Skip to main content
Advertisement

Main menu

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET

User menu

  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Journal of Pharmacology and Experimental Therapeutics
  • Other Publications
    • Drug Metabolism and Disposition
    • Journal of Pharmacology and Experimental Therapeutics
    • Molecular Pharmacology
    • Pharmacological Reviews
    • Pharmacology Research & Perspectives
    • ASPET
  • My alerts
  • Log in
  • My Cart
Journal of Pharmacology and Experimental Therapeutics

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Fast Forward
    • Latest Articles
    • Special Sections
    • Archive
  • Information
    • Instructions to Authors
    • Submit a Manuscript
    • FAQs
    • For Subscribers
    • Terms & Conditions of Use
    • Permissions
  • Editorial Board
  • Alerts
    • Alerts
    • RSS Feeds
  • Virtual Issues
  • Feedback
  • Submit
  • Visit jpet on Facebook
  • Follow jpet on Twitter
  • Follow jpet on LinkedIn
Research ArticleMinireview
Open Access

Stem Cell Therapies in Alzheimer’s Disease: Applications for Disease Modeling

Zizhen Si and Xidi Wang
Journal of Pharmacology and Experimental Therapeutics May 2021, 377 (2) 207-217; DOI: https://doi.org/10.1124/jpet.120.000324
Zizhen Si
Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Xidi Wang
Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF
Loading

Abstract

Alzheimer’s disease (AD) is a neurodegenerative disease with complex pathologic and biologic characteristics. Extracellular β-amyloid deposits, such as senile plaques, and intracellular aggregation of hyperphosphorylated tau, such as neurofibrillary tangles, remain the main neuropathological criteria for the diagnosis of AD. There is currently no effective treatment of the disease, and many clinical trials have failed to prove any benefits of new therapeutics. More recently, there has been increasing interest in harnessing the potential of stem cell technologies for drug discovery, disease modeling, and cell therapies, which have been used to study an array of human conditions, including AD. The recently developed and optimized induced pluripotent stem cell (iPSC) technology is a critical platform for screening anti-AD drugs and understanding mutations that modify AD. Neural stem cell (NSC) transplantation has been investigated as a new therapeutic approach to treat neurodegenerative diseases. Mesenchymal stem cells (MSCs) also exhibit considerable potential to treat neurodegenerative diseases by secreting growth factors and exosomes, attenuating neuroinflammation. This review highlights recent progress in stem cell research and the translational applications and challenges of iPSCs, NSCs, and MSCs as treatment strategies for AD. Even though these treatments are still in relative infancy, these developing stem cell technologies hold considerable promise to combat AD and other neurodegenerative disorders.

SIGNIFICANCE STATEMENT Alzheimer’s disease (AD) is a neurodegenerative disease that results in learning and memory defects. Although some drugs have been approved for AD treatment, fewer than 20% of patients with AD benefit from these drugs. Therapies based on stem cells, including induced pluripotent stem cells, neural stem cells, and mesenchymal stem cells, provide promising therapeutic strategies for AD.

Introduction

Alzheimer’s disease (AD) is the most common cause of dementia, named first in 1906 by Alois Alzheimer. Currently, the presence of extracellular β-amyloid deposits, such as senile plaques, and the intracellular accumulation of hyperphosphorylated tau, such as neurofibrillary tangles, are still the main neuropathological criteria for the diagnosis of AD (Kent et al., 2020; Scearce-Levie et al., 2020). Early-onset AD emerges in patients younger than 65 years of age, accounting for less than 5% of all cases, and most cases of late-onset AD occur after the age of 65 (Sabayan and Sorond, 2017). Patients with AD will inevitably die within 5–12 years of the onset of AD symptoms (Bruni et al., 2020). The clinical manifestations of AD are progressive. Typical features are early neuroinflammation and learning and memory impairments, followed by complex attention, visuospatial function, executive function, praxis, language, gnosis, behavioral, and/or social impairment (Scheltens et al., 2016; Arvanitakis et al., 2019; Lemprière, 2019). In recent years, although significant progress has been made in clarifying key aspects of the biology, the etiological mechanisms of AD are still far from being fully understood (Jafari et al., 2020; Kim et al., 2020). New treatment strategies and drugs attempting to slow or halt cognitive deficiency and neuronal loss of AD are proposed every year (Roberson and Mucke, 2006; Benek et al., 2020). The US Food and Drug Administration has only approved five drugs for the clinical treatment of AD, and these include the cholinesterase inhibitors tacrine, galantamine, donepezil, and rivastigmine, and the glutamate receptor antagonist memantine. However, these five pharmacological agents can only relieve symptoms without affecting the main pathologic features of AD (Kumar et al., 2015; Stakos et al., 2020). In addition, the effects of these drugs vary from person to person; no more than 20% of patients have a moderate efficiency, whereas more than 60% of patients have tolerance, noncompliance, and side effects (Serretti et al., 2007;(Zetterberg and Bendlin, 2021) ). Therefore, effective therapeutic strategies for AD are of great priority.

In the past years, there has been increasing interest in harnessing the potential of stem cell technology for drug discovery, disease modeling, and cell therapies (Mancuso et al., 2019; Lee et al., 2020; Yang et al., 2020). The most commonly used stem cell types in AD research are induced pluripotent stem cells (iPSCs), brain-derived neural stem cells (NSCs), and bone marrow mesenchymal stem cells (MSCs) (Yang et al., 2013; Chen et al., 2014; Penney et al., 2020). Stem cell–based therapy might be a better approach than traditional therapies, as it could reduce neuronal loss, increase synaptic connections, and improve the microenvironment in the brain fundamentally. The mechanisms of action (Fig. 1) include the following. 1) Replacement of injured or lost neuronal cells: stem cells can differentiate into cholinergic neurons, which could integrate with the host, repair neural circuits, and eventually replace the lost neurons (Telias and Ben-Yosef, 2015). 2) Secretion of neurotrophic factors: stem cells can secrete neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) and fibroblast growth factor, to promote cell survival, increase synaptic connections, and improve cognitive function (Blurton-Jones et al., 2009). 3) Anti-amyloid protein production: stem cell transplantation reduces amyloid beta (Aβ) levels and reduces Aβ toxic reactions, which is beneficial for the survival of transplanted cells and cognitive recovery (Bae et al., 2013). 4) Anti-inflammatory response: stem cell transplantation reduces the expression of proinflammatory factors interleukin-1β, interleukin-6, tumor necrosis factor-α, inducible nitric oxide synthase, and exerts neuroprotective effects (Lin et al., 2018). 5) Promotion of the activation of endogenous stem cells: transplantation of exogenous stem cells improves the microenvironment of brain, which facilitates the survival of endogenous stem cells and stimulates their activation (Philips and Robberecht, 2011). 6) Improvement of the metabolic activity of neurons in the brain: stem cell transplantation increases the connection and metabolism between neurons and improves cognitive function (Blurton-Jones et al., 2014). Progress in stem cell–based therapy provides a new perspective for treating neurodegenerative disease, especially in AD. In this review, we underline some key insights into the disease mechanisms derived from studies of iPSCs, NSCs, and MSCs and discuss the pros and cons of these stem cell types as therapeutics. Additionally, we review new research to track stem cell therapy, highlight the most relevant stem cell trials in AD and other neurologic disorders, and discuss the potential applications, as well as the major challenges and future directions in cell-replacement therapy of AD.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Stem cell mechanisms of action to treat AD. 1) Replacement of the injured or lost neuronal cells. 2) Secretion of neurotrophic factors [BDNF and fibroblast growth factor (FGF)]. 3) Anti-amyloid protein production. 4) Anti-inflammatory response [interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α)]. 5) Promotion of the activation of endogenous stem cells. 6) Improvement of the metabolic activity of neurons in the brain.

The Pathogenesis of AD

AD is the most common neurodegenerative disorder causing dementia and is characterized by memory deficit and cognitive decline (Hampel et al., 2018; Ong et al., 2018). Early-onset familial AD (FAD) occurs in people aged 40–60 years, and sporadic late-onset AD (SAD) occurs after the age of 70 (Jeong, 2017). For the neuropathological diagnosis of AD, cerebrospinal fluid (CSF) or positron emission tomography imaging biomarkers can be used as surrogate markers for Aβ and tau deposition in brains (Brier et al., 2016; Leuzy et al., 2016). Studies of cognitive function and changes in CSF and neuroimaging biomarkers in FAD and SAD have determined that the disease is at a preclinical stage at least 10–20 years before the onset of clinical symptoms (Olsson et al., 2016; Tarawneh et al., 2016). The disease is characterized by the early deposition of Aβ in early-onset nerves and other cortical areas, including the default pattern network, followed by regional cortical hypometabolism, decreased hippocampal volume, accumulation of tau pathology, and the onset of symptomatic cognitive impairment. Plasma neurofilament light chain and CSF are emerging biomarkers that track the general level of neurodegeneration in all forms of neurodegenerative dementia (Di Stefano et al., 2016; Han et al., 2016; Rabinovici, 2016; Pascoal et al., 2017).

Apolipoprotein E (ApoE) may affect amyloid pathology by directly binding Aβ in the plaque, regulating AD risk. ApoE can have a regulatory effect on tau pathology and tau-related neurodegeneration and may independently affect neurons and neuronal networks (Li et al., 2019; Mentis et al., 2020). Reactive astrocyte and microglia hyperplasia are prominent pathologic features of the AD brain, and the activation of the immune system is a critical regulator of AD pathology (Sabatino et al., 2019; Heneka, 2020).

Although significant progress has been made in the understanding of the pathology of AD, we have yet to discover disease-relief therapies that are effective in humans. The pathologic biology of AD is very complicated. The older the age, the greater the possibility that other age-related diseases and AD pathology will cause cognitive decline (Congdon and Sigurdsson, 2018; Si et al., 2018; Teipel et al., 2018; Cummings, 2019). The ongoing in-depth research in this area is critical to making discoveries that will eventually reveal novel treatments that can truly change the course of the disease.

Stem Cell Treatment in AD Modeling

Some drugs have been approved to slow down cognitive deficiency and neuronal loss in AD. Although these drugs can improve some AD symptoms, fewer than 20% of patients with AD will benefit, whereas over 60% of patients develop tolerance and side effects (Serretti et al., 2007; Zetterberg and Bendlin, 2021). With rapid growing achievements in stem cell research, stem cell–based therapy provides a new option for AD treatment.

Applications of iPSCs in AD Modeling

Mouse fibroblast cells were first shown to be reprogrammed into iPSCs in 2006 by applying four transcription factors, including sex-determining region Y-box 2 (Sox2), c-Myc, octamer-binding transcription factor 4, and Kruppel-like factor 4 (Takahashi and Yamanaka, 2006). The next year, this technology was applied to human somatic cells to generate iPSCs successfully (Takahashi et al., 2007). Since then, considerable efforts have followed to optimize this technology and reprogram cells by newly defined or fewer factors and more efficient delivery systems (Chuah and Zink, 2017; Di Lullo and Kriegstein, 2017; Pournasr and Duncan, 2017; Devalla and Passier, 2018). Lineage specifiers involved in the ectodermal specification and mesendodermal specification can synergistically induce pluripotency without octamer-binding transcription factor 4 and Sox2 (Aoi, 2008; Nakagawa et al., 2008; Chia et al., 2010; Buganim and Jaenisch, 2012). A growing number of novel reprogramming factors have been identified as maternal and pluripotency-associated factors, such as Esrrb, Tet1, Sall4, and PR domain-containing 14 (Maherali et al., 2008; Doege et al., 2012; Moon et al., 2012; Hu et al., 2014; Chen et al., 2015). Manipulation of microRNAs can replace traditional reprogramming factors to increase the efficiency of reprogramming somatic cells into iPSCs (Judson et al., 2009; Anokye-Danso et al., 2011). Furthermore, the differentiation into iPSCs has been extended to various cell types, including human keratinocytes, fibroblasts, mature B lymphocytes, liver and stomach cells, human amniotic fluid–derived cells, glia cells, and pancreatic β cells, as well as microglia, neurons, astrocytes, endothelial cells, oligodendrocytes, and brain pericytes (Stadtfeld et al., 2008; Tsai et al., 2010; Watanabe et al., 2011; Zhou et al., 2011; Meng et al., 2012; Montserrat et al., 2012). Moreover, co-culture models of multiple brain cell types have been developed to simulate the complex interactions between neuronal cells in vivo. Improvements of differentiation methods to increase the maturity, yield, and purity of brain cell types, and the developments of co-culture and three-dimensional (3D) models to better imitate the pathologies of AD remain in development (Choi et al., 2014). Further improving these reprogramming strategies and models has promising potential to facilitate neurodegenerative disease research and clinical applications (Fig. 2).

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

Potential applications of developing iPSC technology in AD modeling and drug discovery. iPSCs edited from somatic cells can differentiate into multiple neuronal cells, which can simulate the complex interactions between neuronal cells in vivo by 3D co-culture. These reprogramming strategy and models have promising potential to facilitate neurodegenerative disease research, drug discovery, and clinical applications. NPC: neural precursor cells

Age is the primary risk factor for neurodegenerative diseases, including AD; therefore, using stem cells to study AD may seem counterintuitive. However, in the very early stages of differentiation, neurons differentiated from iPSCs with FAD mutations, or iPSCs from patients with AD, exhibit AD-related phenotypes (Ochalek et al., 2017; Ortiz-Virumbrales et al., 2017; Wezyk et al., 2018). These alterations parallel the stages of AD progression, which are understudied in vivo. Genomewide association studies have shown that alterations in many different genes can promote the development of AD, and different genetic changes in patients with AD shared pathologic manifestations in some cases (De Strooper and Karran, 2016). Generating specific individual brain cells of iPSCs has potential applications for patient-specific treatment (Chen et al., 2016; Cota-Coronado et al., 2019).

