Warning: mkdir(): Permission denied in /home/virtual/lib/view_data.php on line 93 Warning: chmod() expects exactly 2 parameters, 3 given in /home/virtual/lib/view_data.php on line 94 Warning: fopen(/home/virtual/pfmjournal/journal/upload/ip_log/ip_log_2024-10.txt): failed to open stream: No such file or directory in /home/virtual/lib/view_data.php on line 100 Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 101 Advances in induced pluripotent stem cell-based in vitro disease modeling for rare neurodegenerative disease: A narrative review
Precis Future Med Search

CLOSE


Precis Future Med > Volume 8(3); 2024 > Article
Lee, Son, Seo, Na, and Kim: Advances in induced pluripotent stem cell-based in vitro disease modeling for rare neurodegenerative disease: A narrative review

Abstract

Understanding the genetic basis and pathomechanisms underlying dementia arising from single-gene mutations is crucial to expand our knowledge in the field of dementia research. In this review, we comprehensively summarize the results of existing research using induced pluripotent stem cells (iPSCs) to investigate familial Alzheimer’s disease caused by mutations in the presenilin-1 (PSEN1), presenilin-2 (PSEN2), or amyloid precursor protein (APP) genes. We further review existing iPSC studies in leukodystrophies, including Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), caused by mutations in the notch receptor 3 (NOTCH3) gene; and adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), caused by mutations in the colony-stimulating factor 1 receptor (CSF1R) gene. We systematically review the advantages and necessity of using iPSCs in research to elucidate the pathogenesis of neurodegenerative diseases, particularly to facilitate disease modeling. Furthermore, we introduce applied research based on iPSC technology. Through this review, we aimed to help elucidate the mechanisms by which causative genes induce dementia symptoms in neurodegenerative diseases, which would help contribute to the development of effective treatment strategies.

INTRODUCTION

Recent advancements in the field of induced pluripotent stem cell (iPSC) research have shown promise, helping to address the complexities of neurodegenerative diseases. Since iPSCs were first introduced by the Yamanaka research team [1], this technology has been applied across numerous fields, revolutionizing various aspects of biomedical research and clinical practice. iPSCs are created from adult somatic cells that have been reprogrammed to an embryonic-like pluripotent state through the introduction of specific transcription factors, including SRY-box transcription factor 2 (Sox2), octamer binding transcription factor 4 (Oct4), KLF4, and c-myc [1]. More recently, iPSCs have been applied in both disease modeling and personalized medicine. Using iPSC technology, specific cell types of interest can be generated from a patient’s somatic cells, allowing for the creation of patient-specific drug testing models.
Neurodegenerative diseases encompass a diverse group of disorders characterized by progressive degeneration of the central nervous system, leading to neuronal loss and impaired glial cell function. Neurodegenerative diseases result in significant impairment of cognitive and motor functions. The application of iPSC technology in the research of neurodegenerative diseases has several advantages. First, the conventional approach for studying neurodegenerative diseases relies heavily on animal models, which show significant physiological differences with humans. Unlike animal models, iPSC-based disease models offer a more accurate context for investigating the complex pathophysiology of human neurodegenerative conditions. Second, iPSCs can be derived relatively easily from a patient’s fibroblasts or peripheral blood mononuclear cells, whereas human brain tissue samples can only be obtained post-mortem. Third, patient-derived iPSCs retain mutant variants of disease-causing genes, possess the properties of embryonic stem cells that can be differentiated into disease-specific brain cells, including neurons, glia, astrocytes, and oligodendrocytes, enabling the investigation of disease pathology in vitro.
Although dementia is a common phenotype of neurodegenerative diseases, rare forms of dementia are caused by single-gene mutations. Understanding the genetic basis and pathomechanism of dementia arising from single-gene mutations is crucial for expanding our knowledge in the field of dementia research. Familial Alzheimer’s disease (AD) accounts for less than 1% of all AD cases. Mutations in the presenilin-1 (PSEN1), presenilin-2 (PSEN2), or amyloid precursor protein (APP) genes lead to the onset of familial AD; however, the exact mechanism of these genes are still being explored. Recent advances in iPSC models have paved the way for investigating the role of these genes in the pathogenesis of AD. Research using iPSC technologies has also facilitated our understanding of rare leukodystrophies, such as Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), caused by mutations in the notch receptor 3 (NOTCH3) gene; and adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), caused by mutations in the colony-stimulating factor 1 receptor (CSF1R) gene.
This review focuses on summarizing the current state of iPSC-based research, particularly in modeling dementia caused by single-gene mutations, highlighting the extent of current investigations and their implications. Furthermore, we summarize existing studies incorporating organoids, three-dimensional (3D) culture systems, drug screening, and clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) technology. By examining these advanced methodologies, this review aimed to delineate and propose future research directions for rare neurodegenerative diseases attributed to specific gene mutations.

