.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Lanoiselee HM, Nicolas G, Wallon D, Rovelet-Lecrux A, Lacour M, Rousseau S, Richard AC, Pasquier F, Rollin-Sillaire A, Martinaud O, et al. (2017). APP, PSEN1, and PSEN2 mutations in early-onset Alzheimer disease: A genetic screening study of familial and sporadic cases. PLoS Medicine 14, e1002270. 10.1371/journal.pmed.1002270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Lauterborn JC, Scaduto P, Cox CD, Schulmann A, Lynch G, Gall CM, Keene CD, and Limon A (2021). Increased excitatory to inhibitory synaptic ratio in parietal cortex samples from individuals with Alzheimer’s disease. Nat Commun 12, 2603. 10.1038/s41467-021-22742-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Lee C, Willerth SM, and Nygaard HB (2020). The Use of Patient-Derived Induced Pluripotent Stem Cells for Alzheimer’s Disease Modeling. Prog Neurobiol 192, 101804. 10.1016/j.pneurobio.2020.101804. [DOI] [PubMed] [Google Scholar]
  95. Levy-Lahad E, Wasco W, Poorkaj P, Romano DM, Oshima J, Pettingell WH, Yu C. e., Jondro PD, Schmidt SD, and Wang K (1995). Candidate gene for the chromosome 1 familial Alzheimer’s disease locus. Science 269, 973–977. [DOI] [PubMed] [Google Scholar]
  96. Leyns CEG, Gratuze M, Narasimhan S, Jain N, Koscal LJ, Jiang H, Manis M, Colonna M, Lee VMY, Ulrich JD, and Holtzman DM (2019). TREM2 function impedes tau seeding in neuritic plaques. Nat Neurosci 22, 1217–1222. 10.1038/s41593-019-0433-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Leyns CEG, Ulrich JD, Finn MB, Stewart FR, Koscal LJ, Remolina Serrano J, Robinson GO, Anderson E, Colonna M, and Holtzman DM (2017). TREM2 deficiency attenuates neuroinflammation and protects against neurodegeneration in a mouse model of tauopathy. Proc Natl Acad Sci U S A 114, 11524–11529. 10.1073/pnas.1710311114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Li L, Roh JH, Chang EH, Lee Y, Lee S, Kim M, Koh W, Chang JW, Kim HJ, Nakanishi M, et al. (2018). iPSC Modeling of Presenilin1 Mutation in Alzheimer’s Disease with Cerebellar Ataxia. Exp Neurobiol 27, 350–364. 10.5607/en.2018.27.5.350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Lian H, Litvinchuk A, Chiang ACA, Aithmitti N, Jankowsky JL, and Zheng H (2016). Astrocyte-Microglia Cross Talk through Complement Activation Modulates Amyloid Pathology in Mouse Models of Alzheimer’s Disease. Journal of Neuroscience 36, 577–589. 10.1523/Jneurosci.2117-15.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Lin Y-T, Seo J, Gao F, Feldman HM, Wen H-L, Penney J, Cam HP, Gjoneska E, Raja WK, and Cheng J (2018). APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron 98, 1141–1154. e1147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Lines G, Casey JM, Preza E, and Wray S (2020). Modelling frontotemporal dementia using patient-derived induced pluripotent stem cells. Mol Cell Neurosci 109, 103553. 10.1016/j.mcn.2020.103553. [DOI] [PubMed] [Google Scholar]
  102. Linnerbauer M, Wheeler MA, and Quintana FJ (2020). Astrocyte Crosstalk in CNS Inflammation. Neuron 108, 608–622. 10.1016/j.neuron.2020.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Madhavan M, Nevin ZS, Shick HE, Garrison E, Clarkson-Paredes C, Karl M, Clayton BLL, Factor DC, Allan KC, Barbar L, et al. (2018). Induction of myelinating oligodendrocytes in human cortical spheroids. Nat Methods 15, 700–706. 10.1038/s41592-018-0081-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Maloney B, Ge YW, Alley GM, and Lahiri DK (2007). Important differences between human and mouse APOE gene promoters: limitation of mouse APOE model in studying Alzheimer’s disease. J Neurochem 103, 1237–1257. 10.1111/j.1471-4159.2007.04831.x. [DOI] [PubMed] [Google Scholar]