Neurons.

Numerous neurodegenerative diseases that occur during aging attest that brain neuronal cells, as nondividing cells, face major challenges in maintaining normal function and health during the multiple decades of life. A better understanding of the mechanisms may help ensure the health and survival of neurons. With the development and application of iPSCs technology, more and more literature has been published demonstrating FAD or SAD modeling using iPSCs (Chambers et al., 2009). iPSCs can differentiate into neural progenitor cells, after which the neural progenitor cells are patterned to different neuronal lineages (Maroof et al., 2013; Nicholas et al., 2013). There are numerous neuron subtypes, including dopaminergic neurons, glutamatergic neurons, GABAergic neurons, and cholinergic neurons (Soldner et al., 2009; Zhang et al., 2013; Begum et al., 2015; Sun et al., 2016). Glutamatergic neurons harboring mutated amyloid precursor protein (APP) V717I were observed to have elevated β-secretase cleavage of APP and increased levels of both Aβ and tau phosphorylation (Muratore et al., 2014). In contrast, neurons harboring mutated APP A673T were found to have reduced β-secretase cleavage of APP and production of Aβ (Maloney et al., 2014). Neurons expressing mutated APP K670N/M671L or APP V717I exhibited impaired low-density lipoprotein endocytosis, reduced mitophagy, cellular uptake defects, and degradation pathway impairment compared with neurons carrying APP duplications (Knappenberger et al., 2004; Israel et al., 2012; Fang et al., 2019). Patients with Down syndrome develop early-onset dementia. The Down syndrome iPSC neurons accumulate tau hyperphosphorylation and Aβ deposits, similar to that caused by mutations in FAD (Shi et al., 2012; Chang et al., 2015; Dashinimaev et al., 2017; Ovchinnikov et al., 2018). Glutamatergic neurons derived from presenilin 1 (PSEN1) null and PSEN1 ΔE9 mutations iPSCs have been shown to gain γ-secretase function via loss or gain of function without loss of other functions (Knappenberger et al., 2004; Wang et al., 2018). Neurons from iPSCs harboring mutated PSEN1 V89L, PSEN1 A246E, and PSEN1 L150P were shown to be more sensitive to oxidative stress and Aβ-induced toxicity than those from healthy individuals (Armijo et al., 2017; Ochalek et al., 2017). iPSCs derived from SAD often share the same phenotypes with those with FAD mutations. In addition to the elevated tau phosphorylation and Aβ accumulation, the SAD-iPSC neurons show activation of endoplasmic reticulum (ER) stress, elevated DNA damage, enlarged endosomes, activation of oxidative stress pathways, and mitochondrial dysfunction (Duan et al., 2014; Birnbaum et al., 2018).

Astrocytes.

Astrocytes are the most abundant cell type in brains, which play essential roles in providing energetic, trophic, physical, and metabolic support to other brain cells (Molofsky and Deneen, 2015; Weber and Barros, 2015; Liddelow et al., 2017). Multiple protocols have been developed to differentiate iPSCs into astrocytes (Krencik et al., 2011; Emdad et al., 2012). Altered marker protein localization and decreased morphologic complexity were exhibited in astrocytes harboring both SAD-linked APOE4 and FAD-linked PSEN1 M146L mutations (Jones et al., 2017). Increased reactive oxygen species production, impaired fatty acid oxidation, elevated release, and reduced uptake of Aβ42 were observed in astrocytes carrying the PSEN1 ΔE9 mutation (Oksanen et al., 2017; Konttinen et al., 2019). When co-cultured with human neurons, astrocytes derived from iPSCs promote the maturation and survival of neurons. However, the effects can be impaired by APOE4 and PSEN1 ΔE9 mutations (Kuijlaars et al., 2016; Zhao et al., 2017). Moreover, the APOE4 mutated astrocytes exhibit a reduced ability to internalize Aβ42 and extensive gene expression alterations compared with APOE3-mutated astrocytes (Zhao et al., 2017; Lin et al., 2018).

Microglia.

Microglia are brain immune cells that play numerous roles, including clearance of dying cells, synaptic pruning, and regulation of neuroinflammation in the brain (Salter and Beggs, 2014; Heppner et al., 2015). In the past 10 years, numerous microglial genes have been identified as risk factors for AD by improved sequencing technologies. These genes include Cas scaffolding protein family member 4, triggering receptor expressed on myeloid cells 2, phosphatidylinositol-3,4,5-trisphosphate 5-phosphatase 1, sialic acid–binding immunoglobulin-like lectin 3, SPI1, and HLA-DRB1 (Guerreiro et al., 2013; Jonsson et al., 2013; Lambert et al., 2013; Huang et al., 2017). These findings indicate that microglial dysfunction might contribute to the development of AD. Microglia induced from healthy individuals iPSCs are capable of Aβ uptake, synaptic pruning, and phagocytosis. After exogenous Aβ treatment, the microglia showed altered gene expression and secreted various cytokines (Muffat et al., 2016; Abud et al., 2017). Microglia derived from iPSCs of patients with SAD exhibited elevated release of specific cytokines and altered phagocytosis after lipopolysaccharide treatment (Xu et al., 2019). Microglia carrying mutated APOE4 showed an impaired ability to internalize Aβ, extensive gene expression, and reduced morphologic complexity compared with isogenic APOE3 controls (Lin et al., 2018).

Oligodendrocytes.

The function of oligodendrocytes is to generate a myelin sheath that wraps the axons of nerve cells and forms the white matter of the brain. Oligodendrocytes also mediate inflammation, providing trophic support and contributing to metabolism regulation in the brain (Simons and Nave, 2015; Dimou and Simons, 2017). Oligodendrocyte dysfunction and white matter loss were observed in mice and patients with AD, which could lead to neuronal dysfunction (Desai et al., 2010; Bartzokis, 2011). Recent studies have reported the induction of oligodendrocytes from 3D organoid models (Hu et al., 2009; Wang et al., 2013; Madhavan et al., 2018). However, there are no studies on human systems to examine AD-related oligodendrocyte function. Selected critical AD studies utilizing iPSCs are summarized in Table 1.

View this table:
  • View inline
  • View popup
TABLE 1

Summary of selected important AD studies utilizing iPSCs

Applications of NSCs in AD Modeling

Transplantation of NSCs has been investigated as a prospective therapeutic approach for neurodegenerative diseases, including AD. NSCs are one type of multipotent stem cells that can differentiate into neurons, oligodendrocytes, microglia, and astrocytes (Massirer et al., 2011; Martínez-Morales et al., 2013; Berger et al., 2020; Si et al., 2020). NSCs can be extracted from brain tissues, differentiated from iPSCs and embryonic stem cells, or reprogrammed from somatic cells (Hermann and Storch, 2013; Wen and Jin, 2014; Shahbazi et al., 2018). Transplanted NSCs are capable of secreting neurotrophic factors and replacing damaged neural circuitry to alter lesion protein levels or counter symptomatic deterioration (Liu et al., 2013; Liu et al., 2014; Telias and Ben-Yosef, 2015; Kim et al., 2018). The neurotrophic factors NSCs secrete have been shown to improve memory function, and NSCs overexpressing β-degrading enzyme have been shown to reduce the aggregation of Aβ (Tang et al., 2008; Wu et al., 2016; Marsh and Blurton-Jones, 2017).

The 3xTg mouse is a FAD-related triple-transgenic mouse model, which carries three mutations: APP Swedish, PSEN1 M146V, and MAPT P301L (Oddo et al., 2003; Billings et al., 2005). After human-derived NSCs transplantation in 3xTg mice, Aβ and tau protein levels remained unchanged, but the memory function and synaptic density improved, indicating that the transplantation of human-derived NSCs may only reverse symptoms (Chen et al., 2014; Ager et al., 2015). Mathew et al. showed that mouse-derived NSCs transplantation in 3xTg mice produced similar results as human-derived NSCs(Blurton-Jones et al., 2009). After transplantation, cognitive impairment was rescued and synaptic density was enhanced without altering the Aβ and tau levels (Blurton-Jones et al., 2014). Transplantation of modified NSCs carrying neprilysin showed to be more effective in delivery than vector-delivered neprilysin, indicating NSCs can act as effective delivery vehicles (Kim et al., 2013; Blurton-Jones et al., 2014). A different source of NSCs may release various neurotrophins and have a distinct neurogenesis. BDNF is a member of the neurotrophin family and is involved in the mouse-derived NSCs recovery of synaptic connectivity, but it remains unknown which trophic factors are involved in synaptogenesis of human-derived NSCs (Ager et al., 2015). Studies on mouse-derived NSCs showed that cognitive improvement depends mainly on the precise differentiation of NSCs, whereas lineage-specific differentiation of human-derived NSCs had limited effect on cognitive function (Blurton-Jones et al., 2014; Chen et al., 2014). Tg2576 mice harbor the human Swedish APP mutation (isoform 695; KM670/671NL) (Hsiao et al., 1996; Kawarabayashi et al., 2001; King and Arendash, 2002), and reduced Aβ production and acetylcholinesterase were observed after NSC transplantation into these mice. Furthermore, many astrocytes expressing α7 nicotinic receptors were found to repair damaged neurons, and the endogenous neurogenesis was enhanced in the transplant region of Tg2576 mice (Lilja et al., 2015). Anti-inflammatory cytokine levels were significantly higher in microglial cells and could inhibit Aβ production and promote Aβ clearance rate when NSCs were transplanted into Tg2576 mice at early stages of disease. Moreover, synaptic density, vascular endothelial growth factor (VEGF), and neurogenesis were increased after transplantation. Timely intervention is essential since these results were not obtained when NSCs were transplanted into Tg2576 mice at later stages (Kim et al., 2015; Haiyan et al., 2016). APP/PS1 mice are widely used as an AD mouse model and harbor both the Swedish and PSEN1 (L166P) mutations (Maia et al., 2013). Enhanced synaptic formation without a change in Aβ levels was observed after transplantation of NSCs into APP/PS1 mice (Li et al., 2016). In contrast, McGinley’s study suggested that NSC transplantation reduces Aβ levels by regulating microglial activation (Zhang et al., 2015; McGinley et al., 2018). Administration of NSCs in APP/PS1 mice also resulted in enhanced levels of tropomyosin receptor kinase B and BDNF. The expression of the NMDA receptor 2B subunit, which plays a critical role in memory and learning function, was also increased, resulting in improved the cognitive function (Zhang et al., 2014). Cholinergic-like neurons derived from NSCs were also introduced into APP/PS1 mice. This showed that cholinergic acetyltransferase’s concentration and activity were elevated, and there was an increase in functional dendrites (Gu et al., 2015). In another study, astrocytes and microglia activity was decreased, which regulates the Toll-like receptor 4 signaling pathway, leading to a decrease in neuroinflammatory response and the improvement of cognitive function (Zhang et al., 2016). Another AD mice model is the 5xFAD mouse, which harbors five mutations: the PSEN1, Florida (I716V), Swedish (K670N/M671L), London (V717I), M146L, and L286V mutations (Oakley et al., 2006; Jawhar et al., 2012). These mice are immune deficient, allowing long-term safety and efficacy evaluation of NSCs transplantation. A clinical-grade NSC line failed to differentiate and had no impact on synapses after successful engraftment into 5xFAD mice for up to 5 months. The protein levels of BDNF and Aβ did not change, and no improvement in behavior impairment was observed (Marsh et al., 2017). In contrast, rapid differentiation and reconstruction of functional neural circuits were observed after transplantation of reprogrammed NPCs from human mononuclear cells. BDNF levels increased, and improvement in behavior impairment was observed after 5–6 months (Zhang et al., 2019). These two studies indicate that, compared with normal NSCs, reprogrammed somatic cells might have greater neural lineage–specific differentiation capacity. Selected AD studies utilizing NSCs are summarized in Table 2.

View this table:
  • View inline
  • View popup
TABLE 2

Summary of selected important AD studies utilizing NSCs

Application of MSCs in AD Modeling

MSCs are a type of pluripotent stem cell with self-renewing, immunomodulatory properties, that have limited differentiation capacity (Song et al., 2020). MSCs can differentiate into chondrocytes, osteocytes, fibroblasts, and adipocytes (Ankrum et al., 2014; Si et al., 2019). Unlike iPSCs and NSCs, MSCs are not expected to replace the impaired neurons and incorporate into neuronal networks because it is controversial whether MSCs can differentiate into ectodermal or endodermal cells (Robert et al., 2020; Varderidou-Minasian and Lorenowicz, 2020). MSCs can be distinguished from other cell types by the expression of CD105, CD90, CD73, and CD44 and by the lack of CD14, CD45, CD19, CD11b, CD34, and HLA-DRB1 expression (Bari et al., 2019; Elahi et al., 2020). MSCs can be harvested from many tissues, including adipose tissue, umbilical cord tissue, bone marrow, fetal tissues, placental tissues, dental pulp, and peripheral blood (Keane et al., 2017; Sá da Bandeira et al., 2017). MSCs have neuroprotective effects in addition to antifibrotic, anti-inflammatory, antibacterial, antitumorigenic, chemoattractive, antiapoptotic, proangiogenic, and tissue repair effects (Pierro et al., 2017; Naji et al., 2019). There are multiple mechanisms behind the neuroprotective effects of MSCs. MSCs can secrete neurotrophic growth factors such as BDNF and glial cell–derived neurotrophic factor to improve the survival of neuronal cells (Teixeira et al., 2017; Hao et al., 2018). It is well known that MSCs can modulate the immune system, and neuroinflammation has been reported to play a pathomechanistic role in neurodegenerative diseases. When MSCs enter the neuroinflammatory milieu, they will release proinflammatory and anti-inflammatory factors, and activated T cells can interact with neuronal cells to reduce neuronal death (Ransohoff, 2016). Secreted biologic factors such as messenger RNA, proteins, or microRNA via extracellular vesicles are other mechanisms to improve neuronal survival (Richards et al., 2016). Finally, a novel hypothesis to the neuroprotective effects of MSCs is that MSCs improve neuronal health by donating their mitochondria to neurons carrying dysfunctional mitochondria (Zhao et al., 2013; Glenn and Whartenby, 2014).