ALZHEIMER’S DISEASE

PSEN1, PSEN2, APP

AD is the most common neurodegenerative disease worldwide. Familial AD is characterized by the early onset of symptoms, caused by mutations in PSEN1, PSEN2, or APP genes. These mutations can increase the production of amyloid-beta (Aβ) peptides, particularly the aggregation-prone Aβ42, contributing to amyloid plaque formation.
The existing studies using iPSC technology to enhance our understanding of AD are summarized in Table 1. To investigate the effect of these causative genes on Aβ mechanistic progress, iPSC technology has been utilized to examine AD pathology in various type of brain cells, including neurons, microglia, and astrocytes [2-5]. Studies using iPSC-derived neurons (iN) and neural progenitor cells (iNPC) with APP and PSEN1 mutations have reported increased production of β-amyloid and a higher Aβ42/40 ratio compared to normal cell lines, thus confirming the modeling of AD [2,5,6]. Regarding the metabolic impact of PSEN1 ∆E9 mutation in AD, studies using iPSC-derived astrocytes revealed pathological hallmarks and changes in neuron-supportive functions, including increased oxidative stress and reduced lactate secretion [3]. In addition, iPSC-derived basal forebrain cholinergic neurons (BFCNs) harboring PSEN2 N141I mutation have been utilized to explore the mechanisms underlying short-term memory formation in AD, revealing electrophysiological deficits in BFCNs from AD lines [4]. Further, to delve deeper on memory formation within APP mutation, Pomeshchik et al. [7] generated iPSC-derived hippocampal spheroids, subsequently uncovering a reduction in synaptic proteins and an increased Aβ 42/40 peptide ratio, thereby elucidating the mechanisms underlying memory deficits in AD. Furthermore, to better mimic the complexity of the human brain, further studies have reported the successful modeling of 3D culture cerebral organoids for sporadic AD, as well as those carrying PSEN1 mutations [8-10].
iPSCs have also been leveraged as a research tool in bioinformatics research. For example, alterations in gene expression have been identified through comprehensive molecular profiling and analysis of single-cell transcriptomes in cells carrying PSEN1 and APP mutations [11,12]. For drug screening, iPSC-derived cortical neurons carrying the G384A mutation in the PSEN1 gene were used to screen 1,000 pharmaceutical compounds, revealing 27 compounds which reduced Aβ load [13]. Another drug screening study using iNs revealed several compounds that target cholesterol metabolism, thereby reducing phospho-Tau accumulation, suggesting therapeutic potential for AD [14].
Aside from studies on rare familial AD-causing genes, research on sporadic AD has aimed to understand the mechanisms of apolipoprotein E4 (APOE4), which is the single greatest risk factor for AD. Using CRISPR/Cas9 engineering, isogenic paired cell lines have been generated to investigate how APOE4 contributes to the impairment of synaptic function, lipid metabolism, and immune responses in iNs, astrocytes, and microglial cells [15-17].