  105. Mancuso R, Van Den Daele J, Fattorelli N, Wolfs L, Balusu S, Burton O, Liston A, Sierksma A, Fourne Y, Poovathingal S, et al. (2019). Stem-cell-derived human microglia transplanted in mouse brain to study human disease. Nat Neurosci 22, 2111–2116. 10.1038/s41593-019-0525-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Martin-Maestro P, Gargini R,A,AS, Garcia E, Anton LC, Noggle S, Arancio O, Avila J, and Garcia-Escudero V (2017). Mitophagy Failure in Fibroblasts and iPSC-Derived Neurons of Alzheimer’s Disease-Associated Presenilin 1 Mutation. Front Mol Neurosci 10, 291. 10.3389/fnmol.2017.00291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Marton RM, Miura Y, Sloan SA, Li Q, Revah O, Levy RJ, Huguenard JR, and Pasca SP (2019). Differentiation and maturation of oligodendrocytes in human three-dimensional neural cultures. Nat Neurosci 22, 484–491. 10.1038/s41593-018-0316-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Masters CL, and Selkoe DJ (2012). Biochemistry of amyloid beta-protein and amyloid deposits in Alzheimer disease. Cold Spring Harbor perspectives in medicine 2, a006262. 10.1101/cshperspect.a006262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Maté de Gérando A, d’Orange M, Augustin E, Joséphine C, Aurégan G, Gaudin-Guérif M, Guillermier M, Hérard A-S, Stimmer L, and Petit F (2021). Neuronal tau species transfer to astrocytes and induce their loss according to tau aggregation state. Brain 144, 1167–1182. [DOI] [PubMed] [Google Scholar]
  110. Mathys H, Davila-Velderrain J, Peng Z, Gao F, Mohammadi S, Young JZ, Menon M, He L, Abdurrob F, and Jiang X (2019). Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 570, 332–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Matsui TK, Tsuru Y, Hasegawa K, and Kuwako KI (2021). Vascularization of human brain organoids. Stem Cells 39, 1017–1024. 10.1002/stem.3368. [DOI] [PubMed] [Google Scholar]
  112. McAlpine CS, Park J, Griciuc A, Kim E, Choi SH, Iwamoto Y, Kiss MG, Christie KA, Vinegoni C, Poller WC, et al. (2021). Astrocytic interleukin-3 programs microglia and limits Alzheimer’s disease. Nature 595, 701–706. 10.1038/s41586-021-03734-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. McQuade A, Coburn M, Tu CH, Hasselmann J, Davtyan H, and Blurton-Jones M (2018). Development and validation of a simplified method to generate human microglia from pluripotent stem cells. Mol Neurodegener 13, 67. 10.1186/s13024-018-0297-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. McQuade A, Kang YJ, Hasselmann J, Jairaman A, Sotelo A, Coburn M, Shabestari SK, Chadarevian JP, Fote G, Tu CH, et al. (2020). Gene expression and functional deficits underlie TREM2-knockout microglia responses in human models of Alzheimer’s disease. Nat Commun 11, 5370. 10.1038/s41467-020-19227-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Meisl G, Hidari E, Allinson K, Rittman T, DeVos SL, Sanchez JS, Xu CK, Duff KE, Johnson KA, Rowe JB, et al. (2021). In vivo rate-determining steps of tau seed accumulation in Alzheimer’s disease. Sci Adv 7, eabh1448. 10.1126/sciadv.abh1448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Meyer K, Feldman HM, Lu T, Drake D, Lim ET, Ling KH, Bishop NA, Pan Y, Seo J, Lin YT, et al. (2019). REST and Neural Gene Network Dysregulation in iPSC Models of Alzheimer’s Disease. Cell Rep 26, 1112–1127 e1119. 10.1016/j.celrep.2019.01.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Mietelska-Porowska A, Wasik U, Goras M, Filipek A, and Niewiadomska G (2014). Tau Protein Modifications and Interactions: Their Role in Function and Dysfunction. Int J Mol Sci 15, 4671–4713. 10.3390/ijms15034671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Muffat J, Li Y, Yuan B, Mitalipova M, Omer A, Corcoran S, Bakiasi G, Tsai LH, Aubourg P, Ransohoff RM, and Jaenisch R (2016). Efficient derivation of microglia-like cells from human pluripotent stem cells. Nat Med 22, 1358–1367. 10.1038/nm.4189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Müller L, Kirschstein T, Köhling R, Kuhla A, and Teipel S (2021). Neuronal Hyperexcitability in APP SWE/PS1dE9 Mouse Models of Alzheimer’s Disease. Journal of Alzheimer’s Disease 81, 855–869. [DOI] [PubMed] [Google Scholar]