In the APP/PS1 mouse model of AD, bone marrow–derived MSCs were transplanted via tail vein injection. These mice were found to have a reduction in microglial numbers without alteration in the numbers of amyloid plaques (Naaldijk et al., 2017). In contrast, a study by Carter et al. showed a significant decrease after intracerebral injection of bone marrow–derived MSCs compared with controls treated with PBS 2 months after injection (Bae et al., 2013). The synaptic transmission-related proteins such as synapsin 1 and dynamin 1 were considerably enhanced in AD mice brains compared with control groups after treatment with bone marrow–derived MSCs (Bae et al., 2013). In another study, human umbilical cord–derived MSCs were induced to neuron-like cells and transplanted into the APP/PS1 AD mouse model. In this model, increased synapsin 1 levels, improved cognitive function, and reduced Aβ deposition were found. The “alternatively activated” microglia (M2-like microglia) and interleukin-4, an anti-inflammatory cytokine associated with M2-like microglia, were increased. Furthermore, the proinflammatory cytokines tumor necrosis factor-α and interleukin-4 were decreased significantly (Yang et al., 2013). Neprilysin and insulin-degrading enzyme, two Aβ-degrading factors, increased after treatment with neuron-like cells from human umbilical cord–derived MSCs (Lee et al., 2012). One study reported that MSC transplantation improved AD cognition, and that the pathology may be mediated through modulating tissue repair factors and inflammatory events (Lee et al., 2010). The Wnt signaling pathway has been reported to be involved in the MSC-regulated neurogenesis in an AD mouse model. Se et al. found that the expression of GFAP, nestin, Ki-67, HuD, and SOX2 significantly increased in Aβ-treated neural progenitor cells co-cultured with MSCs as compared with Aβ treatment alone (Oh et al., 2015). Additionally, β-catenin and Ngn1 expression were enhanced in Aβ-treated neural progenitor cells co-cultured with MSCs (Oh et al., 2015). In AD mouse models, the MSC’s effects on mitochondrial function have not yet been studied. Selected AD studies utilizing MSCs are summarized in Table 3.

View this table:
  • View inline
  • View popup
TABLE 3

Summary of selected important AD studies utilizing MSCs

Challenges and Future Perspectives

The development of stem cell technologies allows the use of differentiated human cells for mutagenesis and drug screening (Hirschi et al., 2014; Sproul, 2015). In the past few decades, many promising preclinical and early clinical findings were obtained. However, many challenges remain regarding the application of stem cells as therapeutic approaches in AD. The development of stem cell technologies also raises the attractive possibility of personalized and regenerative medicine (Sproul, 2015; Chen et al., 2016; Cota-Coronado et al., 2019). Genomic instability of iPSCs, however, is a serious issue for both experimental studies and regenerative medicine. Limited passage numbers and regular checks of genomic alterations in iPSC lines are the most commonly used methods to prevent issues (Kwon et al., 2017; Zhang et al., 2018). Even though the use of integration-free delivery systems has reduced the genomic alterations in iPSCs, it remains an active topic of investigation to reduce genomic instability (Rebuzzini et al., 2016; Yoshihara et al., 2017). Expanding brain cell subtypes generated from iPSCs, improving differentiation protocols of iPSCs, and developing more suitable and complex 3D co-culture systems are important goals for iPSC research. How to improve the reproducibility and consistency of cell subtypes obtained from iPSCs also remains unknown. Despite the use of the same protocol, there is considerable variability in gene expression and cellular morphology (Mills et al., 2013; Volpato et al., 2018). To minimize such variability, better standardization of growth conditions and differentiation techniques, adoption of rigorous statistical analyses, and more thorough reporting of methodologies should be established (Lin, 2011; Sullivan et al., 2018). Another challenge is how to generate iPSC-derived brain cells that can adequately mimic the growth and maturation of various cell types in the brain. Signals from other cell types are critical during this process to shape their identity (Cahoy et al., 2008; Gosselin et al., 2014; Bohlen et al., 2017). To resemble cell counterparts more closely, we need to better understand the critical signals involved in the process. The 3D co-culture systems are important models for stem cell application in neurodegeneration research, promoting the development of hallmark pathologies in AD that cannot be found in two-dimensional cultures (Camp et al., 2015; Sloan et al., 2017). The 3D co-culture systems also provide a platform to help develop a better understanding of the complex, interrelated functions and interactions between all cell types in the brain. An ideal 3D co-culture system for AD should include each type of glial cell, all neuronal subtypes, and the blood-brain barrier components (Choi et al., 2014). In many cases, only some aspects of brain function are established in a reduced system model. In the deeper layers, organoids often show dysfunction and cell death, and the introduction of functional vasculature would likely improve this situation. The “bioreactors” used in culture may also improve the health of the cells (Marion et al., 2009; Corti et al., 2012; Baxter et al., 2015; Huh et al., 2016; Mertens et al., 2018).

The iPSC models to mimic AD have been questioned since the age- and environment-dependent epigenetic and cellular signatures may be lost during reprogramming (Roessler et al., 2014). To overcome this problem, somatic cells are reprogrammed directly into neuronal cells, bypassing the iPSC stage. The reprogrammed have improved capability to retain age-related transcriptomic and cellular alterations compared with iPSCs (Victor et al., 2018). There are also limitations for direct reprogramming due to the low yield of reprogrammed cells and poor reprogramming efficiency (Mertens et al., 2018). Unlike iPSCs, the beneficial role of NSCs in AD is to increase the levels of neurotrophic factors, restore local neuron populations, and increase synaptic density rather than modulate pathologic protein levels (Zhang et al., 2004; Zheng et al., 2017; Omole and Fakoya, 2018). However, how long this phenomenon can persist with altering the pathologic protein levels and what role NSCs may play in this process remains unknown. As stem cell techniques continue to be refined, novel stem cell–based therapies can be adequately validated and may reveal effective therapeutics that lead to further targeted drug development for AD, ultimately moving the field forward.

Although the literature is replete with therapeutic interventions pursued based on expert opinion and patient acceptance, stem cell transplantation risks cannot be ignored. There is a successful example that a patient received multiple injections of different source-derived allogeneic stem cells to reduce neurologic deficits from a middle cerebral artery stroke (Berkowitz et al., 2016). Despite fewer safety concerns than allogeneic stem cells, applications of autologous stem cells may raise notably adverse events. A reported case showed that hematopoietic stem cells injection into kidneys of a patient with renal failure were associated with tumor development and ultimately led to nephrectomy (Thirabanjasak et al., 2010). However, we firmly believe that by resolving the unique challenges in clinic, stem cell therapies can provide an important, safe, and effective strategy to patients who need it.

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Si, Wang.

Footnotes

    • Received September 7, 2020.
    • Accepted February 3, 2021.
  • This work was supported by Harbin Medical University Scientific Research Innovation Fund [Grant 2017JCZX38].

  • https://doi.org/10.1124/jpet.120.000324.

Abbreviations

Aβ
amyloid beta
AD
Alzheimer’s disease
ApoE
apolipoprotein E
APP
amyloid precursor protein
BDNF
brain-derived neurotrophic factor
CSF
cerebrospinal fluid
3D
three-dimensional
FAD
familial Alzheimer’s disease
iPSC
induced pluripotent stem cell
MSC
mesenchymal stem cell
NSC
neural stem cell
SAD
sporadic late-onset Alzheimer’s disease
Sox2
sex determining region Y-box 2
  • Copyright © 2021 by The Author(s)

This is an open access article distributed under the CC BY-NC Attribution 4.0 International license.