CADASIL

CADASIL is a rare genetic small-vessel disease resulting from specific inherited variants of the NOTCH3 gene [18]. Individuals with CADASIL commonly experience recurrent strokes that can lead to cognitive impairment and vascular dementia [19]. The pathological changes observed in CADASIL, including vascular smooth muscle cell (VSMC) degeneration and accumulation of the NOTCH3 protein, all contribute to vascular dysfunction through the deposition of granular osmiophilic materials around VSMCs [20,21]. The central nervous system has a distinct structure, known as the neurovascular unit, that facilitates the functional connections between blood vessels and neurons. This unit primarily comprises brain microvascular endothelial cells (BMEC), mural cells, astrocytes, and neurons [22]. These cells work in concert to regulate the blood-brain barrier (BBB) function and cerebral blood flow, thereby ensuring homeostasis of the central nervous system [23].
To understand the role of NOTCH3 mutations in the neurovascular unit of CADASIL, various in vitro studies have been conducted to observe the pathological changes in multiple cell types derived from iPSCs [24,25]. For example, Zhang et al. [24] generated an iPSC-derived neurovascular unit model including BMECs, astrocytes, and cortical projection neurons, and co-cultured each cell type in a transwell to mimic the BBB in order to investigate the effect of the NOTCH3 mutant on neurovascular interactions and BBB function. Zhang et al. [24] found impaired electrical resistance in mural cells and astrocytes, along with significantly decreased barrier function and disorganized tight junctions in BMECs in the NOTCH3 mutant neurovascular unit model. Another study further showed that mural cells derived from iPSCs carrying the NOTCH3 mutation (R182C, R141C, C106R) exhibited increased platelet-derived growth factor receptor β (PDGFRβ) expression, abnormal structure and distribution of the filamentous actin network, and the Notch3 extracellular domain (N3ECD)/latent-transforming growth factor β-binding protein-1 (LTBP-1)/high temperature requirement A1 (HtrA1)-immunopositive deposits, thus recapitulating pathogenic changes of CADASIL [25]. In another CADASIL research, alterations in gene expression within the NOTCH and nuclear factor κB signaling pathways, as well as cytoskeleton disorganization were observed in VSMCs which were differentiated from patient-derived iPSCs [26].
Moreover, several studies have generated human blood vessel organoids that exhibit CADASIL-like pathology in order to mimic human blood vessel-like structures and investigate vascular cell interactions at both the cellular and tissue levels [27,28].

ALSP

ALSP is a rare, autosomal dominant, white matter degenerative disorder characterized by cognitive impairment arising from mutations in the CSF1R gene [29]. CSF1R is a tyrosine kinase receptor, and approximately 75% of ALSP-CSF1R mutations are located in the tyrosine kinase domain [30]. CSF1R plays a vital role in the proliferation and survival of microglia, as well as in the differentiation of neural progenitor cells [30]. This led to the hypothesis that individuals with CSF1R mutations associated with ALSP may display imbalanced microglial function and impaired neuronal activity.
Since CSF1R mutations impair microglial proliferation and survival, the generation of an in vitro model of patient-derived iPSC differentiation into microglia harboring CSF1R mutations is challenging. Previous studies have generally used CSF1R haploinsufficient (Csf1r+/-) mouse models or zebrafish models with CSF1R mutations to investigate the mechanistic relationship between CSF1R and microglia [31,32]. However, despite these efforts, the exact mechanisms linking ALSP and CSF1R remains elusive, partly because of species differences as mouse or zebrafish models are different from humans.
Researchers have generated ALSP-iPSC disease models from fibroblasts of ALSP patients [33] or by using the CRISPR/Cas9-based CSF1R knockout technology from iPSC lines of normal human [34]. However, as CSF1R gene is critical in the differentiation of microglia and neural progenitor cells, there was a hurdle to differentiate ALSP-iPSCs into these cell types. A recently reported novel protocol allows for more efficient differentiation of iPSCs from ALSP patients with CSF1R mutations into microglia [35]. Using this novel protocol, the differentiated ALSP-induced microglia transcriptomic analysis revealed a gene expression pattern similar to that of human microglia [35]. Moreover, these cells display characteristics of ALSP pathology, including decreased CSF1R autophosphorylation, impaired migratory ability, and reduced purinergic receptor P2Y12 (P2RY12) expression [35].
Following the successful establishment of ALSP-induced microglial lines carrying CSF1R mutations, future studies should focus on exploring the mechanistic correlation between CSF1R and microglia in a human-centric manner. This could be achieved by investigating gene-corrected isogenic lines using CRISPR/Cas9 technology. Furthermore, investigating the phenotype in 3D cultures using a co-culture system of neurons and non-neuronal cells [36] or organoids [37,38] will be crucial in understanding the complex interactions within the brain microenvironment. Additionally, leveraging insights from research on microglial phenotypes in other diseases, and applying them to ALSP pathology, will help enhance our understanding of this disease.

APPLICATIONS

iPSCs have diverse applications, ranging from differentiation into disease-specific cell types, to exploration of the mechanisms using gene-editing technologies in cell models. Additionally, iPSCs have been actively studied for their potential in expanding into 3D culturable organoids, cell replacement therapy, and drug screening. For example, cell replacement therapy involving the injection of iPSC-derived dopamine progenitor cells into a humanized mouse model, has been investigated in Parkinson’s disease, another common neurodegenerative condition [39]. iPSC disease modeling can further be used for drug screening, facilitating not only high-throughput drug discovery but also the development of personalized medicine tailored to individual symptoms, in vivo safety and efficacy testing of developed drugs, and examination of host-microbe interactions [40]. Additionally, gene-editing technologies are expected to be increasingly used in therapeutic studies owing to continuous advancements such as Cas12f, which reduces off-target effects and enhances delivery [41]. These advancements further highlight the critical role of iPSCs in understanding disease pathology and developing therapeutic strategies. The importance of iPSC research in the field of rare diseases is expected to increase, offering new insights and treatment avenues.