  120. Muratore CR, Rice HC, Srikanth P, Callahan DG, Shin T, Benjamin LN, Walsh DM, Selkoe DJ, and Young-Pearse TL (2014). The familial Alzheimer’s disease APPV717I mutation alters APP processing and Tau expression in iPSC-derived neurons. Hum Mol Genet 23, 3523–3536. 10.1093/hmg/ddu064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Najm R, Zalocusky KA, Zilberter M, Yoon SY, Hao Y, Koutsodendris N, Nelson M, Rao A, Taubes A, Jones EA, and Huang Y (2020). In Vivo Chimeric Alzheimer’s Disease Modeling of Apolipoprotein E4 Toxicity in Human Neurons. Cell Rep 32, 107962. 10.1016/j.celrep.2020.107962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Narasimhan S, Changolkar L, Riddle DM, Kats A, Stieber A, Weitzman SA, Zhang B, Li Z, Roberson ED, Trojanowski JQ, and Lee VMY (2020). Human tau pathology transmits glial tau aggregates in the absence of neuronal tau. The Journal of experimental medicine 217. 10.1084/jem.20190783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Nortley R, Korte N, Izquierdo P, Hirunpattarasilp C, Mishra A, Jaunmuktane Z, Kyrargyri V, Pfeiffer T, Khennouf L, and Madry C (2019). Amyloid β oligomers constrict human capillaries in Alzheimer’s disease via signaling to pericytes. Science 365, eaav9518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Oksanen M, Petersen AJ, Naumenko N, Puttonen K, Lehtonen S, Gubert Olive M, Shakirzyanova A, Leskela S, Sarajarvi T, Viitanen M, et al. (2017). PSEN1 Mutant iPSC-Derived Model Reveals Severe Astrocyte Pathology in Alzheimer’s Disease. Stem Cell Reports 9, 1885–1897. 10.1016/j.stemcr.2017.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Ortiz-Virumbrales M, Moreno CL, Kruglikov I, Marazuela P, Sproul A, Jacob S, Zimmer M, Paull D, Zhang B, Schadt EE, et al. (2017). CRISPR/Cas9-Correctable mutation-related molecular and physiological phenotypes in iPSC-derived Alzheimer’s PSEN2 (N141I) neurons. Acta Neuropathol Commun 5, 77. 10.1186/s40478-017-0475-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Ovchinnikov DA, Korn O, Virshup I, Wells CA, and Wolvetang EJ (2018). The Impact of APP on Alzheimer-like Pathogenesis and Gene Expression in Down Syndrome iPSC-Derived Neurons. Stem Cell Reports 11, 32–42. 10.1016/j.stemcr.2018.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Pampuscenko K, Morkuniene R, Sneideris T, Smirnovas V, Budvytyte R, Valincius G, Brown GC, and Borutaite V (2020). Extracellular tau induces microglial phagocytosis of living neurons in cell cultures. J Neurochem 154, 316–329. 10.1111/jnc.14940. [DOI] [PubMed] [Google Scholar]
  128. Pandya H, Shen MJ, Ichikawa DM, Sedlock AB, Choi Y, Johnson KR, Kim G, Brown MA, Elkahloun AG, Maric D, et al. (2017). Differentiation of human and murine induced pluripotent stem cells to microglia-like cells. Nat Neurosci 20, 753–759. 10.1038/nn.4534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Papaspyropoulos A, Tsolaki M, Foroglou N, and Pantazaki AA (2020). Modeling and Targeting Alzheimer’s Disease With Organoids. Front Pharmacol 11, 396. 10.3389/fphar.2020.00396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Papuc E, and Rejdak K (2020). The role of myelin damage in Alzheimer’s disease pathology. Arch Med Sci 16, 345–351. 10.5114/aoms.2018.76863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Park J, Wetzel I, Marriott I, Dreau D, D’Avanzo C, Kim DY, Tanzi RE, and Cho H (2018). A 3D human triculture system modeling neurodegeneration and neuroinflammation in Alzheimer’s disease. Nat Neurosci 21, 941–951. 10.1038/s41593-018-0175-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Park SA, Ahn SI, and Gallo JM (2016). Tau mis-splicing in the pathogenesis of neurodegenerative disorders. BMB Rep 49, 405–413. 