References

  1. ↵
    1. Abud EM,
    2. Ramirez RN,
    3. Martinez ES,
    4. Healy LM,
    5. Nguyen CHH,
    6. Newman SA,
    7. Yeromin AV,
    8. Scarfone VM,
    9. Marsh SE,
    10. Fimbres C, et al.
    (2017) iPSC-derived human microglia-like cells to study neurological diseases. Neuron 94:278–293.e9.
    OpenUrlCrossRefPubMed
  2. ↵
    1. Ager RR,
    2. Davis JL,
    3. Agazaryan A,
    4. Benavente F,
    5. Poon WW,
    6. LaFerla FM, and
    7. Blurton-Jones M
    (2015) Human neural stem cells improve cognition and promote synaptic growth in two complementary transgenic models of Alzheimer’s disease and neuronal loss. Hippocampus 25:813–826.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Ankrum JA,
    2. Ong JF, and
    3. Karp JM
    (2014) Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol 32:252–260.
    OpenUrlCrossRefPubMed
  4. ↵
    1. Anokye-Danso F,
    2. Trivedi CM,
    3. Juhr D,
    4. Gupta M,
    5. Cui Z,
    6. Tian Y,
    7. Zhang Y,
    8. Yang W,
    9. Gruber PJ,
    10. Epstein JA, et al.
    (2011) Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell 8:376–388.
    OpenUrlCrossRefPubMed
  5. ↵
    1. Aoi T
    (2008) [Advance in study of induced pluripotent stem cells (iPS cells)]. Nihon Rinsho 66:850–856.
    OpenUrl
  6. ↵
    1. Armijo E,
    2. Gonzalez C,
    3. Shahnawaz M,
    4. Flores A,
    5. Davis B, and
    6. Soto C
    (2017) Increased susceptibility to Aβ toxicity in neuronal cultures derived from familial Alzheimer’s disease (PSEN1-A246E) induced pluripotent stem cells. Neurosci Lett 639:74–81.
    OpenUrlCrossRefPubMed
  7. ↵
    1. Arvanitakis Z,
    2. Shah RC, and
    3. Bennett DA
    (2019) Diagnosis and management of dementia: review. JAMA 322:1589–1599.
    OpenUrlPubMed
  8. ↵
    1. Bae JS,
    2. Jin HK,
    3. Lee JK,
    4. Richardson JC, and
    5. Carter JE
    (2013) Bone marrow-derived mesenchymal stem cells contribute to the reduction of amyloid-β deposits and the improvement of synaptic transmission in a mouse model of pre-dementia Alzheimer’s disease. Curr Alzheimer Res 10:524–531.
    OpenUrl
  9. ↵
    1. Bari E,
    2. Ferrarotti I,
    3. Torre ML,
    4. Corsico AG, and
    5. Perteghella S
    (2019) Mesenchymal stem/stromal cell secretome for lung regeneration: the long way through “pharmaceuticalization” for the best formulation. J Control Release 309:11–24.
    OpenUrl
  10. ↵
    1. Bartzokis G
    (2011) Alzheimer’s disease as homeostatic responses to age-related myelin breakdown. Neurobiol Aging 32:1341–1371.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Baxter M,
    2. Withey S,
    3. Harrison S,
    4. Segeritz CP,
    5. Zhang F,
    6. Atkinson-Dell R,
    7. Rowe C,
    8. Gerrard DT,
    9. Sison-Young R,
    10. Jenkins R, et al.
    (2015) Phenotypic and functional analyses show stem cell-derived hepatocyte-like cells better mimic fetal rather than adult hepatocytes. J Hepatol 62:581–589.
    OpenUrlCrossRefPubMed
  12. ↵
    1. Begum AN,
    2. Guoynes C,
    3. Cho J,
    4. Hao J,
    5. Lutfy K, and
    6. Hong Y
    (2015) Rapid generation of sub-type, region-specific neurons and neural networks from human pluripotent stem cell-derived neurospheres. Stem Cell Res 15:731–741.
    OpenUrl
  13. ↵
    1. Benek O,
    2. Korabecny J, and
    3. Soukup O
    (2020) A perspective on multi-target drugs for Alzheimer’s disease. Trends Pharmacol Sci 41:434–445.
    OpenUrl
  14. ↵
    1. Berger T,
    2. Lee H,
    3. Young AH,
    4. Aarsland D, and
    5. Thuret S
    (2020) Adult hippocampal neurogenesis in major depressive disorder and Alzheimer’s disease. Trends Mol Med 26:803–818.
    OpenUrl
  15. ↵
    1. Berkowitz AL,
    2. Miller MB,
    3. Mir SA,
    4. Cagney D,
    5. Chavakula V,
    6. Guleria I,
    7. Aizer A,
    8. Ligon KL, and
    9. Chi JH
    (2016) Glioproliferative lesion of the spinal cord as a complication of “stem-cell tourism”. N Engl J Med 375:196–198.
    OpenUrlCrossRefPubMed
  16. ↵
    1. Billings LM,
    2. Oddo S,
    3. Green KN,
    4. McGaugh JL, and
    5. LaFerla FM
    (2005) Intraneuronal Abeta causes the onset of early Alzheimer’s disease-related cognitive deficits in transgenic mice. Neuron 45:675–688.
    OpenUrlCrossRefPubMed
  17. ↵
    1. Birnbaum JH,
    2. Wanner D,
    3. Gietl AF,
    4. Saake A,
    5. Kündig TM,
    6. Hock C,
    7. Nitsch RM, and
    8. Tackenberg C
    (2018) Oxidative stress and altered mitochondrial protein expression in the absence of amyloid-β and tau pathology in iPSC-derived neurons from sporadic Alzheimer’s disease patients. Stem Cell Res 27:121–130.
    OpenUrlCrossRefPubMed
  18. ↵
    1. Blurton-Jones M,
    2. Kitazawa M,
    3. Martinez-Coria H,
    4. Castello NA,
    5. Müller FJ,
    6. Loring JF,
    7. Yamasaki TR,
    8. Poon WW,
    9. Green KN, and
    10. LaFerla FM
    (2009) Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc Natl Acad Sci USA 106:13594–13599.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Blurton-Jones M,
    2. Spencer B,
    3. Michael S,
    4. Castello NA,
    5. Agazaryan AA,
    6. Davis JL,
    7. Müller FJ,
    8. Loring JF,
    9. Masliah E, and
    10. LaFerla FM
    (2014) Neural stem cells genetically-modified to express neprilysin reduce pathology in Alzheimer transgenic models. Stem Cell Res Ther 5:46.
    OpenUrl
  20. ↵
    1. Bohlen CJ,
    2. Bennett FC,
    3. Tucker AF,
    4. Collins HY,
    5. Mulinyawe SB, and
    6. Barres BA
    (2017) Diverse requirements for microglial survival, specification, and function revealed by defined-medium cultures. Neuron 94:759–773.e8.
    OpenUrlCrossRefPubMed
  21. ↵
    1. Brier MR,
    2. Gordon B,
    3. Friedrichsen K,
    4. McCarthy J,
    5. Stern A,
    6. Christensen J,
    7. Owen C,
    8. Aldea P,
    9. Su Y,
    10. Hassenstab J, et al.
    (2016) Tau and Aβ imaging, CSF measures, and cognition in Alzheimer’s disease. Sci Transl Med 8:338ra66.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Bruni AC,
    2. Bernardi L, and
    3. Gabelli C
    (2020) From beta amyloid to altered proteostasis in Alzheimer’s disease. Ageing Res Rev 64:101126.
    OpenUrl
  23. ↵
    1. Buganim Y and
    2. Jaenisch R
    (2012) Transdifferentiation by defined factors as a powerful research tool to address basic biological questions. Cell Cycle 11:4485–4486.
    OpenUrlCrossRef
  24. ↵
    1. Cahoy JD,
    2. Emery B,
    3. Kaushal A,
    4. Foo LC,
    5. Zamanian JL,
    6. Christopherson KS,
    7. Xing Y,
    8. Lubischer JL,
    9. Krieg PA,
    10. Krupenko SA, et al.
    (2008) A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J Neurosci 28:264–278.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Camp JG,
    2. Badsha F,
    3. Florio M,
    4. Kanton S,
    5. Gerber T,
    6. Wilsch-Bräuninger M,
    7. Lewitus E,
    8. Sykes A,
    9. Hevers W,
    10. Lancaster M, et al.
    (2015) Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc Natl Acad Sci USA 112:15672–15677.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Chambers SM,
    2. Fasano CA,
    3. Papapetrou EP,
    4. Tomishima M,
    5. Sadelain M, and
    6. Studer L
    (2009) Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol 27:275–280.
    OpenUrlCrossRefPubMed
  27. ↵
    1. Chang CY,
    2. Chen SM,
    3. Lu HE,
    4. Lai SM,
    5. Lai PS,
    6. Shen PW,
    7. Chen PY,
    8. Shen CI,
    9. Harn HJ,
    10. Lin SZ, et al.
    (2015) N-butylidenephthalide attenuates Alzheimer’s disease-like cytopathy in Down syndrome induced pluripotent stem cell-derived neurons. Sci Rep 5:8744.
    OpenUrlCrossRefPubMed
  28. ↵
    1. Chen IY,
    2. Matsa E, and
    3. Wu JC
    (2016) Induced pluripotent stem cells: at the heart of cardiovascular precision medicine. Nat Rev Cardiol 13:333–349.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Chen J,
    2. Gao Y,
    3. Huang H,
    4. Xu K,
    5. Chen X,
    6. Jiang Y,
    7. Li H,
    8. Gao S,
    9. Tao Y,
    10. Wang H, et al.
    (2015) The combination of Tet1 with Oct4 generates high-quality mouse-induced pluripotent stem cells. Stem Cells 33:686–698.
    OpenUrl
  30. ↵
    1. Chen SQ,
    2. Cai Q,
    3. Shen YY,
    4. Wang PY,
    5. Li MH, and
    6. Teng GY
    (2014) Neural stem cell transplantation improves spatial learning and memory via neuronal regeneration in amyloid-β precursor protein/presenilin 1/tau triple transgenic mice. Am J Alzheimers Dis Other Demen 29:142–149.
    OpenUrlCrossRefPubMed
  31. ↵
    1. Chia NY,
    2. Chan YS,
    3. Feng B,
    4. Lu X,
    5. Orlov YL,
    6. Moreau D,
    7. Kumar P,
    8. Yang L,
    9. Jiang J,
    10. Lau MS, et al.
    (2010) A genome-wide RNAi screen reveals determinants of human embryonic stem cell identity. Nature 468:316–320.
    OpenUrlCrossRefPubMed
  32. ↵
    1. Choi SH,
    2. Kim YH,
    3. Hebisch M,
    4. Sliwinski C,
    5. Lee S,
    6. D’Avanzo C,
    7. Chen H,
    8. Hooli B,
    9. Asselin C,
    10. Muffat J, et al.
    (2014) A three-dimensional human neural cell culture model of Alzheimer’s disease. Nature 515:274–278.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Chuah JKC and
    2. Zink D
    (2017) Stem cell-derived kidney cells and organoids: recent breakthroughs and emerging applications. Biotechnol Adv 35:150–167.
    OpenUrl
  34. ↵
    1. Congdon EE and
    2. Sigurdsson EM
    (2018) Tau-targeting therapies for Alzheimer disease. Nat Rev Neurol 14:399–415.
    OpenUrlCrossRefPubMed
  35. ↵
    1. Corti S,
    2. Nizzardo M,
    3. Simone C,
    4. Falcone M,
    5. Donadoni C,
    6. Salani S,
    7. Rizzo F,
    8. Nardini M,
    9. Riboldi G,
    10. Magri F, et al.
    (2012) Direct reprogramming of human astrocytes into neural stem cells and neurons. Exp Cell Res 318:1528–1541.
    OpenUrlCrossRefPubMed
  36. ↵
    1. Cota-Coronado A,
    2. Ramírez-Rodríguez PB,
    3. Padilla-Camberos E,
    4. Díaz EF,
    5. Flores-Fernández JM,
    6. Ávila-Gónzalez D, and
    7. Diaz-Martinez NE
    (2019) Implications of human induced pluripotent stem cells in metabolic disorders: from drug discovery toward precision medicine. Drug Discov Today 24:334–341.
    OpenUrl
  37. ↵
    1. Cummings J
    (2019) The National Institute on Aging-Alzheimer’s Association framework on Alzheimer’s disease: application to clinical trials. Alzheimers Dement 15:172–178.
    OpenUrl
  38. ↵
    1. Dashinimaev EB,
    2. Artyuhov AS,
    3. Bolshakov AP,
    4. Vorotelyak EA, and
    5. Vasiliev AV
    (2017) Neurons derived from induced pluripotent stem cells of patients with down syndrome reproduce early stages of Alzheimer’s disease type pathology in vitro. J Alzheimers Dis 56:835–847.
    OpenUrl
  39. ↵
    1. De Strooper B and
    2. Karran E
    (2016) The cellular phase of Alzheimer’s disease. Cell 164:603–615.
    OpenUrlCrossRefPubMed
  40. ↵
    1. Desai MK,
    2. Mastrangelo MA,
    3. Ryan DA,
    4. Sudol KL,
    5. Narrow WC, and
    6. Bowers WJ
    (2010) Early oligodendrocyte/myelin pathology in Alzheimer’s disease mice constitutes a novel therapeutic target. Am J Pathol 177:1422–1435.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Devalla HD and
    2. Passier R
    (2018) Cardiac differentiation of pluripotent stem cells and implications for modeling the heart in health and disease. Sci Transl Med 10:eaah5457.
    OpenUrlFREE Full Text
  42. ↵
    1. Di Lullo E and
    2. Kriegstein AR
    (2017) The use of brain organoids to investigate neural development and disease. Nat Rev Neurosci 18:573–584.
    OpenUrlCrossRefPubMed
  43. ↵
    1. Di Stefano F,
    2. Kas A,
    3. Habert MO,
    4. Decazes P,
    5. Lamari F,
    6. Lista S,
    7. Hampel H, and
    8. Teichmann M
    (2016) The phenotypical core of Alzheimer’s disease-related and nonrelated variants of the corticobasal syndrome: a systematic clinical, neuropsychological, imaging, and biomarker study. Alzheimers Dement 12:786–795.
    OpenUrl
  44. ↵
    1. Dimou L and
    2. Simons M
    (2017) Diversity of oligodendrocytes and their progenitors. Curr Opin Neurobiol 47:73–79.
    OpenUrlCrossRefPubMed
  45. ↵
    1. Doege CA,
    2. Inoue K,
    3. Yamashita T,
    4. Rhee DB,
    5. Travis S,
    6. Fujita R,
    7. Guarnieri P,
    8. Bhagat G,
    9. Vanti WB,
    10. Shih A, et al.
    (2012) Early-stage epigenetic modification during somatic cell reprogramming by Parp1 and Tet2. Nature 488:652–655.
    OpenUrlCrossRefPubMed
  46. ↵
    1. Duan L,
    2. Bhattacharyya BJ,
    3. Belmadani A,
    4. Pan L,
    5. Miller RJ, and
    6. Kessler JA
    (2014) Stem cell derived basal forebrain cholinergic neurons from Alzheimer’s disease patients are more susceptible to cell death. Mol Neurodegener 9:3.
    OpenUrlCrossRefPubMed
  47. ↵
    1. Elahi FM,
    2. Farwell DG,
    3. Nolta JA, and
    4. Anderson JD
    (2020) Preclinical translation of exosomes derived from mesenchymal stem/stromal cells. Stem Cells 38:15–21.
    OpenUrl
  48. ↵
    1. Emdad L,
    2. D’Souza SL,
    3. Kothari HP,
    4. Qadeer ZA, and
    5. Germano IM
    (2012) Efficient differentiation of human embryonic and induced pluripotent stem cells into functional astrocytes. Stem Cells Dev 21:404–410.
    OpenUrlCrossRefPubMed
  49. ↵
    1. Fang EF,
    2. Hou Y,
    3. Palikaras K,
    4. Adriaanse BA,
    5. Kerr JS,
    6. Yang B,
    7. Lautrup S,
    8. Hasan-Olive MM,
    9. Caponio D,
    10. Dan X, et al.
    (2019) Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer’s disease. Nat Neurosci 22:401–412.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Glenn JD and
    2. Whartenby KA
    (2014) Mesenchymal stem cells: emerging mechanisms of immunomodulation and therapy. World J Stem Cells 6:526–539.
    OpenUrlCrossRefPubMed
  51. ↵
    1. Gosselin D,
    2. Link VM,
    3. Romanoski CE,
    4. Fonseca GJ,
    5. Eichenfield DZ,
    6. Spann NJ,
    7. Stender JD,
    8. Chun HB,
    9. Garner H,
    10. Geissmann F, et al.