FUTURE DIRECTIONS

Although iPSCs have significant potential in the field of rare neurodegenerative diseases, there are some limitations that should be considered in future research. Owing to their cellular reprogramming to the pluripotent state, iPSCs exhibit rejuvenated epigenetic features, including DNA and cytosine-phosphate-guanine (CpG) methylation, which are both signatures of aging [42]. To overcome this issue, research is currently underway on aging induction by directly reprogramming somatic cells to iNs [43], or by treating iPSC-derived cells with chemicals such as telomerase inhibitors [44]. Further advancements in gene-editing technologies for iPSCs have raised concerns regarding off-target effects and genomic instability [45,46]. Despite technological advancements, iPSCs still face limitations in accurately replicating the mechanical structure and environmental conditions of human organs in dishes, even with the development of 3D-culturable organoids [47-49]. Furthermore, while iPSCs offer the advantage of autologous transplantation [45], their immortalized nature raises concerns regarding issues such as cell engraftment, immunogenicity, and tumorigenicity, which must be carefully addressed to ensure their safety and efficacy [50-52]. As such, a comprehensive understanding of the molecular characteristics of these models is essential to harness their full potential in the context of neurodegenerative diseases.

CONCLUSION

Research utilizing iPSC-based disease modeling has made it feasible to investigate disease phenotypes and their underlying mechanisms in rare neurodegenerative diseases caused by single-gene mutations. Various iPSC-derived brain cell types have successfully replicated the phenotypes of familial AD, CADASIL, and ALSP. Additionally, many studies have employed gene editing to create isogenic iPSC lines for the exploration of pathogenic pathways. These findings underscore the significance of iPSC-based research in advancing our understanding of rare neurodegenerative diseases.

CONFLICTS OF INTEREST

Sang Won Seo has been editorial board of Precision and Future Medicine since December 2017. He was not involved in the review process of this review article. No potential conflict of interest relevant to this article was reported.

Notes

AUTHOR CONTRIBUTIONS

Conception or design: SL, HJK.

Acquisition, interpretation of data: SL, HJK.

Drafting the work or revising: SL, HJS, HJK.

Final approval of the manuscript: SL, HJS, SWS, DLN, HJK.

ACKNOWLEDGEMENTS

This research was supported by the National Research Foundation of Korea (NRF) grant funded by the Ministry of Education, Republic of Korea (NRF-2022R1A2C2092346). Additional funding was provided by the Ministry of Health & Welfare, Republic of Korea (HX22C0027), and by the Ministry of Science and ICT (MSIT), Korea, under the ICT Creative Consilience Program (IITP-2024-2020-0-01821), supervised by the Institute for Information & communications Technology Planning & Evaluation (IITP).