10.5483/bmbrep.2016.49.8.084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Penney J, Ralvenius WT, and Tsai L-H (2020). Modeling Alzheimer’s disease with iPSC-derived brain cells. Molecular psychiatry 25, 148–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Piers TM, Cosker K, Mallach A, Johnson GT, Guerreiro R, Hardy J, and Pocock JM (2020). A locked immunometabolic switch underlies TREM2 R47H loss of function in human iPSC-derived microglia. FASEB J 34, 2436–2450. 10.1096/fj.201902447R. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Pomeshchik Y, Klementieva O, Gil J, Martinsson I, Hansen MG, de Vries T, Sancho-Balsells A, Russ K, Savchenko E, Collin A, et al. (2020). Human iPSC-Derived Hippocampal Spheroids: An Innovative Tool for Stratifying Alzheimer Disease Patient-Specific Cellular Phenotypes and Developing Therapies. Stem Cell Reports 15, 256–273. 10.1016/j.stemcr.2020.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Preman P, Tcw J, Calafate S, Snellinx A, Alfonso-Triguero M, Corthout N, Munck S, Thal DR, Goate AM, and De Strooper B (2021). Human iPSC-derived astrocytes transplanted into the mouse brain undergo morphological changes in response to amyloid-β plaques. Molecular neurodegeneration 16, 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Purhonen J, Grigorjev V, Ekiert R, Aho N, Rajendran J, Pietras R, Truve K, Wikstrom M, Sharma V, Osyczka A, et al. (2020). A spontaneous mitonuclear epistasis converging on Rieske Fe-S protein exacerbates complex III deficiency in mice. Nat Commun 11, 322. 10.1038/s41467-019-14201-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Qu W, and Li L (2020). Loss of TREM2 Confers Resilience to Synaptic and Cognitive Impairment in Aged Mice. The Journal of neuroscience : the official journal of the Society for Neuroscience 40, 9552–9563. 10.1523/JNEUROSCI.2193-20.2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Qu W, and Li L (2021). Microglial TREM2 at the Intersection of Brain Aging and Alzheimer’s Disease. Neuroscientist, 10738584211040786. 10.1177/10738584211040786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Raja WK, Mungenast AE, Lin YT, Ko T, Abdurrob F, Seo J, and Tsai LH (2016). Self-Organizing 3D Human Neural Tissue Derived from Induced Pluripotent Stem Cells Recapitulate Alzheimer’s Disease Phenotypes. PLoS One 11, e0161969. 10.1371/journal.pone.0161969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Ransohoff RM, and El Khoury J (2015). Microglia in Health and Disease. Cold Spring Harb Perspect Biol 8, a020560. 10.1101/cshperspect.a020560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Rauch JN, Luna G, Guzman E, Audouard M, Challis C, Sibih YE, Leshuk C, Hernandez I, Wegmann S, Hyman BT, et al. (2020). LRP1 is a master regulator of tau uptake and spread. Nature 580, 381–385. 10.1038/s41586-020-2156-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Reich M, Paris I, Ebeling M, Dahm N, Schweitzer C, Reinhardt D, Schmucki R, Prasad M, Kochl F, Leist M, et al. (2020). Alzheimer’s Risk Gene TREM2 Determines Functional Properties of New Type of Human iPSC-Derived Microglia. Front Immunol 11, 617860. 10.3389/fimmu.2020.617860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Richetin K, Steullet P, Pachoud M, Perbet R, Parietti E, Maheswaran M, Eddarkaoui S, Begard S, Pythoud C, Rey M, et al. (2020). Tau accumulation in astrocytes of the dentate gyrus induces neuronal dysfunction and memory deficits in Alzheimer’s disease. Nat Neurosci 23, 1567–1579. 10.1038/s41593-020-00728-x. [DOI] [PubMed] [Google Scholar]