    (2014) Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159:1327–1340.
    OpenUrlCrossRefPubMed
  52. ↵
    1. Gu G,
    2. Zhang W,
    3. Li M,
    4. Ni J, and
    5. Wang P
    (2015) Transplantation of NSC-derived cholinergic neuron-like cells improves cognitive function in APP/PS1 transgenic mice. Neuroscience 291:81–92.
    OpenUrl
  53. ↵
    1. Guerreiro R,
    2. Wojtas A,
    3. Bras J,
    4. Carrasquillo M,
    5. Rogaeva E,
    6. Majounie E,
    7. Cruchaga C,
    8. Sassi C,
    9. Kauwe JS,
    10. Younkin S, et al., and Alzheimer Genetic Analysis Group
    (2013) TREM2 variants in Alzheimer’s disease. N Engl J Med 368:117–127.
    OpenUrlCrossRefPubMed
  54. ↵
    1. Haiyan H,
    2. Rensong Y,
    3. Guoqin J,
    4. Xueli Z,
    5. Huaying X, and
    6. Yanwu X
    (2016) Effect of astragaloside IV on neural stem cell transplantation in Alzheimer’s disease rat models. Evid Based Complement Alternat Med 2016:3106980.
    OpenUrl
  55. ↵
    1. Hampel H,
    2. Mesulam MM,
    3. Cuello AC,
    4. Farlow MR,
    5. Giacobini E,
    6. Grossberg GT,
    7. Khachaturian AS,
    8. Vergallo A,
    9. Cavedo E,
    10. Snyder PJ, et al.
    (2018) The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 141:1917–1933.
    OpenUrl
  56. ↵
    1. Han SH,
    2. Park JC, and
    3. Mook-Jung I
    (2016) Amyloid β-interacting partners in Alzheimer’s disease: from accomplices to possible therapeutic targets. Prog Neurobiol 137:17–38.
    OpenUrl
  57. ↵
    1. Hao J,
    2. Li S,
    3. Shi X,
    4. Qian Z,
    5. Sun Y,
    6. Wang D,
    7. Zhou X,
    8. Qu H,
    9. Hu S,
    10. Zuo E, et al.
    (2018) Bone marrow mesenchymal stem cells protect against n-hexane-induced neuropathy through beclin 1-independent inhibition of autophagy. Sci Rep 8:4516.
    OpenUrlCrossRef
  58. ↵
    1. Heneka MT
    (2020) An immune-cell signature marks the brain in Alzheimer’s disease. Nature 577:322–323.
    OpenUrl
  59. ↵
    1. Heppner FL,
    2. Ransohoff RM, and
    3. Becher B
    (2015) Immune attack: the role of inflammation in Alzheimer disease. Nat Rev Neurosci 16:358–372.
    OpenUrlCrossRefPubMed
  60. ↵
    1. Hermann A and
    2. Storch A
    (2013) Induced neural stem cells (iNSCs) in neurodegenerative diseases. J Neural Transm (Vienna) 120 (Suppl 1):S19–S25.
    OpenUrl
  61. ↵
    1. Hirschi KK,
    2. Li S, and
    3. Roy K
    (2014) Induced pluripotent stem cells for regenerative medicine. Annu Rev Biomed Eng 16:277–294.
    OpenUrlCrossRefPubMed
  62. ↵
    1. Hsiao K,
    2. Chapman P,
    3. Nilsen S,
    4. Eckman C,
    5. Harigaya Y,
    6. Younkin S,
    7. Yang F, and
    8. Cole G
    (1996) Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science 274:99–102.
    OpenUrlAbstract/FREE Full Text
  63. ↵
    1. Hu BY,
    2. Du ZW, and
    3. Zhang SC
    (2009) Differentiation of human oligodendrocytes from pluripotent stem cells. Nat Protoc 4:1614–1622.
    OpenUrlCrossRefPubMed
  64. ↵
    1. Hu X,
    2. Zhang L,
    3. Mao SQ,
    4. Li Z,
    5. Chen J,
    6. Zhang RR,
    7. Wu HP,
    8. Gao J,
    9. Guo F,
    10. Liu W, et al.
    (2014) Tet and TDG mediate DNA demethylation essential for mesenchymal-to-epithelial transition in somatic cell reprogramming. Cell Stem Cell 14:512–522.
    OpenUrlCrossRefPubMed
  65. ↵
    1. Huang KL,
    2. Marcora E,
    3. Pimenova AA,
    4. Di Narzo AF,
    5. Kapoor M,
    6. Jin SC,
    7. Harari O,
    8. Bertelsen S,
    9. Fairfax BP,
    10. Czajkowski J, et al., and International Genomics of Alzheimer’s Project; Alzheimer’s Disease Neuroimaging Initiative
    (2017) A common haplotype lowers PU.1 expression in myeloid cells and delays onset of Alzheimer’s disease. Nat Neurosci 20:1052–1061.
    OpenUrlCrossRefPubMed
  66. ↵
    1. Huh CJ,
    2. Zhang B,
    3. Victor MB,
    4. Dahiya S,
    5. Batista LF,
    6. Horvath S, and
    7. Yoo AS
    (2016) Maintenance of age in human neurons generated by microRNA-based neuronal conversion of fibroblasts. eLife 5:e18648.
    OpenUrl
  67. ↵
    1. Israel MA,
    2. Yuan SH,
    3. Bardy C,
    4. Reyna SM,
    5. Mu Y,
    6. Herrera C,
    7. Hefferan MP,
    8. Van Gorp S,
    9. Nazor KL,
    10. Boscolo FS, et al.
    (2012) Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 482:216–220.
    OpenUrlCrossRefPubMed
  68. ↵
    1. Jafari Z,
    2. Kolb BE, and
    3. Mohajerani MH
    (2020) Neural oscillations and brain stimulation in Alzheimer’s disease. Prog Neurobiol 194:101878.
    OpenUrl
  69. ↵
    1. Jawhar S,
    2. Trawicka A,
    3. Jenneckens C,
    4. Bayer TA, and
    5. Wirths O
    (2012) Motor deficits, neuron loss, and reduced anxiety coinciding with axonal degeneration and intraneuronal Abeta aggregation in the 5XFAD mouse model of Alzheimer’s disease. Neurobiol Aging 33:196.e129–140.
    OpenUrl
  70. ↵
    1. Jeong S
    (2017) Molecular and cellular basis of neurodegeneration in Alzheimer’s disease. Mol Cells 40:613–620.
    OpenUrlCrossRef
  71. ↵
    1. Jones VC,
    2. Atkinson-Dell R,
    3. Verkhratsky A, and
    4. Mohamet L
    (2017) Aberrant iPSC-derived human astrocytes in Alzheimer’s disease. Cell Death Dis 8:e2696.
    OpenUrlCrossRef
  72. ↵
    1. Jonsson T,
    2. Stefansson H,
    3. Steinberg S,
    4. Jonsdottir I,
    5. Jonsson PV,
    6. Snaedal J,
    7. Bjornsson S,
    8. Huttenlocher J,
    9. Levey AI,
    10. Lah JJ, et al.
    (2013) Variant of TREM2 associated with the risk of Alzheimer’s disease. N Engl J Med 368:107–116.
    OpenUrlCrossRefPubMed
  73. ↵
    1. Judson RL,
    2. Babiarz JE,
    3. Venere M, and
    4. Blelloch R
    (2009) Embryonic stem cell-specific microRNAs promote induced pluripotency. Nat Biotechnol 27:459–461.
    OpenUrlCrossRefPubMed
  74. ↵
    1. Kawarabayashi T,
    2. Younkin LH,
    3. Saido TC,
    4. Shoji M,
    5. Ashe KH, and
    6. Younkin SG
    (2001) Age-dependent changes in brain, CSF, and plasma amyloid (beta) protein in the Tg2576 transgenic mouse model of Alzheimer’s disease. J Neurosci 21:372–381.
    OpenUrlAbstract/FREE Full Text
  75. ↵
    1. Keane C,
    2. Jerkic M, and
    3. Laffey JG
    (2017) Stem cell-based therapies for sepsis. Anesthesiology 127:1017–1034.
    OpenUrl
  76. ↵
    1. Kent SA,
    2. Spires-Jones TL, and
    3. Durrant CS
    (2020) The physiological roles of tau and Aβ: implications for Alzheimer’s disease pathology and therapeutics. Acta Neuropathol 140:417–447.
    OpenUrl
  77. ↵
    1. Kim DH,
    2. Lim H,
    3. Lee D,
    4. Choi SJ,
    5. Oh W,
    6. Yang YS,
    7. Oh JS,
    8. Hwang HH, and
    9. Jeon HB
    (2018) Thrombospondin-1 secreted by human umbilical cord blood-derived mesenchymal stem cells rescues neurons from synaptic dysfunction in Alzheimer’s disease model. Sci Rep 8:354.
    OpenUrl
  78. ↵
    1. Kim JA,
    2. Ha S,
    3. Shin KY,
    4. Kim S,
    5. Lee KJ,
    6. Chong YH,
    7. Chang KA, and
    8. Suh YH
    (2015) Neural stem cell transplantation at critical period improves learning and memory through restoring synaptic impairment in Alzheimer’s disease mouse model. Cell Death Dis 6:e1789.
    OpenUrl
  79. ↵
    1. Kim K,
    2. Lee CH, and
    3. Park CB
    (2020) Chemical sensing platforms for detecting trace-level Alzheimer’s core biomarkers. Chem Soc Rev 49:5446–5472.
    OpenUrl
  80. ↵
    1. Kim SU,
    2. Lee HJ, and
    3. Kim YB
    (2013) Neural stem cell-based treatment for neurodegenerative diseases. Neuropathology 33:491–504.
    OpenUrlCrossRefPubMed
  81. ↵
    1. King DL and
    2. Arendash GW
    (2002) Behavioral characterization of the Tg2576 transgenic model of Alzheimer’s disease through 19 months. Physiol Behav 75:627–642.
    OpenUrlCrossRefPubMed
  82. ↵
    1. Knappenberger KS,
    2. Tian G,
    3. Ye X,
    4. Sobotka-Briner C,
    5. Ghanekar SV,
    6. Greenberg BD, and
    7. Scott CW
    (2004) Mechanism of gamma-secretase cleavage activation: is gamma-secretase regulated through autoinhibition involving the presenilin-1 exon 9 loop? Biochemistry 43:6208–6218.
    OpenUrlCrossRefPubMed
  83. ↵
    1. Konttinen H,
    2. Gureviciene I,
    3. Oksanen M,
    4. Grubman A,
    5. Loppi S,
    6. Huuskonen MT,
    7. Korhonen P,
    8. Lampinen R,
    9. Keuters M,
    10. Belaya I, et al.
    (2019) PPARβ/δ-agonist GW0742 ameliorates dysfunction in fatty acid oxidation in PSEN1ΔE9 astrocytes. Glia 67:146–159.
    OpenUrl
  84. ↵
    1. Krencik R,
    2. Weick JP,
    3. Liu Y,
    4. Zhang ZJ, and
    5. Zhang SC
    (2011) Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nat Biotechnol 29:528–534.
    OpenUrlCrossRefPubMed
  85. ↵
    1. Kuijlaars J,
    2. Oyelami T,
    3. Diels A,
    4. Rohrbacher J,
    5. Versweyveld S,
    6. Meneghello G,
    7. Tuefferd M,
    8. Verstraelen P,
    9. Detrez JR,
    10. Verschuuren M, et al.
    (2016) Sustained synchronized neuronal network activity in a human astrocyte co-culture system. Sci Rep 6:36529.
    OpenUrlCrossRef
  86. ↵
    1. Kumar A,
    2. Singh A, and
    3. Ekavali
    (2015) A review on Alzheimer’s disease pathophysiology and its management: an update. Pharmacol Rep 67:195–203.
    OpenUrlCrossRefPubMed
  87. ↵
    1. Kwon EM,
    2. Connelly JP,
    3. Hansen NF,
    4. Donovan FX,
    5. Winkler T,
    6. Davis BW,
    7. Alkadi H,
    8. Chandrasekharappa SC,
    9. Dunbar CE,
    10. Mullikin JC, et al.
    (2017) iPSCs and fibroblast subclones from the same fibroblast population contain comparable levels of sequence variations. Proc Natl Acad Sci USA 114:1964–1969.
    OpenUrlAbstract/FREE Full Text
  88. ↵
    1. Lambert JC,
    2. Ibrahim-Verbaas CA,
    3. Harold D,
    4. Naj AC,
    5. Sims R,
    6. Bellenguez C,
    7. DeStafano AL,
    8. Bis JC,
    9. Beecham GW,
    10. Grenier-Boley B, et al., and European Alzheimer’s Disease Initiative (EADI); Genetic and Environmental Risk in Alzheimer’s Disease; Alzheimer’s Disease Genetic Consortium; Cohorts for Heart and Aging Research in Genomic Epidemiology
    (2013) Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet 45:1452–1458.
    OpenUrlCrossRefPubMed
  89. ↵
    1. Lee C,
    2. Willerth SM, and
    3. Nygaard HB
    (2020) The use of patient-derived induced pluripotent stem cells for Alzheimer’s disease modeling. Prog Neurobiol 192:101804.
    OpenUrl
  90. ↵
    1. Lee HJ,
    2. Lee JK,
    3. Lee H,
    4. Carter JE,
    5. Chang JW,
    6. Oh W,
    7. Yang YS,
    8. Suh JG,
    9. Lee BH,
    10. Jin HK, et al.
    (2012) Human umbilical cord blood-derived mesenchymal stem cells improve neuropathology and cognitive impairment in an Alzheimer’s disease mouse model through modulation of neuroinflammation. Neurobiol Aging 33:588–602.
    OpenUrlCrossRefPubMed
  91. ↵
    1. Lee JK,
    2. Jin HK, and
    3. Bae JS
    (2010) Bone marrow-derived mesenchymal stem cells attenuate amyloid β-induced memory impairment and apoptosis by inhibiting neuronal cell death. Curr Alzheimer Res 7:540–548.
    OpenUrlPubMed
  92. ↵
    1. Lemprière S
    (2019) Age-related microglial activation accelerated in AD. Nat Rev Neurol 15:369.
    OpenUrl
  93. ↵
    1. Leuzy A,
    2. Chiotis K,
    3. Hasselbalch SG,
    4. Rinne JO,
    5. de Mendonça A,
    6. Otto M,
    7. Lleó A,
    8. Castelo-Branco M,
    9. Santana I,
    10. Johansson J, et al.
    (2016) Pittsburgh compound B imaging and cerebrospinal fluid amyloid-β in a multicentre European memory clinic study. Brain 139:2540–2553.
    OpenUrlCrossRefPubMed
  94. ↵
    1. Li W,
    2. Li Y,
    3. Qiu Q,
    4. Sun L,
    5. Yue L,
    6. Li X, and
    7. Xiao S
    (2019) Associations between the apolipoprotein E ε4 allele and reduced serum levels of high density lipoprotein a cognitively normal aging han Chinese population. Front Endocrinol (Lausanne) 10:827.
    OpenUrl
  95. ↵
    1. Li X,
    2. Zhu H,
    3. Sun X,
    4. Zuo F,
    5. Lei J,
    6. Wang Z,
    7. Bao X, and
    8. Wang R
    (2016) Human neural stem cell transplantation rescues cognitive defects in APP/PS1 model of Alzheimer’s disease by enhancing neuronal connectivity and metabolic activity. Front Aging Neurosci 8:282.
    OpenUrl
  96. ↵
    1. Liddelow SA,
    2. Guttenplan KA,
    3. Clarke LE,
    4. Bennett FC,
    5. Bohlen CJ,
    6. Schirmer L,
    7. Bennett ML,
    8. Münch AE,
    9. Chung WS,
    10. Peterson TC, et al.
    (2017) Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541:481–487.
    OpenUrlCrossRefPubMed
  97. ↵
    1. Lilja AM,
    2. Malmsten L,
    3. Röjdner J,
    4. Voytenko L,
    5. Verkhratsky A,
    6. Ögren SO,
    7. Nordberg A, and
    8. Marutle A
    (2015) Neural stem cell transplant-induced effect on neurogenesis and cognition in Alzheimer Tg2576 mice is inhibited by concomitant treatment with amyloid-lowering or cholinergic α7 nicotinic receptor drugs. Neural Plast 2015:370432.
    OpenUrl
  98. ↵
    1. Lin SL
    (2011) Concise review: deciphering the mechanism behind induced pluripotent stem cell generation. Stem Cells 29:1645–1649.
    OpenUrlCrossRefPubMed
  99. ↵
    1. Lin YT,
    2. Seo J,
    3. Gao F,
    4. Feldman HM,
    5. Wen HL,
    6. Penney J,
    7. Cam HP,
    8. Gjoneska E,
    9. Raja WK,
    10. Cheng J, et al.
    (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.e7.
    OpenUrl
  100. ↵
    1. Liu F,
    2. Xuan A,
    3. Chen Y,
    4. Zhang J,
    5. Xu L,
    6. Yan Q, and
    7. Long D
    (2014) Combined effect of nerve growth factor and brain-derived neurotrophic factor on neuronal differentiation of neural stem cells and the potential molecular mechanisms. Mol Med Rep 10:1739–1745.
    OpenUrl
  101. ↵
    1. Liu Y,
    2. Weick JP,
    3. Liu H,
    4. Krencik R,
    5. Zhang X,
    6. Ma L,
    7. Zhou GM,
    8. Ayala M, and
    9. Zhang SC
    (2013) Medial ganglionic eminence-like cells derived from human embryonic stem cells correct learning and memory deficits. Nat Biotechnol 31:440–447.
    OpenUrlCrossRefPubMed
  102. ↵
    1. Madhavan M,
    2. Nevin ZS,
    3. Shick HE,
    4. Garrison E,
    5. Clarkson-Paredes C,
    6. Karl M,
    7. Clayton BLL,
    8. Factor DC,
    9. Allan KC,
    10. Barbar L, et al.
    (2018) Induction of myelinating oligodendrocytes in human cortical spheroids. Nat Methods 15:700–706.
    OpenUrlCrossRefPubMed
  103. ↵
    1. Maherali N,
    2. Ahfeldt T,
    3. Rigamonti A,
    4. Utikal J,
    5. Cowan C, and
    6. Hochedlinger K
    (2008) A high-efficiency system for the generation and study of human induced pluripotent stem cells. Cell Stem Cell 3:340–345.
    OpenUrlCrossRefPubMed
  104. ↵
    1. Maia LF,
    2. Kaeser SA,
    3. Reichwald J,
    4. Hruscha M,
    5. Martus P,
    6. Staufenbiel M, and
    7. Jucker M
    (2013) Changes in amyloid-β and Tau in the cerebrospinal fluid of transgenic mice overexpressing amyloid precursor protein. Sci Transl Med 5:194re2.
    OpenUrlFREE Full Text
  105. ↵
    1. Maloney JA,
    2. Bainbridge T,
    3. Gustafson A,
    4. Zhang S,
    5. Kyauk R,
    6. Steiner P,
    7. van der Brug M,
    8. Liu Y,
    9. Ernst JA,
    10. Watts RJ, et al.
    (2014) Molecular mechanisms of Alzheimer disease protection by the A673T allele of amyloid precursor protein. J Biol Chem 289:30990–31000.
    OpenUrlAbstract/FREE Full Text
  106. ↵
    1. Mancuso R,
    2. Van Den Daele J,
    3. Fattorelli N,
    4. Wolfs L,
    5. Balusu S,
    6. Burton O,
    7. Liston A,
    8. Sierksma A,
    9. Fourne Y,
    10. Poovathingal S, et al.
    (2019) Stem-cell-derived human microglia transplanted in mouse brain to study human disease. Nat Neurosci 22:2111–2116.
    OpenUrlCrossRefPubMed
  107. ↵
    1. Marion RM,
    2. Strati K,
    3. Li H,
    4. Tejera A,
    5. Schoeftner S,
    6. Ortega S,
    7. Serrano M, and
    8. Blasco MA
    (2009) Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells. Cell Stem Cell 4:141–154.
    OpenUrlCrossRefPubMed
  108. ↵
    1. Maroof AM,
    2. Keros S,
    3. Tyson JA,
    4. Ying SW,
    5. Ganat YM,
    6. Merkle FT,
    7. Liu B,
    8. Goulburn A,
    9. Stanley EG,
    10. Elefanty AG, et al.
    (2013) Directed differentiation and functional maturation of cortical interneurons from human embryonic stem cells. Cell Stem Cell 12:559–572.
    OpenUrlCrossRefPubMed
  109. ↵
    1. Marsh SE and
    2. Blurton-Jones M
    (2017) Neural stem cell therapy for neurodegenerative disorders: the role of neurotrophic support. Neurochem Int 106:94–100.
    OpenUrl
  110. ↵
    1. Marsh SE,
    2. Yeung ST,
    3. Torres M,
    4. Lau L,
    5. Davis JL,
    6. Monuki ES,
    7. Poon WW, and
    8. Blurton-Jones M
    (2017) HuCNS-SC human NSCs fail to differentiate, form ectopic clusters, and provide no cognitive benefits in a transgenic model of Alzheimer’s disease. Stem Cell Reports 8:235–248.
    OpenUrl
  111. ↵
    1. Martínez-Morales PL,
    2. Revilla A,
    3. Ocaña I,
    4. González C,
    5. Sainz P,
    6. McGuire D, and
    7. Liste I
    (2013) Progress in stem cell therapy for major human neurological disorders. Stem Cell Rev Rep 9:685–699.
    OpenUrl
  112. ↵
    1. Massirer KB,
    2. Carromeu C,
    3. Griesi-Oliveira K, and
    4. Muotri AR
    (2011) Maintenance and differentiation of neural stem cells. Wiley Interdiscip Rev Syst Biol Med 3:107–114.
    OpenUrlCrossRefPubMed
  113. ↵
    1. McGinley LM,
    2. Kashlan ON,
    3. Bruno ES,
    4. Chen KS,
    5. Hayes JM,
    6. Kashlan SR,
    7. Raykin J,
    8. Johe K,
    9. Murphy GG, and
    10. Feldman EL
    (2018) Human neural stem cell transplantation improves cognition in a murine model of Alzheimer’s disease. Sci Rep 8:14776.
    OpenUrl
  114. ↵
    1. Meng X,
    2. Neises A,
    3. Su RJ,
    4. Payne KJ,
    5. Ritter L,
    6. Gridley DS,
    7. Wang J,
    8. Sheng M,
    9. Lau KH,
    10. Baylink DJ, et al.
    (2012) Efficient reprogramming of human cord blood CD34+ cells into induced pluripotent stem cells with OCT4 and SOX2 alone. Mol Ther 20:408–416.
    OpenUrlCrossRefPubMed
  115. ↵
    1. Mentis AA,
    2. Dardiotis E, and
    3. Chrousos GP
    (2020) Apolipoprotein E4 and meningeal lymphatics in Alzheimer disease: a conceptual framework. Mol Psychiatry DOI: 10.1038/s41380-020-0731-7 [published ahead of print].
  116. ↵
    1. Mertens J,
    2. Reid D,
    3. Lau S,
    4. Kim Y, and
    5. Gage FH
    (2018) Aging in a dish: iPSC-derived and directly induced neurons for studying brain aging and age-related neurodegenerative diseases. Annu Rev Genet 52:271–293.
    OpenUrlCrossRef
  117. ↵
    1. Mills JA,
    2. Wang K,
    3. Paluru P,
    4. Ying L,
    5. Lu L,
    6. Galvão AM,
    7. Xu D,
    8. Yao Y,
    9. Sullivan SK,
    10. Sullivan LM, et al.
    (2013) Clonal genetic and hematopoietic heterogeneity among human-induced pluripotent stem cell lines. Blood 122:2047–2051.
    OpenUrlAbstract/FREE Full Text
  118. ↵
    1. Molofsky AV and
    2. Deneen B
    (2015) Astrocyte development: a guide for the perplexed. Glia 63:1320–1329.
    OpenUrlCrossRefPubMed
  119. ↵
    1. Montserrat N,
    2. Ramírez-Bajo MJ,
    3. Xia Y,
    4. Sancho-Martinez I,
    5. Moya-Rull D,
    6. Miquel-Serra L,
    7. Yang S,
    8. Nivet E,
    9. Cortina C,
    10. González F, et al.
    (2012) Generation of induced pluripotent stem cells from human renal proximal tubular cells with only two transcription factors, OCT4 and SOX2. J Biol Chem 287:24131–24138.
    OpenUrlAbstract/FREE Full Text
  120. ↵
    1. Moon JH,
    2. Heo JS,
    3. Kwon S,
    4. Kim J,
    5. Hwang J,
    6. Kang PJ,
    7. Kim A,
    8. Kim HO,
    9. Whang KY,
    10. Yoon BS, et al.
    (2012) Two-step generation of induced pluripotent stem cells from mouse fibroblasts using Id3 and Oct4. J Mol Cell Biol 4:59–62.
    OpenUrlCrossRefPubMed
  121. ↵
    1. Muffat J,
    2. Li Y,
    3. Yuan B,
    4. Mitalipova M,
    5. Omer A,
    6. Corcoran S,
    7. Bakiasi G,
    8. Tsai LH,
    9. Aubourg P,
    10. Ransohoff RM, et al.
    (2016) Efficient derivation of microglia-like cells from human pluripotent stem cells. Nat Med 22:1358–1367.
    OpenUrlCrossRefPubMed
  122. ↵
    1. Muratore CR,
    2. Rice HC,
    3. Srikanth P,
    4. Callahan DG,
    5. Shin T,
    6. Benjamin LN,
    7. Walsh DM,
    8. Selkoe DJ, and
    9. 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.
    OpenUrlCrossRefPubMed
  123. ↵
    1. Naaldijk Y,
    2. Jäger C,
    3. Fabian C,
    4. Leovsky C,
    5. Blüher A,
    6. Rudolph L,
    7. Hinze A, and
    8. Stolzing A
    (2017) Effect of systemic transplantation of bone marrow-derived mesenchymal stem cells on neuropathology markers in APP/PS1 Alzheimer mice. Neuropathol Appl Neurobiol 43:299–314.
    OpenUrlCrossRefPubMed
  124. ↵
    1. Naji A,
    2. Eitoku M,
    3. Favier B,
    4. Deschaseaux F,
    5. Rouas-Freiss N, and
    6. Suganuma N
    (2019) Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci 76:3323–3348.
    OpenUrl
  125. ↵
    1. Nakagawa M,
    2. Koyanagi M,
    3. Tanabe K,
    4. Takahashi K,
    5. Ichisaka T,
    6. Aoi T,
    7. Okita K,
    8. Mochiduki Y,
    9. Takizawa N, and
    10. Yamanaka S
    (2008) Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol 26:101–106.
    OpenUrlCrossRefPubMed
  126. ↵
    1. Nicholas CR,
    2. Chen J,
    3. Tang Y,
    4. Southwell DG,
    5. Chalmers N,
    6. Vogt D,
    7. Arnold CM,
    8. Chen YJ,
    9. Stanley EG,
    10. Elefanty AG, et al.
    (2013) Functional maturation of hPSC-derived forebrain interneurons requires an extended timeline and mimics human neural development. Cell Stem Cell 12:573–586.
    OpenUrlCrossRefPubMed
  127. ↵
    1. Oakley H,
    2. Cole SL,
    3. Logan S,
    4. Maus E,
    5. Shao P,
    6. Craft J,
    7. Guillozet-Bongaarts A,
    8. Ohno M,
    9. Disterhoft J,
    10. Van Eldik L, et al.
    (2006) Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J Neurosci 26:10129–10140.
    OpenUrlAbstract/FREE Full Text
  128. ↵
    1. Ochalek A,
    2. Mihalik B,
    3. Avci HX,
    4. Chandrasekaran A,
    5. Téglási A,
    6. Bock I,
    7. Giudice ML,
    8. Táncos Z,
    9. Molnár K,
    10. László L, et al.
    (2017) Neurons derived from sporadic Alzheimer’s disease iPSCs reveal elevated TAU hyperphosphorylation, increased amyloid levels, and GSK3B activation. Alzheimers Res Ther 9:90.
    OpenUrlCrossRefPubMed
  129. ↵
    1. Oddo S,
    2. Caccamo A,
    3. Shepherd JD,
    4. Murphy MP,
    5. Golde TE,
    6. Kayed R,
    7. Metherate R,
    8. Mattson MP,
    9. Akbari Y, and
    10. LaFerla FM
    (2003) Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron 39:409–421.
    OpenUrlCrossRefPubMed
  130. ↵
    1. Oh SH,
    2. Kim HN,
    3. Park HJ,
    4. Shin JY, and
    5. Lee PH
    (2015) Mesenchymal stem cells increase hippocampal neurogenesis and neuronal differentiation by enhancing the wnt signaling pathway in an Alzheimer’s disease model. Cell Transplant 24:1097–1109.
    OpenUrl
  131. ↵
    1. Oksanen M,
    2. Petersen AJ,
    3. Naumenko N,
    4. Puttonen K,
    5. Lehtonen Š,
    6. Gubert Olivé M,
    7. Shakirzyanova A,
    8. Leskelä S,
    9. Sarajärvi T,
    10. Viitanen M, et al.
    (2017) PSEN1 mutant iPSC-derived model reveals severe astrocyte pathology in Alzheimer’s disease. Stem Cell Reports 9:1885–1897.
    OpenUrl
  132. ↵
    1. Olsson B,
    2. Lautner R,
    3. Andreasson U,
    4. Öhrfelt A,
    5. Portelius E,
    6. Bjerke M,
    7. Hölttä M,
    8. Rosén C,
    9. Olsson C,
    10. Strobel G, et al.
    (2016) CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol 15:673–684.
    OpenUrlCrossRefPubMed
  133. ↵
    1. Omole AE and
    2. Fakoya AOJ
    (2018) Ten years of progress and promise of induced pluripotent stem cells: historical origins, characteristics, mechanisms, limitations, and potential applications. PeerJ 6:e4370.
    OpenUrlCrossRef
  134. ↵
    1. Ong SS,
    2. Doraiswamy PM, and
    3. Lad EM
    (2018) Controversies and future directions of ocular biomarkers in Alzheimer disease. JAMA Neurol 75:650–651.
    OpenUrl
  135. ↵
    1. Ortiz-Virumbrales M,
    2. Moreno CL,
    3. Kruglikov I,
    4. Marazuela P,
    5. Sproul A,
    6. Jacob S,
    7. Zimmer M,
    8. Paull D,
    9. Zhang B,
    10. 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.
    OpenUrl
  136. ↵
    1. Ovchinnikov DA,
    2. Korn O,
    3. Virshup I,
    4. Wells CA, and
    5. 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.
    OpenUrl
  137. ↵
    1. Pascoal TA,
    2. Mathotaarachchi S,
    3. Mohades S,
    4. Benedet AL,
    5. Chung CO,
    6. Shin M,
    7. Wang S,
    8. Beaudry T,
    9. Kang MS,
    10. Soucy JP, et al.
    (2017) Amyloid-β and hyperphosphorylated tau synergy drives metabolic decline in preclinical Alzheimer’s disease. Mol Psychiatry 22:306–311.
    OpenUrlCrossRef
  138. ↵
    1. Penney J,
    2. Ralvenius WT, and
    3. Tsai LH
    (2020) Modeling Alzheimer’s disease with iPSC-derived brain cells. Mol Psychiatry 25:148–167.
    OpenUrlPubMed
  139. ↵
    1. Philips T and
    2. Robberecht W
    (2011) Neuroinflammation in amyotrophic lateral sclerosis: role of glial activation in motor neuron disease. Lancet Neurol 10:253–263.
    OpenUrlCrossRefPubMed
  140. ↵
    1. Pierro M,
    2. Thébaud B, and
    3. Soll R
    (2017) Mesenchymal stem cells for the prevention and treatment of bronchopulmonary dysplasia in preterm infants. Cochrane Database Syst Rev 11:CD011932.
    OpenUrl
  141. ↵
    1. Pournasr B and
    2. Duncan SA
    (2017) Modeling inborn errors of hepatic metabolism using induced pluripotent stem cells. Arterioscler Thromb Vasc Biol 37:1994–1999.
    OpenUrlAbstract/FREE Full Text
  142. ↵
    1. Rabinovici GD
    (2016) Amyloid biomarkers: pushing the limits of early detection. Brain 139:1008–1010.
    OpenUrlCrossRefPubMed
  143. ↵
    1. Ransohoff RM
    (2016) How neuroinflammation contributes to neurodegeneration. Science 353:777–783.
    OpenUrlAbstract/FREE Full Text
  144. ↵
    1. Rebuzzini P,
    2. Zuccotti M,
    3. Redi CA, and
    4. Garagna S
    (2016) Achilles’ heel of pluripotent stem cells: genetic, genomic and epigenetic variations during prolonged culture. Cell Mol Life Sci 73:2453–2466.
    OpenUrl
  145. ↵
    1. Richards RI,
    2. Robertson SA,
    3. O’Keefe LV,
    4. Fornarino D,
    5. Scott A,
    6. Lardelli M, and
    7. Baune BT
    (2016) The enemy within: innate surveillance-mediated cell death, the common mechanism of neurodegenerative disease. Front Neurosci 10:193.
    OpenUrl
  146. ↵
    1. Roberson ED and
    2. Mucke L
    (2006) 100 years and counting: prospects for defeating Alzheimer’s disease. Science 314:781–784.
    OpenUrlAbstract/FREE Full Text
  147. ↵
    1. Robert AW,
    2. Marcon BH,
    3. Dallagiovanna B, and
    4. Shigunov P
    (2020) Adipogenesis, osteogenesis, and chondrogenesis of human mesenchymal stem/stromal cells: a comparative transcriptome approach. Front Cell Dev Biol 8:561.
    OpenUrl
  148. ↵
    1. Roessler R,
    2. Smallwood SA,
    3. Veenvliet JV,
    4. Pechlivanoglou P,
    5. Peng SP,
    6. Chakrabarty K,
    7. Groot-Koerkamp MJ,
    8. Pasterkamp RJ,
    9. Wesseling E,
    10. Kelsey G, et al.
    (2014) Detailed analysis of the genetic and epigenetic signatures of iPSC-derived mesodiencephalic dopaminergic neurons. Stem Cell Reports 2:520–533.
    OpenUrl
  149. ↵
    1. Sá da Bandeira D,
    2. Casamitjana J, and
    3. Crisan M
    (2017) Pericytes, integral components of adult hematopoietic stem cell niches. Pharmacol Ther 171:104–113.
    OpenUrlCrossRef
  150. ↵
    1. Sabatino JJ Jr.,
    2. Pröbstel AK, and
    3. Zamvil SS
    (2019) B cells in autoimmune and neurodegenerative central nervous system diseases. Nat Rev Neurosci 20:728–745.
    OpenUrl
  151. ↵
    1. Sabayan B and
    2. Sorond F
    (2017) Reducing risk of dementia in older age. JAMA 317:2028.
    OpenUrl
  152. ↵
    1. Salter MW and
    2. Beggs S
    (2014) Sublime microglia: expanding roles for the guardians of the CNS. Cell 158:15–24.
    OpenUrlCrossRefPubMed
  153. ↵
    1. Scearce-Levie K,
    2. Sanchez PE, and
    3. Lewcock JW
    (2020) Leveraging preclinical models for the development of Alzheimer disease therapeutics. Nat Rev Drug Discov 19:447–462.
    OpenUrlCrossRef
  154. ↵
    1. Scheltens P,
    2. Blennow K,
    3. Breteler MM,
    4. de Strooper B,
    5. Frisoni GB,
    6. Salloway S, and
    7. Van der Flier WM
    (2016) Alzheimer’s disease. Lancet 388:505–517.
    OpenUrlCrossRefPubMed
  155. ↵
    1. Serretti A,
    2. Olgiati P, and
    3. De Ronchi D
    (2007) Genetics of Alzheimer’s disease. A rapidly evolving field. J Alzheimers Dis 12:73–92.
    OpenUrlPubMed
  156. ↵
    1. Shahbazi E,
    2. Mirakhori F,
    3. Ezzatizadeh V, and
    4. Baharvand H
    (2018) Reprogramming of somatic cells to induced neural stem cells. Methods 133:21–28.
    OpenUrl
  157. ↵
    1. Shi Y,
    2. Kirwan P,
    3. Smith J,
    4. MacLean G,
    5. Orkin SH, and
    6. Livesey FJ
    (2012) A human stem cell model of early Alzheimer’s disease pathology in Down syndrome. Sci Transl Med 4:124ra29.
    OpenUrlAbstract/FREE Full Text
  158. ↵
    1. Si Z,
    2. Wang X,
    3. Kang Y,
    4. Wang X,
    5. Sun C,
    6. Li Y,
    7. Xu J,
    8. Wu J,
    9. Zhang Z,
    10. Li L, et al.
    (2020) Heme oxygenase 1 inhibits adult neural stem cells proliferation and survival via modulation of Wnt/β-catenin signaling. J Alzheimers Dis 76:623–641.
    OpenUrl
  159. ↵
    1. Si Z,
    2. Wang X,
    3. Sun C,
    4. Kang Y,
    5. Xu J,
    6. Wang X, and
    7. Hui Y
    (2019) Adipose-derived stem cells: sources, potency, and implications for regenerative therapies. Biomed Pharmacother 114:108765.
    OpenUrl
  160. ↵
    1. Si Z,
    2. Wang X,
    3. Zhang Z,
    4. Wang J,
    5. Li J,
    6. Li J,
    7. Li L,
    8. Li Y,
    9. Peng Y,
    10. Sun C, et al.
    (2018) Heme oxygenase 1 induces tau oligomer formation and synapse aberrations in hippocampal neurons. J Alzheimers Dis 65:409–419.
    OpenUrl
  161. ↵
    1. Simons M and
    2. Nave KA
    (2015) Oligodendrocytes: myelination and axonal support. Cold Spring Harb Perspect Biol 8:a020479.
    OpenUrlCrossRefPubMed
  162. ↵
    1. Sloan SA,
    2. Darmanis S,
    3. Huber N,
    4. Khan TA,
    5. Birey F,
    6. Caneda C,
    7. Reimer R,
    8. Quake SR,
    9. Barres BA, and
    10. Paşca SP
    (2017) Human astrocyte maturation captured in 3D cerebral cortical spheroids derived from pluripotent stem cells. Neuron 95:779–790.e6.
    OpenUrlCrossRefPubMed
  163. ↵
    1. Soldner F,
    2. Hockemeyer D,
    3. Beard C,
    4. Gao Q,
    5. Bell GW,
    6. Cook EG,
    7. Hargus G,
    8. Blak A,
    9. Cooper O,
    10. Mitalipova M, et al.
    (2009) Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell 136:964–977.
    OpenUrlCrossRefPubMed
  164. ↵
    1. Song N,
    2. Scholtemeijer M, and
    3. Shah K
    (2020) Mesenchymal stem cell immunomodulation: mechanisms and therapeutic potential. Trends Pharmacol Sci 41:653–664.
    OpenUrlPubMed
  165. ↵
    1. Sproul AA
    (2015) Being human: the role of pluripotent stem cells in regenerative medicine and humanizing Alzheimer’s disease models. Mol Aspects Med 43–44:54–65.
    OpenUrl
  166. ↵
    1. Stadtfeld M,
    2. Brennand K, and
    3. Hochedlinger K
    (2008) Reprogramming of pancreatic beta cells into induced pluripotent stem cells. Curr Biol 18:890–894.
    OpenUrlCrossRefPubMed
  167. ↵
    1. Stakos DA,
    2. Stamatelopoulos K,
    3. Bampatsias D,
    4. Sachse M,
    5. Zormpas E,
    6. Vlachogiannis NI,
    7. Tual-Chalot S, and
    8. Stellos K
    (2020) The Alzheimer’s disease amyloid-beta hypothesis in cardiovascular aging and disease: JACC focus seminar. J Am Coll Cardiol 75:952–967.
    OpenUrlFREE Full Text
  168. ↵
    1. Sullivan S,
    2. Stacey GN,
    3. Akazawa C,
    4. Aoyama N,
    5. Baptista R,
    6. Bedford P,
    7. Bennaceur Griscelli A,
    8. Chandra A,
    9. Elwood N,
    10. Girard M, et al.
    (2018) Quality control guidelines for clinical-grade human induced pluripotent stem cell lines. Regen Med 13:859–866.
    OpenUrl
  169. ↵
    1. Sun AX,
    2. Yuan Q,
    3. Tan S,
    4. Xiao Y,
    5. Wang D,
    6. Khoo AT,
    7. Sani L,
    8. Tran HD,
    9. Kim P,
    10. Chiew YS, et al.
    (2016) Direct induction and functional maturation of forebrain GABAergic neurons from human pluripotent stem cells. Cell Rep 16:1942–1953.
    OpenUrlCrossRefPubMed
  170. ↵
    1. Takahashi K,
    2. Tanabe K,
    3. Ohnuki M,
    4. Narita M,
    5. Ichisaka T,
    6. Tomoda K, and
    7. Yamanaka S
    (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131:861–872.
    OpenUrlCrossRefPubMed
  171. ↵
    1. Takahashi K and
    2. Yamanaka S
    (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663–676.
    OpenUrlCrossRefPubMed
  172. ↵
    1. Tang J,
    2. Xu H,
    3. Fan X,
    4. Li D,
    5. Rancourt D,
    6. Zhou G,
    7. Li Z, and
    8. Yang L
    (2008) Embryonic stem cell-derived neural precursor cells improve memory dysfunction in Abeta(1-40) injured rats. Neurosci Res 62:86–96.
    OpenUrlPubMed
  173. ↵
    1. Tarawneh R,
    2. D’Angelo G,
    3. Crimmins D,
    4. Herries E,
    5. Griest T,
    6. Fagan AM,
    7. Zipfel GJ,
    8. Ladenson JH,
    9. Morris JC, and
    10. Holtzman DM
    (2016) Diagnostic and prognostic utility of the synaptic marker neurogranin in Alzheimer disease. JAMA Neurol 73:561–571.
    OpenUrl
  174. ↵
    1. Teipel S,
    2. König A,
    3. Hoey J,
    4. Kaye J,
    5. Krüger F,
    6. Robillard JM,
    7. Kirste T, and
    8. Babiloni C
    (2018) Use of nonintrusive sensor-based information and communication technology for real-world evidence for clinical trials in dementia. Alzheimers Dement 14:1216–1231.
    OpenUrl
  175. ↵
    1. Teixeira FG,
    2. Carvalho MM,
    3. Panchalingam KM,
    4. Rodrigues AJ,
    5. Mendes-Pinheiro B,
    6. Anjo S,
    7. Manadas B,
    8. Behie LA,
    9. Sousa N, and
    10. Salgado AJ
    (2017) Impact of the secretome of human mesenchymal stem cells on brain structure and animal behavior in a rat model of Parkinson’s disease. Stem Cells Transl Med 6:634–646.
    OpenUrl
  176. ↵
    1. Telias M and
    2. Ben-Yosef D
    (2015) Neural stem cell replacement: a possible therapy for neurodevelopmental disorders? Neural Regen Res 10:180–182.
    OpenUrl
  177. ↵
    1. Thirabanjasak D,
    2. Tantiwongse K, and
    3. Thorner PS
    (2010) Angiomyeloproliferative lesions following autologous stem cell therapy. J Am Soc Nephrol 21:1218–1222.
    OpenUrlAbstract/FREE Full Text
  178. ↵
    1. Tsai SY,
    2. Clavel C,
    3. Kim S,
    4. Ang YS,
    5. Grisanti L,
    6. Lee DF,
    7. Kelley K, and
    8. Rendl M
    (2010) Oct4 and klf4 reprogram dermal papilla cells into induced pluripotent stem cells. Stem Cells 28:221–228.
    OpenUrlPubMed
  179. ↵
    1. Varderidou-Minasian S and
    2. Lorenowicz MJ
    (2020) Mesenchymal stromal/stem cell-derived extracellular vesicles in tissue repair: challenges and opportunities. Theranostics 10:5979–5997.
    OpenUrl
  180. ↵
    1. Victor MB,
    2. Richner M,
    3. Olsen HE,
    4. Lee SW,
    5. Monteys AM,
    6. Ma C,
    7. Huh CJ,
    8. Zhang B,
    9. Davidson BL,
    10. Yang XW, et al.
    (2018) Striatal neurons directly converted from Huntington’s disease patient fibroblasts recapitulate age-associated disease phenotypes. Nat Neurosci 21:341–352.
    OpenUrl
  181. ↵
    1. Volpato V,
    2. Smith J,
    3. Sandor C,
    4. Ried JS,
    5. Baud A,
    6. Handel A,
    7. Newey SE,
    8. Wessely F,
    9. Attar M,
    10. Whiteley E, et al.
    (2018) Reproducibility of molecular phenotypes after long-term differentiation to human iPSC-derived neurons: a multi-site omics study. Stem Cell Reports 11:897–911.
    OpenUrl
  182. ↵
    1. Wang C,
    2. Najm R,
    3. Xu Q,
    4. Jeong DE,
    5. Walker D,
    6. Balestra ME,
    7. Yoon SY,
    8. Yuan H,
    9. Li G,
    10. 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.
    OpenUrlCrossRefPubMed
  183. ↵
    1. Wang S,
    2. Bates J,
    3. Li X,
    4. Schanz S,
    5. Chandler-Militello D,
    6. Levine C,
    7. Maherali N,
    8. Studer L,
    9. Hochedlinger K,
    10. Windrem M, et al.
    (2013) Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 12:252–264.
    OpenUrlCrossRefPubMed
  184. ↵
    1. Watanabe S,
    2. Hirai H,
    3. Asakura Y,
    4. Tastad C,
    5. Verma M,
    6. Keller C,
    7. Dutton JR, and
    8. Asakura A
    (2011) MyoD gene suppression by Oct4 is required for reprogramming in myoblasts to produce induced pluripotent stem cells. Stem Cells 29:505–516.
    OpenUrlCrossRefPubMed
  185. ↵
    1. Weber B and
    2. Barros LF
    (2015) The astrocyte: powerhouse and recycling center. Cold Spring Harb Perspect Biol 7:a020396.
    OpenUrlAbstract/FREE Full Text
  186. ↵
    1. Wen Y and
    2. Jin S
    (2014) Production of neural stem cells from human pluripotent stem cells. J Biotechnol 188:122–129.
    OpenUrl
  187. ↵
    1. Wezyk M,
    2. Szybinska A,
    3. Wojsiat J,
    4. Szczerba M,
    5. Day K,
    6. Ronnholm H,
    7. Kele M,
    8. Berdynski M,
    9. Peplonska B,
    10. 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.
    OpenUrl
  188. ↵
    1. Wu CC,
    2. Lien CC,
    3. Hou WH,
    4. Chiang PM, and
    5. Tsai KJ
    (2016) Gain of BDNF function in engrafted neural stem cells promotes the therapeutic potential for Alzheimer’s disease. Sci Rep 6:27358.
    OpenUrl
  189. ↵
    1. Xu M,
    2. Zhang L,
    3. Liu G,
    4. Jiang N,
    5. Zhou W, and
    6. Zhang Y
    (2019) Pathological changes in Alzheimer’s disease analyzed using induced pluripotent stem cell-derived human microglia-like cells. J Alzheimers Dis 67:357–368.
    OpenUrl
  190. ↵
    1. Yang H,
    2. Xie Z,
    3. Wei L,
    4. Yang H,
    5. Yang S,
    6. Zhu Z,
    7. Wang P,
    8. Zhao C, and
    9. Bi J
    (2013) Human umbilical cord mesenchymal stem cell-derived neuron-like cells rescue memory deficits and reduce amyloid-beta deposition in an AβPP/PS1 transgenic mouse model. Stem Cell Res Ther 4:76.
    OpenUrlCrossRefPubMed
  191. ↵
    1. Yang L,
    2. Zhai Y,
    3. Hao Y,
    4. Zhu Z, and
    5. Cheng G
    (2020) The regulatory functionality of exosomes derived from hUMSCs in 3D culture for Alzheimer’s disease therapy. Small 16:e1906273.
    OpenUrl
  192. ↵
    1. Yoshihara M,
    2. Hayashizaki Y, and
    3. Murakawa Y
    (2017) Genomic instability of iPSCs: challenges towards their clinical applications. Stem Cell Rev Rep 13:7–16.
    OpenUrl
  193. ↵
    1. Zetterberg H and
    2. Bendlin BB
    (2021) Biomarkers for Alzheimer’s disease-preparing for a new era of disease-modifying therapies. Mol Psychiatry 26:296–308.
    OpenUrl
  194. ↵
    1. Zhang M,
    2. Wang L,
    3. An K,
    4. Cai J,
    5. Li G,
    6. Yang C,
    7. Liu H,
    8. Du F,
    9. Han X,
    10. Zhang Z, et al.
    (2018) Lower genomic stability of induced pluripotent stem cells reflects increased non-homologous end joining. Cancer Commun (Lond) 38:49.
    OpenUrl
  195. ↵
    1. Zhang Q,
    2. Wu HH,
    3. Wang Y,
    4. Gu GJ,
    5. Zhang W, and
    6. Xia R
    (2016) Neural stem cell transplantation decreases neuroinflammation in a transgenic mouse model of Alzheimer’s disease. J Neurochem 136:815–825.
    OpenUrl
  196. ↵
    1. Zhang T,
    2. Ke W,
    3. Zhou X,
    4. Qian Y,
    5. Feng S,
    6. Wang R,
    7. Cui G,
    8. Tao R,
    9. Guo W,
    10. Duan Y, et al.
    (2019) Human neural stem cells reinforce hippocampal synaptic network and rescue cognitive deficits in a mouse model of Alzheimer’s disease. Stem Cell Reports 13:1022–1037.
    OpenUrl
  197. ↵
    1. Zhang W,
    2. Gu GJ,
    3. Shen X,
    4. Zhang Q,
    5. Wang GM, and
    6. Wang PJ
    (2015) Neural stem cell transplantation enhances mitochondrial biogenesis in a transgenic mouse model of Alzheimer’s disease-like pathology. Neurobiol Aging 36:1282–1292.
    OpenUrl
  198. ↵
    1. Zhang W,
    2. Wang PJ,
    3. Sha HY,
    4. Ni J,
    5. Li MH, and
    6. Gu GJ
    (2014) Neural stem cell transplants improve cognitive function without altering amyloid pathology in an APP/PS1 double transgenic model of Alzheimer’s disease. Mol Neurobiol 50:423–437.
    OpenUrl
  199. ↵
    1. Zhang Y,
    2. Pak C,
    3. Han Y,
    4. Ahlenius H,
    5. Zhang Z,
    6. Chanda S,
    7. Marro S,
    8. Patzke C,
    9. Acuna C,
    10. Covy J, et al.
    (2013) Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78:785–798.
    OpenUrlCrossRefPubMed
  200. ↵
    1. Zhang Z,
    2. Jiang Q,
    3. Jiang F,
    4. Ding G,
    5. Zhang R,
    6. Wang L,
    7. Zhang L,
    8. Robin AM,
    9. Katakowski M, and
    10. Chopp M
    (2004) In vivo magnetic resonance imaging tracks adult neural progenitor cell targeting of brain tumor. Neuroimage 23:281–287.
    OpenUrlCrossRefPubMed
  201. ↵
    1. Zhao J,
    2. Davis MD,
    3. Martens YA,
    4. Shinohara M,
    5. Graff-Radford NR,
    6. Younkin SG,
    7. Wszolek ZK,
    8. Kanekiyo T, and
    9. Bu G
    (2017) APOE ε4/ε4 diminishes neurotrophic function of human iPSC-derived astrocytes. Hum Mol Genet 26:2690–2700.
    OpenUrlCrossRefPubMed
  202. ↵
    1. Zhao W,
    2. Beers DR, and
    3. Appel SH
    (2013) Immune-mediated mechanisms in the pathoprogression of amyotrophic lateral sclerosis. J Neuroimmune Pharmacol 8:888–899.
    OpenUrlCrossRefPubMed
  203. ↵
    1. Zheng Y,
    2. Huang J,
    3. Zhu T,
    4. Li R,
    5. Wang Z,
    6. Ma F, and
    7. Zhu J
    (2017) Stem cell tracking technologies for neurological regenerative medicine purposes. Stem Cells Int 2017:2934149.
    OpenUrl
  204. ↵
    1. Zhou T,
    2. Benda C,
    3. Duzinger S,
    4. Huang Y,
    5. Li X,
    6. Li Y,
    7. Guo X,
    8. Cao G,
    9. Chen S,
    10. Hao L, et al.
    (2011) Generation of induced pluripotent stem cells from urine. J Am Soc Nephrol 22:1221–1228.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

Journal of Pharmacology and Experimental Therapeutics: 377 (2)
Journal of Pharmacology and Experimental Therapeutics
Vol. 377, Issue 2
1 May 2021
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Editorial Board (PDF)
  • Front Matter (PDF)
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Journal of Pharmacology and Experimental Therapeutics article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Stem Cell Therapies in Alzheimer’s Disease: Applications for Disease Modeling
(Your Name) has forwarded a page to you from Journal of Pharmacology and Experimental Therapeutics
(Your Name) thought you would be interested in this article in Journal of Pharmacology and Experimental Therapeutics.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Research ArticleMinireview

Stem Cells Therapies in AD

Zizhen Si and Xidi Wang
Journal of Pharmacology and Experimental Therapeutics May 1, 2021, 377 (2) 207-217; DOI: https://doi.org/10.1124/jpet.120.000324

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Research ArticleMinireview

Stem Cells Therapies in AD

Zizhen Si and Xidi Wang
Journal of Pharmacology and Experimental Therapeutics May 1, 2021, 377 (2) 207-217; DOI: https://doi.org/10.1124/jpet.120.000324
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • The Pathogenesis of AD
    • Stem Cell Treatment in AD Modeling
    • Applications of iPSCs in AD Modeling
    • Applications of NSCs in AD Modeling
    • Application of MSCs in AD Modeling
    • Challenges and Future Perspectives
    • Authorship Contributions
    • Footnotes
    • Abbreviations
    • References
  • Figures & Data
  • Info & Metrics
  • eLetters
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Traumatic Brain Injury Pharmacology Trials
  • Generic Substitution of Antiseizure Medications
  • Potential Therapeutic Approaches to Nociplastic Pain
Show more Minireview

Similar Articles

Advertisement
  • Home
  • Alerts
Facebook   Twitter   LinkedIn   RSS

Navigate

  • Current Issue
  • Fast Forward by date
  • Fast Forward by section
  • Latest Articles
  • Archive
  • Search for Articles
  • Feedback
  • ASPET

More Information

  • About JPET
  • Editorial Board
  • Instructions to Authors
  • Submit a Manuscript
  • Customized Alerts
  • RSS Feeds
  • Subscriptions
  • Permissions
  • Terms & Conditions of Use

ASPET's Other Journals

  • Drug Metabolism and Disposition
  • Molecular Pharmacology
  • Pharmacological Reviews
  • Pharmacology Research & Perspectives
ISSN 1521-0103 (Online)

Copyright © 2023 by the American Society for Pharmacology and Experimental Therapeutics