Table 1.
Summary of the results of studies utilizing iPSCs for neurodegenerative diseases
Disease Gene mutations Differentiated cell type Application Major findings Reference
AD APP (V171I) PSEN1 (int4del, Y115H, M139V, M146I, R278I) Neuron Disease modeling Supported Aβ mechanistic tenets in a human physiological model and substantiate iPSC-neurons for modeling fAD [2]
AD PSEN1 (∆E9) Astrocyte Disease modeling Increased oxidative stress [3]
Reduced lactate secretion
Role of astrocytes in AD pathology, including changes in neuron-supportive function
AD PSEN2 (N141I) Basal forebrain cholinergic neurons Disease modeling Defective electrophysiological properties [4]
AD APP (V717I) Neuron Disease modeling Increased Aβ42 and Aβ38 [5]
Increased total and phosphorylated tau
AD PSEN1 (M146L, A246E) Neural progenitor cells Disease modeling Increased in ABβ42/40 peptide ratio [6]
AD APP (V717I, PSEN1, R278K) Hippocampal spheroids Disease modeling Increased Aβ42/Aβ40 peptide ratio [7]
Decreased synaptic protein levels
Examination of early pathological changes in humans
AD hippocampal parenchyma-like model
AD PSEN1 (A246E) Cerebral organoid Disease modeling AD-like pathology [8]
PSEN2 (N141I) React to β- and g-secretase inhibitors by decreasing levels of Aβ peptide
Developmental and tissue patterning defects
Single cell-sequencing revealed altered development and signs of premature differentiation
AD APP knockin or knockout Neuron Genome-wide molecular profiling An isogenic APP Swe/PSEN1 M146V ‘‘double-mutant’’ iPSC line (dAP) homozygous for both APP swe and PSEN1 M146V was developed, in addition to other various fAD mutated iPSC lines [12]
PSEN knockin Total of 16 iPSC fAD lines (8 APP mutated lines, PSEN1 mutated lines, and 1 dAP iPSC line) were used. All iPSC lines were edited by CRISPR
Common alterations in early endosomes mediated by accumulation of β-CTF, not Aβ
AD PSEN1 (G384A) Cortical neuron Drug screening A combination of existing drugs (anti-Aβ cocktail, bromocriptine, cromolyn, and topiramate) synergistically improved Aβ phenotypes of AD [13]
This anti-Aβ cocktail decreases toxic Aβ levels in neurons derived from patient cells
CADASIL NOTCH3 (R153C, C224Y) BMEC Disease modeling Impaired electrical resistance in mural cells and astrocytes [24]
Mural cell Decreased barrier function and disorganized tight junctions in BMECs
Astrocyte
CADASIL NOTCH3 (R182C, R141C, C106R) Mural cell Disease modeling Increased PDGFRβ expression, abnormal structure and distribution of the filamentous actin network, presence of N3ECD/LTBP-1/HtrA1-immunopositive deposits [25]
CADASIL NOTCH3 (R1076C) VSMC Disease modeling Alterations in gene expression within the NOTCH and NF-κB signaling pathways, as well as cytoskeleton disorganization [26]
CADASIL NOTCH3 (R153C, R182C) Blood vessel organoid Disease modeling Induction of mutations by CRISPR/Cas9 base editing [27]
Reduced vessel diameter, accumulation of NOTCH3 extracellular domain, degeneration of mural cells, and increased apoptosis and cytoskeletal alterations
CADASIL NOTCH3 (R421C) VSMC Disease modeling iPSC-derived VSMC showed NOTCH3 deposition and abnormal actin cytoskeleton structure [28]
Blood vessel organoid Gene expression downregulation of cell adhesion, vessel development in CADASIL blood vessel organoid
NOTCH3 mutations could be corrected using the dual Adeno associated virus split- adenine based editor max system
ALSP CSF1R (V838L) - Disease modeling Generation of iPSCs from ALSP patient fibroblasts [33]
ALSP CSF1R knockout (CSF1R+/-, CSF1R-/-) - Disease modeling CSF1R Knockout by CRISPR/Cas9 based gene editing [34]
ALSP CSF1R (V784M) Microglia Disease modeling Transcriptomic analysis revealed similarities to human primary microglia [35]
Decreased CSF1R autophosphorylation
Impaired migratory ability
Reduced P2RY12 expression

iPSC, induced pluripotent stem cell; AD, Alzheimer’s disease; APP, amyloid precursor protein; PSEN1, presenilin-1; Aβ, amyloid-beta; fAD, familial Alzheimer’s disease; PSEN2, presenilin-2; CRISPR/Cas9, clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9; β-CTF, β c-terminal fragment; CADASIL, Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy; BMEC, brain microvascular endothelial cell; PDGFRβ, platelet-derived growth factor receptor β; N3ECD, Notch3 extracellular domain; LTBP-1, latent-transforming growth factor β-binding protein-1; HtrA1, high temperature requirement A1; VSMC, vascular smooth muscle cell; NF-κB, nuclear factor κB; ALSP, adult-onset leukoencephalopathy with axonal spheroids and pigmented glia; CSF1R, colony-stimulating factor 1 receptor; P2RY12, purinergic receptor P2Y12

REFERENCES

1. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007;131:861–72.
crossref pmid
2. Arber C, Toombs J, Lovejoy C, Ryan NS, Paterson RW, Willumsen N, et al. Familial Alzheimer’s disease patient-derived neurons reveal distinct mutation-specific effects on amyloid beta. Mol Psychiatry 2020;25:2919–31.
crossref pmid pmc pdf
3. Oksanen M, Petersen AJ, Naumenko N, Puttonen K, Lehtonen S, Gubert Olive M, et al. PSEN1 mutant iPSC-derived model reveals severe astrocyte pathology in Alzheimer’s disease. Stem Cell Reports 2017;9:1885–97.
crossref pmid pmc
4. Ortiz-Virumbrales M, Moreno CL, Kruglikov I, Marazuela P, Sproul A, Jacob S, et al. CRISPR/Cas9-correctable mutation-related molecular and physiological phenotypes in iPSC-derived Alzheimer’s PSEN2 N141I neurons. Acta Neuropathol Commun 2017;5:77.
crossref pmid pmc pdf
5. Muratore CR, Rice HC, Srikanth P, Callahan DG, Shin T, Benjamin LN, et al. The familial Alzheimer’s disease APPV717I mutation alters APP processing and Tau expression in iPSC-derived neurons. Hum Mol Genet 2014;23:3523–36.
crossref pmid pmc
6. Sproul AA, Jacob S, Pre D, Kim SH, Nestor MW, Navarro-Sobrino M, et al. Characterization and molecular profiling of PSEN1 familial Alzheimer’s disease iPSC-derived neural progenitors. PLoS One 2014;9:e84547.
crossref pmid pmc
7. Pomeshchik Y, Klementieva O, Gil J, Martinsson I, Hansen MG, de Vries T, et al. Human iPSC-derived hippocampal spheroids: an innovative tool for stratifying Alzheimer disease patient-specific cellular phenotypes and developing therapies. Stem Cell Reports 2020;15:256–73.
crossref pmid pmc
8. Vanova T, Sedmik J, Raska J, Amruz Cerna K, Taus P, Pospisilova V, et al. Cerebral organoids derived from patients with Alzheimer’s disease with PSEN1/2 mutations have defective tissue patterning and altered development. Cell Rep 2023;42:113310.
crossref pmid
9. Chen X, Sun G, Tian E, Zhang M, Davtyan H, Beach TG, et al. Modeling sporadic Alzheimer’s disease in human brain organoids under serum exposure. Adv Sci (Weinh) 2021;8:e2101462.
crossref pmid pmc pdf
10. Sreenivasamurthy S, Laul M, Zhao N, Kim T, Zhu D. Current progress of cerebral organoids for modeling Alzheimer’s disease origins and mechanisms. Bioeng Transl Med 2022;8:e10378.
crossref pmid pmc pdf
11. Sun N, Victor MB, Park YP, Xiong X, Scannail AN, Leary N, et al. Human microglial state dynamics in Alzheimer’s disease progression. Cell 2023;186:4386–403.
crossref pmid pmc
12. Kwart D, Gregg A, Scheckel C, Murphy EA, Paquet D, Duffield M, et al. A large panel of isogenic APP and PSEN1 mutant human iPSC neurons reveals shared endosomal abnormalities mediated by APP β-CTFs, not Aβ. Neuron 2019;104:256–70.
crossref pmid
13. Kondo T, Imamura K, Funayama M, Tsukita K, Miyake M, Ohta A, et al. iPSC-based compound screening and in vitro trials identify a synergistic anti-amyloid β combination for Alzheimer’s disease. Cell Rep 2017;21:2304–12.
crossref pmid
14. van der Kant R, Langness VF, Herrera CM, Williams DA, Fong LK, Leestemaker Y, et al. Cholesterol metabolism is a druggable axis that independently regulates tau and amyloid-β in iPSC-derived Alzheimer’s disease neurons. Cell Stem Cell 2019;24:363–75.
crossref pmid pmc
15. Lee H, Cho S, Kim MJ, Park YJ, Cho E, Jo YS, et al. ApoE4-dependent lysosomal cholesterol accumulation impairs mitochondrial homeostasis and oxidative phosphorylation in human astrocytes. Cell Rep 2023;42:113183.
crossref pmid
16. Lin YT, Seo J, Gao F, Feldman HM, Wen HL, Penney J, et al. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron 2018;98:1141–54.
crossref pmid pmc
17. Raman S, Brookhouser N, Brafman DA. Using human induced pluripotent stem cells (hiPSCs) to investigate the mechanisms by which apolipoprotein E (APOE) contributes to Alzheimer’s disease (AD) risk. Neurobiol Dis 2020;138:104788.
crossref pmid pmc
18. Joutel A, Corpechot C, Ducros A, Vahedi K, Chabriat H, Mouton P, et al. Notch3 mutations in CADASIL, a hereditary adult-onset condition causing stroke and dementia. Nature 1996;383:707–10.
crossref pmid pdf
19. Chabriat H, Joutel A, Dichgans M, Tournier-Lasserve E, Bousser MG. Cadasil. Lancet Neurol 2009;8:643–53.
crossref pmid
20. Gravesteijn G, Hack RJ, Mulder AA, Cerfontaine MN, van Doorn R, Hegeman IM, et al. NOTCH3 variant position is associated with NOTCH3 aggregation load in CADASIL vasculature. Neuropathol Appl Neurobiol 2022;48:e12751.
crossref pmid pmc pdf
21. Joutel A, Andreux F, Gaulis S, Domenga V, Cecillon M, Battail N, et al. The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. J Clin Invest 2000;105:597–605.
crossref pmid pmc
22. Coomber BL, Stewart PA. Morphometric analysis of CNS microvascular endothelium. Microvasc Res 1985;30:99–115.
crossref pmid
23. Daneman R. Prat A. The blood-brain barrier. Cold Spring Harb Perspect Biol 2015;7:a020412.
crossref pmid pmc
24. Zhang W, Zhao X, Qi X, Kimber SJ, Hooper NM, Wang T. Induced pluripotent stem cell model revealed impaired neurovascular interaction in genetic small vessel disease Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy. Front Cell Neurosci 2023;17:1195470.
crossref pmid pmc
25. Yamamoto Y, Kojima K, Taura D, Sone M, Washida K, Egawa N, et al. Human iPS cell-derived mural cells as an in vitro model of hereditary cerebral small vessel disease. Mol Brain 2020;13:38.
crossref pmid pmc pdf
26. Ling C, Liu Z, Song M, Zhang W, Wang S, Liu X, et al. Modeling CADASIL vascular pathologies with patient-derived induced pluripotent stem cells. Protein Cell 2019;10:249–71.
crossref pmid pmc pdf
27. Ahn Y, An JH, Yang HJ, Lee WJ, Lee SH, Park YH, et al. Blood vessel organoids generated by base editing and harboring single nucleotide variation in Notch3 effectively recapitulate CADASIL-related pathogenesis. Mol Neurobiol 2024;Apr 9 [Epub]. https://doi.org/10.1007/s12035-024-04141-4.
crossref pmid
28. Wang J, Zhang L, Wu G, Wu J, Zhou X, Chen X, et al. Correction of a CADASIL point mutation using adenine base editors in hiPSCs and blood vessel organoids. J Genet Genomics 2024;51:197–207.
crossref pmid
29. Rademakers R, Baker M, Nicholson AM, Rutherford NJ, Finch N, Soto-Ortolaza A, et al. Mutations in the colony stimulating factor 1 receptor (CSF1R) gene cause hereditary diffuse leukoencephalopathy with spheroids. Nat Genet 2011;44:200–5.
pmid pmc
30. Hu B, Duan S, Wang Z, Li X, Zhou Y, Zhang X, et al. Insights into the role of CSF1R in the central nervous system and neurological disorders. Front Aging Neurosci 2021;13:789834.
crossref pmid pmc
31. Oosterhof N, Kuil LE, van der Linde HC, Burm SM, Berdowski W, van Ijcken WF, et al. Colony-stimulating factor 1 receptor (CSF1R) regulates microglia density and distribution, but not microglia differentiation in vivo. Cell Rep 2018;24:1203–17.
crossref pmid
32. Biundo F, Chitu V, Shlager GG, Park ES, Gulinello ME, Saha K, et al. Microglial reduction of colony stimulating factor-1 receptor expression is sufficient to confer adult onset leukodystrophy. Glia 2021;69:779–91.
crossref pmid pmc pdf
33. Hayer SN, Schelling Y, Hoeflinger P, Hauser S, Schols L. Generation of an induced pluripotent stem cell line from a patient with adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP): HIHCNi003-A. Stem Cell Res 2018;30:206–9.
crossref pmid
34. Schmitz AS, Korneck M, Raju J, Lamsfus-Calle A, Daniel-Moreno A, Antony JS, et al. Generation of a heterozygous and a homozygous CSF1R knockout line from iPSC using CRISPR/Cas9. Stem Cell Res 2023;69:103066.
crossref pmid
35. Dorion MF, Casas D, Shlaifer I, Yaqubi M, Fleming P, Karpilovsky N, et al. An adapted protocol to derive microglia from stem cells and its application in the study of CSF1R-related disorders. Mol Neurodegener 2024;19:31.
crossref pmid pmc pdf
36. Beghini DG, Kasai-Brunswick TH, Henriques-Pons A. Induced pluripotent stem cells in drug discovery and neurodegenerative disease modelling. Int J Mol Sci 2024;25:2392.
crossref pmid pmc
37. Pazzin DB, Previato TT, Budelon Goncalves JI, Zanirati G, Xavier FA, da Costa JC, et al. Induced pluripotent stem cells and organoids in advancing neuropathology research and therapies. Cells 2024;13:745.
crossref pmid pmc
38. Kim J, Koo BK, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol 2020;21:571–84.
crossref pmid pmc pdf
39. Schweitzer JS, Song B, Herrington TM, Park TY, Lee N, Ko S, et al. Personalized iPSC-derived dopamine progenitor cells for Parkinson’s disease. N Engl J Med 2020;382:1926–32.
crossref pmid pmc
40. Marei HE, Khan MU, Hasan A. Potential use of iPSCs for disease modeling, drug screening, and cell-based therapy for Alzheimer’s disease. Cell Mol Biol Lett 2023;28:98.
crossref pmid pmc pdf
41. Harrington LB, Burstein D, Chen JS, Paez-Espino D, Ma E, Witte IP, et al. Programmed DNA destruction by miniature CRISPR-Cas14 enzymes. Science 2018;362:839–42.
crossref pmid pmc
42. Aversano S, Caiazza C, Caiazzo M. Induced pluripotent stem cell-derived and directly reprogrammed neurons to study neurodegenerative diseases: the impact of aging signatures. Front Aging Neurosci 2022;14:1069482.
crossref pmid pmc
43. Mertens J, Herdy JR, Traxler L, Schafer ST, Schlachetzki JC, Bohnke L, et al. Age-dependent instability of mature neuronal fate in induced neurons from Alzheimer’s patients. Cell Stem Cell 2021;28:1533–48.
crossref pmid pmc
44. Vera E, Bosco N, Studer L. Generating late-onset human iPSC-based disease models by inducing neuronal age-related phenotypes through telomerase manipulation. Cell Rep 2016;17:1184–92.
crossref pmid pmc
45. Chuang K, Fields MA, Del Priore LV. Potential of gene editing and induced pluripotent stem cells (iPSCs) in treatment of retinal diseases. Yale J Biol Med 2017;90:635–42.
pmid pmc
46. Zhang Z, Bao X, Lin CP. Progress and prospects of gene editing in pluripotent stem cells. Biomedicines 2023;11:2168.
crossref pmid pmc
47. Qian X, Song H, Ming GL. Brain organoids: advances, applications and challenges. Development 2019;146:dev166074.
crossref pmid pmc pdf
48. Xu R, Boreland AJ, Li X, Erickson C, Jin M, Atkins C, et al. Developing human pluripotent stem cell-based cerebral organoids with a controllable microglia ratio for modeling brain development and pathology. Stem Cell Reports 2021;16:1923–37.
crossref pmid pmc
49. Zhang W, Jiang J, Xu Z, Yan H, Tang B, Liu C, et al. Microglia-containing human brain organoids for the study of brain development and pathology. Mol Psychiatry 2023;28:96–107.
crossref pmid pmc pdf
50. Park TY, Jeon J, Cha Y, Kim KS. Past, present, and future of cell replacement therapy for Parkinson’s disease: a novel emphasis on host immune responses. Cell Res 2024;34:479–92.
crossref pmid pmc pdf
51. Liu L, Huang JS, Han C, Zhang GX, Xu XY, Shen Y, et al. Induced pluripotent stem cells in Huntington’s disease: disease modeling and the potential for cell-based therapy. Mol Neurobiol 2016;53:6698–708.
crossref pmid pmc pdf
52. Nori S, Okada Y, Nishimura S, Sasaki T, Itakura G, Kobayashi Y, et al. Long-term safety issues of iPSC-based cell therapy in a spinal cord injury model: oncogenic transformation with epithelial-mesenchymal transition. Stem Cell Reports 2015;4:360–73.
crossref pmid pmc


ABOUT
ARTICLES

Browse all articles >

ISSUES
TOPICS

Browse all articles >

EDITORIAL
POLICY
AUTHOR
INFORMATION
Editorial Office
Sungkyunkwan University School of Medicine
2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Korea
Tel: +82-31-299-6038    Fax: +82-31-299-6029    E-mail: pfmjournal@skku.edu                

Copyright © 2024 by Sungkyunkwan University School of Medicine.

Developed in M2PI

Close layer
prev next