  145. Sagare AP, Bell RD, Zhao Z, Ma Q, Winkler EA, Ramanathan A, and Zlokovic BV (2013). Pericyte loss influences Alzheimer-like neurodegeneration in mice. Nature communications 4, 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  146. Selkoe DJ, and Hardy J (2016). The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO molecular medicine 8, 595–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Serio A, Bilican B, Barmada SJ, Ando DM, Zhao C, Siller R, Burr K, Haghi G, Story D, Nishimura AL, et al. (2013). Astrocyte pathology and the absence of non-cell autonomy in an induced pluripotent stem cell model of TDP-43 proteinopathy. Proc Natl Acad Sci U S A 110, 4697–4702. 10.1073/pnas.1300398110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Serrano-Pozo A, Frosch MP, Masliah E, and Hyman BT (2011). Neuropathological alterations in Alzheimer disease. Cold Spring Harbor perspectives in medicine 1, a006189. 10.1101/cshperspect.a006189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Sharma A, Sances S, Workman MJ, and Svendsen CN (2020). Multi-lineage Human iPSC-Derived Platforms for Disease Modeling and Drug Discovery. Cell Stem Cell 26, 309–329. 10.1016/j.stem.2020.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Shaw LM, Vanderstichele H, Knapik-Czajka M, Clark CM, Aisen PS, Petersen RC, Blennow K, Soares H, Simon A, Lewczuk P, et al. (2009). Cerebrospinal fluid biomarker signature in Alzheimer’s disease neuroimaging initiative subjects. Ann Neurol 65, 403–413. 10.1002/ana.21610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Shen J, and Kelleher RJ 3rd (2007). The presenilin hypothesis of Alzheimer’s disease: evidence for a loss-of-function pathogenic mechanism. Proc Natl Acad Sci U S A 104, 403–409. 10.1073/pnas.0608332104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Sherrington R, Rogaev E, Liang Y, Rogaeva E, Levesque G, Ikeda M, Chi H, Lin C, Li G, and Holman K (1995). Cloning of a novel gene bearing missense mutations in early-onset Alzheimer’s disease. [DOI] [PubMed] [Google Scholar]
  153. Shi Y, Andhey PS, Ising C, Wang K, Snipes LL, Boyer K, Lawson S, Yamada K, Qin W, Manis M, et al. (2021). Overexpressing low-density lipoprotein receptor reduces tau-associated neurodegeneration in relation to apoE-linked mechanisms. Neuron 109, 2413–2426 e2417. 10.1016/j.neuron.2021.05.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Shi Y, and Holtzman DM (2018). Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat Rev Immunol 18, 759–772. 10.1038/s41577-018-0051-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Shi Y, Kirwan P, Smith J, Robinson HP, and Livesey FJ (2012). Human cerebral cortex development from pluripotent stem cells to functional excitatory synapses. Nat Neurosci 15, 477–486, S471. 10.1038/nn.3041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Shi Y, Manis M, Long J, Wang K, Sullivan PM, Remolina Serrano J, Hoyle R, and Holtzman DM (2019). Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. The Journal of experimental medicine 216, 2546–2561. 10.1084/jem.20190980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Sienski G, Narayan P, Bonner JM, Kory N, Boland S, Arczewska AA, Ralvenius WT, Akay L, Lockshin E, He L, et al. (2021). APOE4 disrupts intracellular lipid homeostasis in human iPSC-derived glia. Sci Transl Med 13. 10.1126/scitranslmed.aaz4564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Simons M, and Nave KA (2015). Oligodendrocytes: Myelination and Axonal Support. Cold Spring Harb Perspect Biol 8, a020479. 10.1101/cshperspect.a020479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Sims R, van der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, Kunkle BW, Boland A, Raybould R, Bis JC, et al. (2017). Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat Genet 49, 1373–1384. 10.1038/ng.3916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Sposito T, Preza E, Mahoney CJ, Seto-Salvia N, Ryan NS, Morris HR, Arber C, Devine MJ, Houlden H, Warner TT, et al. (2015). Developmental regulation of tau splicing is disrupted in stem cell-derived neurons from frontotemporal dementia patients with the 10 + 16 splice-site mutation in MAPT. Hum Mol Genet 24, 5260–5269. 10.1093/hmg/ddv246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Sproul AA, Jacob S, Pre D, Kim SH, Nestor MW, Navarro-Sobrino M, Santa-Maria I, Zimmer M, Aubry S, Steele JW, et al. (2014). Characterization and molecular profiling of PSEN1 familial Alzheimer’s disease iPSC-derived neural progenitors. PLoS One 9, e84547. 10.1371/journal.pone.0084547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Stancu IC, Vasconcelos B, Terwel D, and Dewachter I (2014). Models of beta-amyloid induced Tau-pathology: the long and “folded” road to understand the mechanism. Mol Neurodegener 9, 51. 10.1186/1750-1326-9-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Svoboda DS, Barrasa MI, Shu J, Rietjens R, Zhang S, Mitalipova M, Berube P, Fu D, Shultz LD, Bell GW, and Jaenisch R (2019). Human iPSC-derived microglia assume a primary microglia-like state after transplantation into the neonatal mouse brain. Proc Natl Acad Sci U S A 116, 25293–25303. 10.1073/pnas.1913541116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Sweeney MD, Ayyadurai S, and Zlokovic BV (2016). Pericytes of the neurovascular unit: key functions and signaling pathways. Nature neuroscience 19, 771–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Sweeney MD, Sagare AP, and Zlokovic BV (2018). Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat Rev Neurol 14, 133–150. 10.1038/nrneurol.2017.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Tábuas-Pereira M, Santana I, Guerreiro R, and Brás J (2020). Alzheimer’s disease genetics: Review of Novel Loci associated with disease. Current Genetic Medicine Reports 8, 1–16. [Google Scholar]
  167. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, and Yamanaka S (2007). Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872. 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
  168. Takata K, Kozaki T, Lee CZW, Thion MS, Otsuka M, Lim S, Utami KH, Fidan K, Park DS, Malleret B, et al. (2017). Induced-Pluripotent-Stem-Cell-Derived Primitive Macrophages Provide a Platform for Modeling Tissue-Resident Macrophage Differentiation and Function. Immunity 47, 183–198 e186. 10.1016/j.immuni.2017.06.017. [DOI] [PubMed] [Google Scholar]
  169. Tarawneh R, and Holtzman DM (2012). The clinical problem of symptomatic Alzheimer disease and mild cognitive impairment. Cold Spring Harb Perspect Med 2, a006148. 10.1101/cshperspect.a006148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Tchieu J, Calder EL, Guttikonda SR, Gutzwiller EM, Aromolaran KA, Steinbeck JA, Goldstein PA, and Studer L (2019). NFIA is a gliogenic switch enabling rapid derivation of functional human astrocytes from pluripotent stem cells. Nature biotechnology 37, 267–275. 10.1038/s41587-019-0035-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Trabzuni D, Wray S, Vandrovcova J, Ramasamy A, Walker R, Smith C, Luk C, Gibbs JR, Dillman A, Hernandez DG, et al. (2012). MAPT expression and splicing is differentially regulated by brain region: relation to genotype and implication for tauopathies. Human Molecular Genetics 21, 4094–4103. 10.1093/hmg/dds238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Varela EV, Etter G, and Williams S (2019). Excitatory-inhibitory imbalance in Alzheimer’s disease and therapeutic significance. Neurobiol Dis 127, 605–615. 10.1016/j.nbd.2019.04.010. [DOI] [PubMed] [Google Scholar]
  173. Wang C, Najm R, Xu Q, Jeong DE, Walker D, Balestra ME, Yoon SY, Yuan H, Li G, Miller ZA, et al. (2018). Gain of toxic apolipoprotein E4 effects in human iPSC-derived neurons is ameliorated by a small-molecule structure corrector. Nat Med 24, 647–657. 10.1038/s41591-018-0004-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Wang C, Xiong M, Gratuze M, Bao X, Shi Y, Andhey PS, Manis M, Schroeder C, Yin Z, Madore C, et al. (2021). Selective removal of astrocytic APOE4 strongly protects against tau-mediated neurodegeneration and decreases synaptic phagocytosis by microglia. Neuron 109, 1657–1674 e1657. 10.1016/j.neuron.2021.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Wezyk M, Szybinska A, Wojsiat J, Szczerba M, Day K, Ronnholm H, Kele M, Berdynski M, Peplonska B, Fichna JP, et al. (2018). Overactive BRCA1 Affects Presenilin 1 in Induced Pluripotent Stem Cell-Derived Neurons in Alzheimer’s Disease. J Alzheimers Dis 62, 175–202. 10.3233/JAD-170830. [DOI] [PubMed] [Google Scholar]
  176. Wisniewski K, Wisniewski H, and Wen G (1985). Occurrence of Alzheimer’s neuropathology and dementia in Down syndrome. Annals of Neurology 17, 278–282. [DOI] [PubMed] [Google Scholar]
  177. Wolfe CM, Fitz NF, Nam KN, Lefterov I, and Koldamova R (2019). The role of APOE and TREM2 in Alzheimer′ s disease—current understanding and perspectives. International journal of molecular sciences 20, 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Wu JW, Hussaini SA, Bastille IM, Rodriguez GA, Mrejeru A, Rilett K, Sanders DW, Cook C, Fu H, Boonen RA, et al. (2016). Neuronal activity enhances tau propagation and tau pathology in vivo. Nat Neurosci 19, 1085–1092. 10.1038/nn.4328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Xu M, Zhang L, Liu G, Jiang N, Zhou W, and Zhang Y (2019). Pathological changes in Alzheimer’s disease analyzed using induced pluripotent stem cell-derived human microglia-like cells. Journal of Alzheimer’s Disease 67, 357–368. [DOI] [PubMed] [Google Scholar]
  180. Xu R, Li X, Boreland AJ, Posyton A, Kwan K, Hart RP, and Jiang P (2020). Human iPSC-derived mature microglia retain their identity and functionally integrate in the chimeric mouse brain. Nat Commun 11, 1577. 10.1038/s41467-020-15411-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, Yoshizaki T, Yamanaka S, Okano H, and Suzuki N (2011). Modeling familial Alzheimer’s disease with induced pluripotent stem cells. Human molecular genetics 20, 4530–4539. [DOI] [PubMed] [Google Scholar]
  182. Yang J, Zhao H, Ma Y, Shi G, Song J, Tang Y, Li S, Li T, Liu N, Tang F, et al. (2017). Early pathogenic event of Alzheimer’s disease documented in iPSCs from patients with PSEN1 mutations. Oncotarget 8, 7900–7913. 10.18632/oncotarget.13776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Zhang D, Pekkanen-Mattila M, Shahsavani M, Falk A, Teixeira AI, and Herland A (2014). A 3D Alzheimer’s disease culture model and the induction of P21-activated kinase mediated sensing in iPSC derived neurons. Biomaterials 35, 1420–1428. 10.1016/j.biomaterials.2013.11.028. [DOI] [PubMed] [Google Scholar]
  184. Zhang S, Wan Z, and Kamm RD (2021). Vascularized organoids on a chip: strategies for engineering organoids with functional vasculature. Lab Chip 21, 473–488. 10.1039/d0lc01186j. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. 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. 10.1016/j.neuron.2013.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Zhao J, Davis MD, Martens YA, Shinohara M, Graff-Radford NR, Younkin SG, Wszolek ZK, Kanekiyo T, and Bu G (2017). APOE ε4/ε4 diminishes neurotrophic function of human iPSC-derived astrocytes. Human molecular genetics 26, 2690–2700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Zhao J, Fu Y, Yamazaki Y, Ren Y, Davis MD, Liu CC, Lu W, Wang X, Chen K, Cherukuri Y, et al. (2020). APOE4 exacerbates synapse loss and neurodegeneration in Alzheimer’s disease patient iPSC-derived cerebral organoids. Nat Commun 11, 5540. 10.1038/s41467-020-19264-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Zhao J, O’Connor T, and Vassar R (2011). The contribution of activated astrocytes to Abeta production: implications for Alzheimer’s disease pathogenesis. J Neuroinflammation 8, 150. 10.1186/1742-2094-8-150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Zhou Y, Song WM, Andhey PS, Swain A, Levy T, Miller KR, Poliani PL, Cominelli M, Grover S, and Gilfillan S (2020). Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer’s disease. Nature medicine 26, 131–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Zhou ZD, Chan CH, Ma QH, Xu XH, Xiao ZC, and Tan EK (2011). The roles of amyloid precursor protein (APP) in neurogenesis: Implications to pathogenesis and therapy of Alzheimer disease. Cell Adh Migr 5, 280–292. 10.4161/cam.5.4.16986. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES