Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Mar 13.
Published in final edited form as: Annu Rev Genet. 2018 Sep 12;52:271–293. doi: 10.1146/annurev-genet-120417-031534

Aging in a Dish: iPSC-Derived and Directly Induced Neurons for Studying Brain Aging and Age-Related Neurodegenerative Diseases

Jerome Mertens 1,2, Dylan Reid 1, Shong Lau 1, Yongsung Kim 1, Fred H Gage 1
PMCID: PMC6415910  NIHMSID: NIHMS1009811  PMID: 30208291

Abstract

Age-associated neurological diseases represent a profound challenge in biomedical research as we are still struggling to understand the interface between the aging process and the manifestation of disease. Various pathologies in the elderly do not directly result from genetic mutations, toxins, or infectious agents but are primarily driven by the many manifestations of biological aging. Therefore, the generation of appropriate model systems to study human aging in the nervous system demands new concepts that lie beyond transgenic and drug-induced models. Although access to viable human brain specimens is limited and induced pluripotent stem cell models face limitations due to reprogramming-associated cellular rejuvenation, the direct conversion of somatic cells into induced neurons allows for the generation of human neurons that capture many aspects of aging. Here, we review advances in exploring age-associated neurodegenerative diseases using human cell reprogramming models, and we discuss general concepts, promises, and limitations of the field.

Keywords: aging, induced neurons, iNs, disease modeling, age-associated neurodegeneration, induced pluripotent stem cells, iPSCs, rejuvenation

INTRODUCTION

Humans are exceptionally long-lived in comparison to other primates, with a projected maximum lifespan of approximately 115 years (24). This extreme longevity presents unique challenges to many of the postmitotic cells that compose our bodies and makes us highly susceptible to age-associated diseases (AADs) such as cancer, diabetes, and various neurodegenerative diseases (NDDs). Although many different research approaches are being pursued to better understand the process of aging (81), as well as to assess the interface between aging and disease, there is no unifying concept that fully tackles the complexity of the biology of aging (36). As the global mean population age continues to rapidly increase, AADs constitute an enormous burden on societies’ health care systems. Further complicating the issue is the fact that many AADs lack effective therapies, due in large part to failed drug discovery efforts in recent years. These shortfalls are united by the common thread that our basic understanding of AADs has been limited by our ability to successfully generate workable human models in the laboratory. Although the animal models we rely on so heavily to model AADs are useful tools to grasp generalized concepts related to aging and the genes involved in age-related decline, they may not reflect specific mechanisms of human disease. In order to bridge this knowledge gap, better understand the process of aging in humans and its contribution to disease, and translate our findings into new therapeutic advances, we must develop better models that are more reflective of our biology (108).

To understand AADs, many laboratories have adopted an approach that relies on modeling diseases in a dish with induced pluripotent stem cells (iPSCs) (7). Primary human cells can be collected directly from patients, and the cells can then be reset to an embryonic-like state with the transient expression of the Yamanaka transcription factors, OCT4, KLF4, SOX2, and C-MYC (OKSM) (127). These cells can then be coaxed to differentiate into various cell types such as neurons, myocytes, or hepatocytes, thus allowing for cell-autonomous features to be probed in the context of the disease of interest (90). In recent years, this iPSC approach has expanded to the development of organoids—collections of cells that reflect features of whole tissues (71). However, the iPSC approach is fundamentally limited in its ability to model aging because expression of the OKSM transcription factors acts as a developmental reset, erasing many of the desired features required for AAD modeling (121).

As an alternative to iPSC reprogramming and differentiation, so-called direct conversion or transdifferentiation strategies to maintain the age-associated features of donor cells are being employed (20, 90). Transcription factors, microRNAs, and chemical cues are used to encourage terminally differentiated cells to directly transform between lineages and thereby bypass the rejuvenating embryonic state. Although many induced cell models are now available, this review largely focuses on the modeling of aging in induced neurons (iNs). Neurons are among the longest living cells in the human body; most are generated in embryonic development, with few new neurons being produced after birth (29). Neurons from primary human sources are also exceptionally difficult to obtain and maintain, making direct study highly challenging. Thus, iNs generated from human donors present a unique opportunity to begin to understand how the aging process impacts these cells and can give rise to various NDDs. This review addresses the key features of aging that appear to drive neurodegeneration, compares the iPSC and direct conversion approaches for next-generation human cellular modeling, and discusses the extent to which iNs reflect aged neurons. We hope the review provokes a thoughtful examination of the nature of neuron-specific aging in humans and of the power of this model to address human disease.

AGING AND NEURODEGENERATION

Aging is the progressive loss of cellular homeostasis, leading to an overall decline of organismal fitness. Over the past 30 years, the contributions of numerous molecular mechanisms and pathways that define the etiology of the aging process have been elucidated (81). Many of these aging pathways are also involved in NDDs such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD). The risk of developing these diseases highly correlates with age, and it, is difficult to predict susceptibility because the vast majority of cases are sporadic, i.e., lacking a well-defined genetic component (35, 59). Although many of the key features that define aging are associated with these diseases, we wish to primarily address features of age that are highly challenging to neurons (Figure 1).

Figure 1.

Figure 1

Intersection of age-related dysfunction. Most age-related pathways are often thought of as independent with little overlap between the various components. ➊ In neurons, as advancing age occurs, nuclear transport becomes altered and pore leakiness increases. ➋ Damaged nuclear pores lose their capability to regulate nuclear homeostasis, genome architecture, DNA modification, and transcription. ➌ Additionally, DNA lesions can accumulate via errors in transcription or in the removal of epigenetic methylation markers, and persistent lesions fundamentally alter metabolism due to bioenergetic stress; ➍ this leads to changes at the mitochondrial level, which in turn increase the amount of reactive oxygen species (ROS) produced and damage the genome further. ➎ DNA damage triggers global stress responses in cells and leads to proteostasis disruption, potentially further destabilizing nuclear pores.

Genomic Integrity and Metabolism

As the longest living cells in the human body, neurons are faced with the unique problem of maintaining their genetic material for decades of life (21). In the process of becoming postmitotic cells, neurons inactivate or transcriptionally downregulate many of the pathways associated with the DNA damage response (DDR) and focus the bulk of their DNA repair efforts on transcriptionally active genes (40). As neurons age, they accumulate lesions in these neuronal genes, resulting in decreased transcriptional output and a shift in cellular metabolism to maintain cell viability (55, 141). Additionally, mitochondria become more compromised with age, leading to an increase in free reactive oxygen species (ROS) production, which is a potent DNA damaging agent (31). DNA repair can often be compromised further in neurodegeneration (88). In ALS/FTD, mutations in the protein FUS are known to disrupt genomic integrity, and the C9orf72 repeat expansion RNA and proteins promote the formation of R-loops, leading to DNA damage (83). Neurons also appear to employ induced DNA breaks to trigger transcription of immediate early genes, the repair of which can be delayed by the extracellular amyloid β (Aβ) found in AD (83, 122). Most interesting, however, is the strong link found between persistent DNA damage and altered metabolism in neurons.

Our understanding of this process is largely viewed through the lens of developmental neurodegeneration models such ERCC1−/Δ7 mice and human patients with Cockayne syndrome (CS). ERCC1−/Δ7 mice are deficient for the nucleotide excision repair complex formed by ERCC1–XPF, leading to a progeria phenotype, a progressive loss of neurons, and greater risk for PD development (117). As the neurons in these mice accumulate unrepaired DNA lesions, the persistent DDR signaling then subsequently suppresses insulin-like growth factor-I signaling, leading to decreased function of these cells but greater longevity (141). The effects can be further augmented by dietary intervention through caloric restriction. Similarly, in CS, the key DNA repair helicase ERCC6 is less than optimally functional, leading to unrepaired DNA lesions or unresolved G-quadruplex secondary structures in ribosomal DNA (115). These act as persistent sources of damage that induce poly (ADP-ribose) polymerase (PARP) activity and consume nuclear NAD+ (114). Depletion of NAD+ locally in the nucleus decreases SIRT1 activity, and it, along with DNA-PK and ATM kinase, downregulates mitochondrial activity and biogenesis (30). Finally, telomeres in aging neurons present a potentially unique source of persistent damage, as the shelterin complex protecting them can prevent repair of lesions and lead to persistent signaling (34).

Transcription and Epigenetics

There is no unifying measure for the age of a cell or tissue, and the deviation between chronological age and different measures of biological age is currently a matter of debate and ongoing research (46, 161). Neurons must maintain their cellular identity for a considerably longer period of time than do most other postmitotic cells. In large part, cellular identity can be defined by the appropriate transcription of neuronal genes and the maintenance of appropriate epigenetic marks on the chromatin. Aged tissues display a general increase in transcriptional noise and a loss of regulation, contributing mechanistically to features of the aging process (6, 42). Cellular aging has been further refined through newer single-cell sequencing techniques, showing that the accumulation of mutation and transcriptional noise drives loss of cellular identity (28, 80). As part of the aging transcriptome, global hypomethylation of the genome occurs, but certain key regions become hypermethylated (10, 46). This altered methylation can be calibrated to accurately predict the age of cells, allowing for accurate age typing of iNs.

Nuclear Pores and Proteostasis

Nuclear pore complexes are composed of nucleoporins that control the flow of information between the nucleus and the cytoplasm of eukaryotic cells (45). These pores fenestrate the nucleus and appropriately traffic various transcription factors and RNAs, allowing cells to respond to signals in their local environment (14). Furthermore, these nuclear pore complexes appear to act as platforms for gene regulation, transcription, and global nuclear organization (18). A recent surprising discovery is that nuclear pore permeability becomes altered with age (22, 113, 136). Pores get more permeable to cytoplasmic proteins entering the nucleus and are increasingly leaky for nuclear proteins with advancing age. This leakiness is in part due to the low turnover of extremely long-lived nucleoporins that form the scaffold and core of the channel. These proteins are being efficiently incorporated only during mitosis; therefore, it is likely that many nucleoporins are as old as the neurons themselves. Thus, nuclear pore-associated damage and functional defects are a prime example of age-related protein damage that likely has tremendous downstream effects on the subcellular localization of transcription factors and other regulatory proteins and directly impacts chromatin structure and transcription (51). Finally, in ALS/FTD, low-complexity proteins such as poly-PR, FUS, or TDP-43 often further compromise pores by blocking them (57, 62). These and other disease-related proteins coaggregate with FG nuclear pore proteins in the center channel of pores (39, 44, 118). Collectively, these observations suggest a unique nexus of age-related dysfunction that may arise in neurons.

INDUCED PLURIPOTENT STEM CELL MODELS FOR AGE-ASSOCIATED DISEASE—SOMETHING MISSING?

The study of NDDs has been hindered by the inability to access living human brain tissue for research purposes. Further, postmortem samples from patients reflect only the end stage of the disease, making it particularly difficult to unravel the specific pathogenic mechanisms involved in initiating the disease. The advent of human iPSC technology and refined differentiation strategies have provided well-defined patient-derived neural cells, including various types of neural stem cells (NSCs), dopaminergic neurons, glutamatergic neurons, GABAergic neurons, motor neurons, astrocytes, oligodendrocytes, and many others, to model NDDs (5, 19, 47, 67, 68, 76, 86, 159). Although many NDDs can be caused by genetic mutations, the majority of cases arise sporadically and exclusively at advanced ages. This age dependency highlights the need for patient-specific models that not only reflect the genotype of the patients but also reflect molecular aging signatures. Here iPSC-based models face a major challenge: iPSCs are, as advertised, reprogrammed to an earlier epigenetic state (101, 110). This iPSC rejuvenation has been demonstrated at the level of epigenetic methylated DNA (mDNA) and the transcriptome (33, 46, 79, 89, 91), as well as in telomeres (84, 124) and several functional phenotypes, including mitochondrial function, cellular senescence, DNA damage, and others (72, 100, 107, 125). Although iPSC reprogramming can be seen as an advantage for cell replacement strategies and rejuvenation-directed research (101), it represents a major challenge for modeling AADs.

Primary cell samples obtained from donors have unique features that have accumulated throughout the lifespan and disease development of the individual. These features are important for understanding AAD mechanisms. Because iPSC reprogramming rejuvenates cells on many levels, and because clonal selection is typically used to further select and screen the fittest cells, neurons from typical iPSC clones may not be the most characteristic cells to model AADs. In fact, most iPSC-based disease models for NDDs have demonstrated a requirement for additional stressors applied to normal culture conditions to elicit disease-specific phenotypes. For example, studies using iPSC-derived dopaminergic neurons to model familial PD consistently used ROS or mitochondria-related stressors such as H2O2, 6-OHDA, valinomycin, or CCCP to detect PD neuron-specific malfunctions (98, 109, 111, 116). Similarly, proteasome inhibition using compounds such as MG132 was required to obtain protein aggregates in Huntington’s disease (HD) iPSC neurons (97), and excitotoxic glutamate exposure was needed to stimulate the formation of ataxin-3-containing microaggregates in iPSC neurons from Machado-Joseph disease patients (64). Further, overexpression of pseudohyperphosphorylated tau in human embryonic stem cell (hESC)-derived neurons induced neurite transport defects and cell death only when antioxidants were removed from the culture (92). Interestingly, familial AD-causing mutations in PS1 or APP, which can lead to early onset of AD symptoms at the age of 40, were shown to consistently alter Aβ production without the need for additional stressors (54, 66, 93, 148, 151). However, excitotoxic glutamate exposure was needed to induce cell death (26). These findings reflect how aberrant Aβ is already present in very young familial AD patient neurons but fails to induce significant neuronal malfunctions unless features of aging also come into play. Though not inconsistent with the amyloid hypothesis of AD, these data are in line with the paradigm shift in the AD field where more and more focus is dedicated to alternative disease mechanisms, including age-related pathways (21, 36). Taken together, even if application of stressors helps reveal disease-specific phenotypes in iPSC neurons, it remains unclear whether they elicit a type of stress that is adequate to better understand the disease in phenotypically and epigenetically young neurons.

A more systematic and comprehensive strategy to induce broad aspects of aging in iPSC models was first introduced in a study utilizing expression of a premature aging-associated protein variant in iPSC neurons (94). Expression of the Hutchinson-Gilford progeria syndrome (HGPS)-causing variant of the LMNA gene progerin resulted in the induction of a series of aging markers, including γH2AX and H2K9me, in otherwise rejuvenated neurons. Progerin also triggered pathological phenotypes in PD patient-specific dopamine neurons, such as dendritic degeneration and impairment of AKT signaling (94). Another study showed that telomerase inhibition at the iPSC stage using the compound BIBR1532 could also induce age-related phenotypes in neurons derived from treated iPSCs (140). Although telomere shortening with age in postmitotic neurons is an up-and-coming field of research (27), more work needs to be done to clarify the role of overall telomere length, as well as active telomere shortening, in mature postmitotic human neurons. The mere existence of telomerase activity in iPSC-derived neurons warrants further investigation. Although both age-inducing strategies represent significant steps forward in modeling AADs with iPSCs, it remains unclear whether the latter approach imitates normal human aging or represents only a pathological type of aging. Clearly, these strategies to induce aging in iPSC derivatives can recapitulate only certain aspects of aging; it will be interesting to determine to what extent different strategies can be combined to provide a more complete representation of actual cellular aging in neurons. This review focuses on direct iN conversion, an alternative strategy for generating human neurons to study aging and AADs that allows circumvention of the pluripotent stem cell stage.

DIRECT LINEAGE CONVERSION INTO INDUCED NEURONS

Despite the rapid progress in the directed differentiation of iPSCs (96, 126), extensive effort has also gone into the development of alternative strategies for obtaining functional cells. Direct reprogramming, which is defined as the direct conversion of cells from one lineage to another without going through the pluripotent stage (38), holds great promise for biomedical applications. The first study to demonstrate the possibility of this strategy was the conversion of fibroblasts into myoblasts by overexpressing MYOD1 (23). The concept that lineage conversion could be achieved by cell type-specific transcription factors and was possible between distant cell types led to protocols for the direct conversion of many cell types, including blood cell lineages, neurons, cardiomyocytes, hepatocytes, pancreatic cells, and macrophages (49, 52, 70, 144, 163).

The reprogramming of mouse fibroblasts into functional iNs was first achieved by overexpressing a set of proneural transcription factors, BRN2, ASCL1, and MYT1L, also known as BAM (144). A year later, a study from the same group used BAM together with NEUROD1 to convert fetal human fibroblasts into functional neurons (103). A number of other studies also reported successful conversion of other cell types into functional neurons (60, 85). The field has since progressed rapidly to investigate the mechanisms involved in direct conversion and pursue the best strategy, including use of novel transcription factors and compounds, for generating neurons that have comparable properties to in vivo or iPSC-differentiated counterparts (Table 1).

Table 1.

Neuronal reprogramming of human cells without going through the pluripotent stage

Neuronal subtype(s) iN method(s) Cell source(s) Developmental stage of source Reference
Dopaminergic neurons ASCL1, BRN2, MYT1L, LMX1A, FOXA2 Fibroblasts Fetal 106
Dopaminergic neurons ASCL1, LMX1A, NURR1 Fibroblasts Fetal, adult 16
Glutamatergic neurons ASCL1, BRN2, MYT1L, NEUROD1 Fibroblasts Fetal 103
Glutamatergic neurons miR-124, BRN2, MYT1L Fibroblasts Adult 4
Glutamatergic neurons, GABAergic neurons miR-9/9*, miR-124, ASCL1, MYT1L, NEUROD2 Fibroblasts Fetal, adult 156
Motor neurons ASCL1, BRN2, MYT1L, LHX3, HB9, ISL1, NGN2 Fibroblasts Fetal 120
Glutamatergic neurons ASCL1, SOX2 Pericytes Adult 60
Glutamatergic neurons, GABAergic neurons ASCL1, NGN2 plus small molecules Fibroblasts Neonatal, adult 69
Cholinergic neurons NGN2 plus small molecules Fibroblasts Fetal, neonatal 78
Dopaminergic neurons ASCL1, BRN2, MYT1L, LMX1A, LMX1B, FOXA2, OTX2 Fibroblasts Fetal 134
Retinal pigment epithelium PAX6, RAX, CRX, MITF-A, OTX2, NRL, KLF4, C-MYC Fibroblasts Adult 158
Neurons, unspecified subtype(s) PTB inhibition Multiple cell lines Embryonic and adult-derived cell lines 150
Neurons, unspecified subtype(s) ASCL1, BRN2, MYT1L, miR-124 negative regulation Fibroblasts Fetal 73
Glutamatergic neurons, GABAergic neurons ASCL1, BRN2, MYT1L Fibroblasts Fetal 105
GABAergic, medium spiny neurons miR-9/9*, miR-124, CTIP2, DLX1, DLX2, MYT1L Fibroblasts Adult 142
Sensory neurons NGN1, NGN2, BRN3A Fibroblasts Embryonic, adult 8
Glutamatergic neurons plus GABAergic neurons and other subtypes NGN2, ASCL1 plus small molecules Fibroblasts Adult 91
Glutamatergic neurons and other subtypes VPA, CHIR99021, Repsox, Forskolin, SP600125, GO6983, Y-27632, Dorsomorphin Fibroblasts Adult 48
Serotonergic neurons NGN2, ASCL1, LMX1B, FEV, NKX2.2, GATA2 Fibroblasts Adult 139
Serotonergic neurons ASCL1, LMX1B, FEV, FOXA2 Fibroblasts Fetal, neonatal, adult 149
Spinal motor neurons NGN2, SOX11, ISL1, LHX3 Fibroblasts Adult 77
Neurons, unspecified subtype(s) ASCL1, BRN2, REST inhibition Fibroblasts Fetal, adult 25
Dopaminergic neurons ASCL1, LMX1A, NURR1, NEUROD1, miR-218 Astrocytes Fetal (immortalized), embryonic 112

Abbreviations: iN, induced neuron; REST, RE1-silencing transcription factor.

Conversion Factors for Induced Neurons

The reprogramming of one cell type to another requires appropriate changes in epigenetic status, which are mediated by transcription factors. During development, cell fate is determined by stepwise changes in the cellular epigenetic profile mediated by a genetically defined cascade of transcription factors. In the process of reprogramming, the pioneering factors play a crucial role in initiating lineage conversion. Pioneer factors are capable of accessing the closed region of chromatin just by themselves and can help other factors to initiate cell fate change. In BAM iN conversion, ASCL1 acts as a pioneer factor, accessing the closed chromatin region and recruiting BRN2 to ASCL1 target sites (146). This mechanism suggests that, although simultaneously expressed, the transcription factors still coordinate in a hierarchical manner. Neurogenin2 (NGN2) is another well studied proneural transcription factor that has been widely reported to initiate iN reprogramming in human fibroblasts with the aid of small molecules (78). Several research groups have used a dual-pioneer factor strategy (ASCL1 and NGN2) that yields high conversion efficiencies, even in adult and aged human fibroblasts (69, 160). This approach indicated the feasibility of using adult donor-derived fibroblasts as a starting cell source for the study of AADs, including NDDs. Importantly, several studies showed that the transient expression of transcription factors is sufficient to induce neural conversion; prolonged expression is not necessary. In most studies, an inducible expression system is used (69, 91, 103, 134); however, it is also possible to employ an endogenous neuron-specific miRNA to negatively regulate the factors and achieve the optimal timing for factor expression (73).

In addition to transcription factors, alternative factors such as miRNAs, short hairpin RNA (shRNA), and small molecules can facilitate direct fate conversion. For iN, strategies include the overexpression of the neuron-specific miRNAs miR-9/9* and miR-124, which, however, still requires co-overexpression of transcription factors (4, 156). Furthermore, mouse iNs have been generated by small interfering RNAs to inhibit PTB, which blocks miRNA-mediated activity of the RE1-silencing transcription factor (REST) complex (150), or a shRNA targeting the REST complex directly (25). Both studies showed that inhibition of the REST complex triggers the expression of epigenetically silenced proneural genes in fibroblasts and therefore facilitates the neuron induction process, at least in the mouse system.

Although most iN methods are based on the use of lentiviral vectors, these viruses raise safety concerns with regard to clinical applications and potential integration-induced artifacts (see also the section titled Induced Neuron Models for Age-Associated Diseases). Recently, cocktails of small molecules have been reported to promote iN conversion and reduce the requirement for exogenous factors (69, 78, 105). Inhibition of TGFβ/SMAD signaling with Noggin and SB431542, together with GSK3β inhibitor, forskolin, and cyclic-AMP, has been shown to improve iN conversion. Notably, recent studies have demonstrated that it is possible to induce neurons with only a cocktail of small molecules and without exogenous transcription factors. One study induced mouse neurons with SMAD and GSK3β inhibition and the addition of the proneurogenic molecules ISX9 and I-BET151, which inhibit fibroblast-specific gene expression (74). In another study, human neurons were induced with histone deacetylase inhibitor VPA, GSK3β inhibitor RepSox, JNK inhibitor SP600125, PKC inhibitor GO6983, and ROCK inhibitor Y-27632 (48). Both studies marked important steps in replacing exogenous factors with small molecules, which have several major advantages such as cost effectiveness, comparatively easy synthesis and preservation, and possible standardization. However, identifying small molecules that reproducibly and completely replace exogenous transcription factors during lineage reprogramming remains a major challenge for iN conversion.

Subtype-Specific Induced Neurons

The originally identified BAM iNs often exhibited glutamatergic neuronal properties (144), and NGN2/ASCL1 iNs yielded a major fraction of glutamatergic neurons, a minor fraction of GABAergic neurons, and single dopaminergic and serotonergic neurons (91). Since the neural lineage is constituted of a vast number of neuronal subtypes, researchers are actively investigating the induction of different functional neuronal subtypes such as dopaminergic, cholinergic, GABAergic, serotonergic, medium spiny, and motor neurons (Table 1) (16, 77, 78, 106, 120, 138, 139, 142, 149, 152). These iN subtypes have all been shown to possess the required basic neuronal functional properties, and they also exhibit subtype-specific functions such as dopamine release or responsiveness to selective serotonin reuptake inhibitors (SSRIs), for example (106, 139). iN induction involves hierarchical coordination between the pioneer factor and other transcription factors. The strategies used to generate subtype-specific iNs involve pioneer factors such as ASCL1 and NGN2, which are used alongside a collection of factors that have been shown to be important in neural subtype determination. Combinations include NGN2 and SOX11 for cholinergic neurons; NGN2 with SOX11, ISL1, and LHX3 for motor neurons (77); the pioneer factors alongside LMX1A, LMX1B, NURR1, PITX3, OTX2, and FOXA2 for dopaminergic iNs (16, 106); the pioneer factors alongside CTIP2, DLX1, and DLX2 for GABAergic medium spiny neurons (142); the pioneer factors alongside LMX1B, FEV with FOXA2, or NKX2.2 and GATA2 for serotonergic neurons (139, 149); and NGN2 alongside NGN1 and BRN3A for sensory neurons (8). In general, the induction of functional lineage subtypes indicates that iN conversion is highly plastic and holds great application potential in modeling subtype-specific neurological disorders. However, identifying the cocktail of subtype-determining factors that faithfully convert fibroblasts into mature and fully functional neurons that are comparable to their in vivo counterparts remains a major focus.

INDUCED PLURIPOTENT STEM CELLS VERSUS INDUCED NEURONS

It is known that both iPSC differentiation and direct conversion can generate functional neurons, but it is important to choose the best method for the questions being asked. Differentiation protocols for human pluripotent stem cells, including human iPSCs and hESCs have been developed and optimized over a much longer period of time than for those of iNs, but the relatively new technology of direct conversion is rapidly developing, with many new protocols being published in recent years. There are several technical and conceptual differences between differentiation and direct conversion protocols.

Cell Source

The starting cell sources described for iNs are limited. Although iN conversion has been reported successfully with multiple starting cell types such as astrocytes, NG2 glia, hepatocytes, and adipocytes (43, 85, 112, 134, 155), most of these cell types represent, or are derived from, fetal rather than adult murine or human tissues. Currently, reports on successful iN conversions with human adult cells, use of which is a necessity for modeling AADs with iNs, have been limited to pericytes and skin fibroblasts. However, pericytes are relatively hard to obtain from donors, so skin fibroblasts remain the most feasible option as a cell source at the moment. In contrast, iPSC reprogramming protocols using many human cell types, including skin fibroblasts, blood cells (102), keratinocytes (1), and renal epithelial cells (162), are successful and robust. It is important to mention that pluripotent stem cells are also important cell sources for direct reprogramming. Since hESCs and iPSCs are epigenetically naïve, they possess the full potential to faithfully convert into the desired lineage by expressing appropriate transcription factors that determine cell fate commitment. iPSCs have been successfully induced into mature neurons (iPSC-iNs) by overexpressing NGN2 (132) and into subtype-specific iNs (95, 130).

Time and Cost Efficiency

Because research budgets are unfortunately limited, time and money play important roles when deciding between differentiation and conversion protocols. The time to generate stable iPSC clones is typically 2–3 months, including basic characterization. Once the clone is successfully established, it can be expanded indefinitely and provide a stable supply of cells. The differentiation of iPSCs to neurons takes around 6–15 weeks, depending on the protocol and desired level of maturity (99). Many iPSC differentiation protocols, involving a stable and expandable progenitor cell stage that can be generated from iPSCs within 2–3 weeks, are used by many laboratories as stable intermediates (65). The iPSC differentiation protocol requires application of several different recipes of media over a precise timeline to mimic molecular signaling changes in their niche during development.

iN conversion from fibroblasts generates neurons that express neuronal markers in 2–4 weeks and show basic functional properties such as evoked action potentials. It takes another 5–6 weeks coculturing with astrocytes before iNs typically show mature physiological features such as spontaneous action potentials and postsynaptic currents (91, 103). The conversion procedure is relatively simple, usually involving one conversion medium for the conversion phase and one maturation medium for the maturation phase.

Expandability

iPSCs, once derived, can theoretically expand infinitely. The progenitors derived during differentiation also possess reliable self-renewing ability. Furthermore, iPSCs and NSCs are comparably small cells and grow at very high density. These properties make iPSC differentiation suitable for the generation of large numbers of neurons for subsequent applications like transplantation and metabolomic, proteomic, transcriptomic, and genomic analyses that require relatively large amounts of material. Direct iN conversion does not involve a highly expandable intermediate stage, and the neurons generated are postmitotic. Therefore, the expandable phase of iN conversion depends entirely on the proliferation of fibroblasts. As fibroblasts cannot infinitely expand and will go senescent after multiple passages, there is a limitation of starting materials even if a proliferating “iN-ready” transgenic cell line can be established using antibiotic-selectable vectors.

INDUCED NEURON MODELS FOR AGE-ASSOCIATED DISEASES

The fundamental challenge in the generation of human cellular disease models is the ability to represent the diseased cell in the body in a meaningful and predictive way. For studies using directly reprogrammed or iPSC-derived neuronal cells to study cellular and molecular mechanisms in the human system, the cellular composition of the cultures, neuronal subtype identity, and maturation state of the cultures are of central importance. When it comes to harnessing generated neurons as model systems to study aspects of monogenic or multigenic neurological diseases, the genetic identity of the generated cells needs to adequately reflect the genetic identity of the donor’s cells in the body. Specifically, as mutations are acquired through tissue culture and the reprogramming process (123), and due to the potential importance of human somatic mosaicism (2, 87), issues associated with the clonality of the derived cell lines emerged as a focus of researchers. Additionally, in the case of using reprogrammed neurons for modeling AADs, the biological age of the generated neurons is thought to be a most influential risk factor for a variety of modeled diseases and thus deserves special attention. Among the various risks for human diseases, age stands out as being a special multifaceted factor. Unlike other risk factors, biological age interacts with other risk factors to drive NDDs, many of which we have mentioned previously. Below, we summarize how different iN-based studies have addressed aging and disease-related phenotypes in iNs (see also Table 2).

Table 2.

Studies that employed induced neuron (iN) conversion to study aging and age-associated diseases (AADs)

Donor age(s) Disease-related mutation(s) iN methods Neuronal subtype(s) Phenotypes Compared to iPSCs Reference
1 year (n = 1 mouse) None ASCL1, BRN2, MYT1L Glutamatergic neurons and GABAergic neurons Age-related gene expression, ROS, cell death Yes 154
0–89 years (n = 18) None NGN2, ASCL1 Glutamatergic neurons, GABAergic neurons, and other Transcriptomic aging, RanBP17 loss, compartmentalization defects, nuclear transport rates Yes 91
43–71 years (n = 5) Healthy and ALS C9orf72 repeat expansions NGN2, ASCL1 Glutamatergic neurons, GABAergic neurons, and other ALS-specific: nucleo-cytoplasmic RanGEF/RCC1 mislocalization No 56
37–71 years (n = 6) Healthy and ALS FUS mutations NGN2, SOX11, ISL1, LHX3 Spinal motor neurons ALS-specific: nucleo-cytoplasmic FUS mislocalization, defects in morphology, physiology, survival, and impaired NMJ formation No 77
0–96 years (n = 15) Unknown miR-9/9*, miR-124, CTIP2, DLX1, DLX2, MYT1L GABAergic striatal neurons Epigenetic (mDNA) age, transcriptomic aging, ROS, DNA damage (comet assay) No 50
0–71 years (n = 6) None and ALS FUS mutations NGN2, SOX11, ISL1, LHX3 Spinal motor neurons DNA damage (γH2AX), dystrophic nuclear envelope, loss of epigenetic markers Yes 128
0–89 years (n = 12) None NGN2, ASCL1 Glutamatergic neurons, GABAergic neurons, and other Mitochondrial dysfunctions (gene expression, morphology, membrane potential, ATP), oxidized proteins, neurite beading Yes 63
6–71 years (n = 29) mHTT (CAG repeat expansions) miR-9/9*, miR-124, CTIP2, DLX1, DLX2, MYT1L GABAergic striatal neurons HD-related gene expression, peri- and intranuclear mHTT aggregates, impaired proteostasis, ROS, DNA damage, neuronal cell death Yes (proteostasis phenotype only) 143

Abbreviations: ALS, amyotrophic lateral sclerosis; HD, Huntington’s disease; iPSCs, induced pluripotent stem cells; mHTT, mutated huntingtin, NMJ, neuromuscular junction; RanGEF, Ran guanine nucleotide exchange factor; ROS, reactive oxygen species.

Age-Related DNA Lesions are Retained in Induced Neurons and, with Exceptions, also in Induced Pluripotent Stem Cells

The accumulation of DNA lesions that become somatic mutations is thought to play a role in aging and AADs. Cumulative DNA mutations are believed to be the central age-dependent component in the development of cancers (21), but they might also play an instrumental role in the initiation of AD (15). Neither iN conversion nor iPSC reprogramming generally alters the genetic identity, including somatic mutations, of reprogrammed cells. However, there are several notable exceptions. First, the use of retro- and lentiviral vectors results in the irreversible integration of DNA sequences into the genome, even if tetracycline-regulated or self-silencing strategies are employed. Whereas many improved systems to minimize reprogramming-associated integrations have been applied in iPSC reprogramming (61, 147), and the use of commercially available nonintegrating Sendai virus has become almost a standard in the iPSC field, efforts to generate integration-free iNs has received some, but markedly less, attention (3, 73). One reason clearly is that iN generation is by far a less widespread and younger technology, and Sendai viruses for iN factors such as ASCL1, BRN2, NGN2, NEUROD1, and others are not yet commercially available. Another more specific reason is that iN conversion, in contrast to iPSC reprogramming, is not clonal, meaning that each converted cell harbors a different pattern of integrations, thereby minimizing integration-related artifacts when bulk cultures or larger numbers of single iNs are assessed. This concept clonality versus random multicellular integration extends from studies of nuclear DNA to mitochondrial DNA (mtDNA), where elegant work has described the transmission of age-related mtDNA mutations in different hetero- and homoplasmic iPSC clones from aged human donors (53, 58). Importantly, the existence of endogenous somatic mosaicism, age-related or not, is an important issue to consider when interpreting and comparing results from iPSC-based studies (2, 87). Further, genomic artifacts have been widely reported in iPSCs and have made the careful karyotype analysis of all generated iPSC clones obligatory. However, it is not clear to what extent clonal artifacts are truly the result of the reprogramming process or if they are caused by cell culture in general, or if they actually reflect a mosaic genotype and just become visible in clonal cell lines.

More important for the sustained functionality and viability of neurons than the number of accumulated DNA mutations is the error-free functionality of DDR pathways that, according to calculations, repair over 100,000 DNA lesions per cell every day (75). Thus, it would be interesting to measure not only mutation load in aging iNs but also the efficiency of DDR pathways; however, their functionality is technically difficult to measure. Indications that DDR might be compromised in aged human neurons comes from an elegant study that found increased numbers of γH2AX foci in aged donor fibroblast-derived iNs, whereas iPSC-derived neurons from the same aged donors did not show more γH2AX foci than their young donor-derived counterparts (128).

Induced Neurons Retain Long-Lived Protein Damage; Induced Pluripotent Stem Cell Reprogramming Erases it

As opposed to DNA mutations, protein damage generally does not persist in the numerous cell divisions and anabolic requirements of iPSC reprogramming. In fact, even during the first days of iPSC reprogramming, the cells appear to largely reset their proteome, demonstrating the extremely high protein turnover and anabolic need associated with acquiring iPSC identity (41). Interestingly, the nucleoporin NUP210, which is part of the oldest protein complex of the cell forming the nuclear pore scaffold structure, undergoes dramatic protein expression changes during early reprogramming, illustrating the rejuvenating character of iPSC reprogramming on the level of damaged proteins of the nuclear pore (41). In contrast, iN conversion does not involve cell division and conserves nuclear pore-associated aging features. Fibroblast-derived glutamatergic/GABAergic iNs from 1- to 89-year-old donors showed a marked decline in nucleo-cytoplasmic compartmentalization and nuclear shuttling rates with age, which is in part caused by the age-related loss of the importin-β family transport factor RanBP17 (91). Consistently, induced motor neurons from aged donors also showed changes in the nuclear envelope structure, as assessed by LaminB1 (128). As expected, protein compartmentalization in iPSCs as well as in iPSC-derived neurons from the same donors did not show age-related differences, thus rendering iN the model system of choice for studying age-related changes that might involve nuclear pore function and protein localization (91, 128). Indeed, glutamatergic/GABAergic iN cultures from middle-aged ALS patients carrying the disease-causing C9orf72 repeat expansions were shown to possess faulty localization of the nuclear transport regulator Ran guanine nucleotide exchange factor (RanGEF/RCC1) (56). These results using patient-specific iNs confirmed data from yeast and Drosophila showing that nuclear transport factors were major modulators of C9orf72 toxicity (32, 56, 157). Further, induced motor neurons from 37- to 50-year-old ALS patients carrying FUS mutations were shown to possess impaired nucleo-cytoplasmic localization of the low complexity protein FUS, and they were unable to form neuromuscular junctions, a phenotype that could be ameliorated, however, by a kinase inhibitor known to inhibit CDK1, 2, and 5 as well as GSK-3β (77). Similarly, unlike iPSC neurons (97), iNs from HD patients showed age-related proteostatic impairments and spontaneously developed aggregates composed of mutated huntingtin (mHTT) with peri- and intranuclear localization (143). Together, these studies suggest that both aging-related and ALS/HD-genotype-related stressors might converge in parallel at the nuclear pore level to promote protein mislocalization, neuronal dysfunction, and eventually neurodegeneration.

Another important example of age-related cumulative protein damage is mitochondria; a major pillar of the theory of age-related mitochondrial dysfunction is that increased production of ROS damages mtDNA and oxidizes mitochondrial proteins, which in turn further increases mitochondrial dysfunction (12). Indeed, data from healthy aged donor iNs that showed neurite beading and a decrease in synaptic transmission genes also revealed stark age-related mitochondrial defects, including suppression of tricarboxylic acid cycle (TCA) and oxidative phosphorylation (OXPHOS)-related genes and proteins, increased levels of oxidized proteins, mitochondrial morphology defects, loss of mitochondrial membrane potential, and decreased ATP production (63). Interestingly, the fibroblasts from which the iNs were derived showed only mild mitochondrial aging defects that, however, could be artificially induced by forcing the fibroblasts from their typical glycolytic bioenergetics into a neuron-like OXPHOS-dependent bioenergetic profile. Affirmation of these observations also comes from other studies where iNs from aged donors showed increased levels of ROS (50). ROS levels were found to be even higher in HD patients’ iNs, where they were also accompanied by signs of DNA damage and neuronal degeneration (143).

Epigenetic and Transcriptional Aging in Induced Neurons

The epigenome can be regarded as a convergence platform for the features of many intrinsic and extrinsic factors that confront a cell over its lifetime. Age-dependent epigenetic changes (also referred to as epigenetic drift) are among the best-studied features of aging research (46, 129, 161). Although iPSCs possess only minimal epigenetic memory of their tissue of origin, epigenetic (mDNA) profiling using the aging clock has demonstrated that iPSCs even from very old donors reset to a prenatal epigenetic age (46, 79). In contrast, iNs possess the same epigenetic age as their parental fibroblasts, demonstrating that the epigenetics can be transferred from fibroblasts to neurons (50). This is an important finding also because it shows that, even though iN conversion reorganizes large parts of the human epigenome (119), it apparently leaves the most age-related marks virtually untouched. Further, comparative studies between iNs and iPSCs from young and aged donors showed that iNs derived from aged donors possessed age-related differences in the levels of H3K9me3 in humans (128), as well as other epigenetic marks (154). Consequently, even on the transcriptome level, iNs possess stark age-related signatures that share similarities with the transcriptomic aging signatures detected in postmortem human brain samples (11, 82, 91). Notably, transcriptomic aging signatures between iNs and their parental fibroblasts differ dramatically with only a small overlap, thus suggesting a model where cell type–independent epigenetic aging changes that remain untouched during iN conversion lead to distinct transcriptomic changes in different aging cell types. Unlike iNs, iPSCs were shown to be reliably erased of transcriptomic aging information (see the section titled Induced Pluripotent Stem Cell Models for Age-Associated Disease—Something Missing?). It will be interesting to explore to what extent aging iNs display transcriptional noise and transcriptional identity drift on the single-cell level (28) and if this erosion of epigenetic marks leads to transcriptional changes that relate to AADs.

A CONCEPT FOR AGE-PRESERVATION IN LINEAGE CONVERSION

Studies employing direct lineage conversion have demonstrated that features of age can be directly transferred from one cell type to another (Table 2). Importantly, these findings demonstrate that the functional manifestation of old age in a cell is not just a hodgepodge of different dysfunctional pathways but that aging follows a certain hierarchical degree of organization. All the way upstream, at the top of the hierarchy, reside aging regulator pathways (ARPs) that are mainly tissue- and cell type-independent and that have the power to induce different cell type-specific downstream aging features in different cell types. This is good news, because this hierarchical concept of aging suggests that a limited number of target features of cellular aging control most other aging phenotypes. Currently, it still appears too early to point to one master ARP, but data seem to focus attention on epigenetic (mDNA) aging as well as nuclear pore defects. Epigenetic changes directly control the transcriptome output of a cell, which, however, differs largely between cell types (i.e., fibroblast aging genes are different from iN aging genes). Surprisingly, epigenetic (mDNA) signatures that define age were shown to be left untouched during iN conversion (50) and were not overwritten during the major conversion-related epigenetic overhaul (119,145, 146). Similarly impactful is control by the nuclear pore over subcellular protein and RNA localization, transcription factor activation and deactivation, chromatin structure, and gene expression (51,133). Age-dependent damage to the long-lived proteins of the nuclear pore is a common observation in aging and appears to be specifically relevant to postmitotic neurons (22, 113, 135, 136). Several iN aging studies have consistently described nuclear transport-related defects in aging and AAD iNs (56, 77, 91). Another strong argument for the nuclear pore being an upstream ARP in the hierarchy stems from the fact that the expression of progerin, which can be regarded as a general insult to the nuclear envelope and embedded pores, is sufficient to trigger AAD-related phenotypes in otherwise rejuvenated iPSC-derived neurons (37, 94).

In the quest to generate the perfect model of aging human brain neurons in vitro, iN conversion represents a major step forward, but it also has limitations. Many technical limitations of the iN technology would be solved by having a proliferative intermediate; for instance, it will be interesting to explore to what extent directly induced NSCs retain aging features (131). Further, due to their postmitotic state and the absence of neural progenitors, iNs face technical challenges in adapting into organoid-like 3D culture systems (71). Conceptual limitations will be even more challenging to address. A simplified view of what can be expected from iNs and iPSC models in general is illustrated in Figure 2, which shows that fibroblasts partially contain the relevant aging information for modeling aged brain neurons that is completely encoded by ARPs. This information is mostly preserved in iNs but largely erased in iPSCs—yet there are strategies to artificially induce age in the latter (Figure 2). In the future, it will be important to explore the mechanisms that define the most impactful ARPs, to identify the best conversion strategies to maintain maximum ARP information, to develop new methods of inducing aspects of aging in iPSC models, and to harness the tools at hand for navigating the interface between aging and disease at the disease-, age-, and patient-specific levels.

Figure 2.

Figure 2

Conceptual model of relevant aging information encoded in upstream aging regulator pathways (ARPs) reflected in induced neurons (iNs) and induced pluripotent stem cells (iPSCs) as compared with those of the brain. (a) Fibroblasts contain a large portion of ARP-encoded aging information (dark gray) that is relevant also for (c) brain aging. (b) After iN conversion, the cells preserve a large part of this aging information within ARPs (dark gray), but some information (orange in panel a) has been lost. This would be the case for information that results from neuron–glia interactions for example, which might not be present without adding glial cells to the culture. In contrast, (d) iPSC reprogramming erases the vast majority of aging information in ARPs, leaving only DNA-encoded aging lesions. (e) Approaches to induce aging in rejuvenated iPSC derivatives such as progerin expression (green), telomere attrition (blue), or oxidative stress (red) can be used to fill the glass, but likely also fall short in mimicking major parts of the aging information encoded in the ARPs.

OUTLOOK

Going forward, it behooves us to ask what the future holds for the iN technology and other direct conversion strategies. An interesting and potentially incredibly valuable difference between iPSC differentiation and direct conversion stems from the fact that direct conversion skips certain intermediate developmental stages (13). By combining differentiation and direct conversion, this concept might help to identify disease-related phenotypes that require the intermediate neural progenitor cell stage to unfold in mature neurons. Although iNs are a valuable model for studying aging human neurons in a dish, they remain challenging to generate economically at scale. This is in part due to the choice of dermal fibroblasts as the primary starting material for generating iNs. These cells do not grow very densely, and usually most good patient cohorts are limited to collections of fibroblasts and peripheral blood monocytes. We will need to improve our ability to generate more materials and achieve higher conversion efficiencies before the induced cell model is translatable to large-scale pharmacological drug discovery in the treatment of AADs. For this reason, many investigators are working to better understand how direct conversion of neurons takes place at the single-cell level (137). Many advances in this technology are on the horizon, offering us an improved understanding of what it means to be not just an old neuron but also an old region-specific neuron. Further, although neurons steal much of the limelight, many of the other cells in the nervous system are also extremely old, though quiescent as opposed to postmitotic. These other cell types play support roles in the brain and are often associated with the dysregulation of various pathways in neurodegeneration. Many of the risk alleles for AD, such as TREM2 and APOE4, relate directly to astrocytes and microglia and their ability to destroy Aβ in the extracellular space. Some efforts have been made to demonstrate in animal models the direct reprogramming of fibroblasts to either astrocytes or oligodendrocytes (17, 153). As these studies are translated to the human system, they might provide valuable tools to model AD and ischemic demyelination. Collectively, iNs and related nervous system direct conversion models will eventually allow us to build models for the aging human brain, potentially in the form of induced cell organoids that may recapitulate disease. By directly studying both the cell-autonomous features of each type and the collective cell-nonautonomous disease pathology of the resulting organoids, we will gain a deeper understanding of neurodegeneration and potentially develop treatments for disorders that replace cells using in vivo reprogramming to create new neurons in patients with AD, PD, ALS, stroke, and other age-associated conditions.

ACKNOWLEDGMENTS

We thank Ken Diffenderfer, Simon T. Schafer, and Joseph R. Herdy (Salk Institute); Frank Edenhofer, Larissa Traxler, Lena Böhnke, and Katharina Günther (University of Innsbruck); and Douglas Galasko [University of California, San Diego (UCSD)] for helpful discussions, and Mary Lynn Gage for editorial comments. This review was supported by the Paul G. Allen Family Foundation, the National Institute on Aging R01AG056306–01, the JPB Foundation, the Leona M. and Harry B. Helmsley Charitable Trust, Annette C. Merle-Smith, the G. Harold & Leila Y. Mathers Charitable Foundation, and the Ray and Dagmar Dolby Family Fund to F.H.G., the National Institutes of Health Pathway to Independence Award K99-AG056679–01 and the Leopold-Franzens University of Innsbruck to J.M., the Alzheimer’s Association Research Fellowship to D.R. (AARF-17-504089), and the Shiley-Marcos Alzheimer’s Disease Research Center at UCSD.

Footnotes

DISCLOSURE STATEMENT

The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.

LITERATURE CITED

  • 1.Aasen T, Raya A, Barrero MJ, Garreta E, Consiglio A, et al. 2008. Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nat. Biotechnol 26(11):1276–84 [DOI] [PubMed] [Google Scholar]
  • 2.Abyzov A, Mariani J, Palejev D, Zhang Y, Haney MS, et al. 2012. Somatic copy number mosaicism in human skin revealed by induced pluripotent stem cells. Nature 492(7429):438–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Adler AF, Grigsby CL, Kulangara K, Wang H, Yasuda R, Leong KW. 2012. Nonviral direct conversion of primary mouse embryonic fibroblasts to neuronal cells. Mol. Ther. Nucleic Acids 1(7):e32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ambasudhan R, Talantova M, Coleman R, Yuan X, Zhu S, et al. 2011. Direct reprogramming of adult human fibroblasts to functional neurons under defined conditions. Cell Stem Cell 9(2):113–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Aubry L, Bugi A, Lefort N, Rousseau F, Peschanski M, Perrier AL. 2008. Striatal progenitors derived from human ES cells mature into DARPP32 neurons in vitro and in quinolinic acid-lesioned rats. PNAS 105(43):16707–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bahar R, Hartmann CH, Rodriguez KA, Denny AD, Busuttil RA, et al. 2006. Increased cell-to-cell variation in gene expression in ageing mouse heart. Nature 441(7096):1011–14 [DOI] [PubMed] [Google Scholar]
  • 7.Bellin M, Marchetto MC, Gage FH, Mummery CL. 2012. Induced pluripotent stem cells: the new patient? Nat. Rev. Mol. Cell Biol. 13(11):713–26 [DOI] [PubMed] [Google Scholar]
  • 8.Blanchard JW, Eade KT, Szűcs A, Sardo Lo V, Tsunemoto RK, et al. 2015. Selective conversion of fibroblasts into peripheral sensory neurons. Nat. Neurosci 18(1):25–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Deleted in proof
  • 10.Bormann F, Rodríguez-Paredes M, Hagemann S, Manchanda H, Kristof B, et al. 2016. Reduced DNA methylation patterning and transcriptional connectivity define human skin aging. Aging Cell 15(3):563–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bozdag S, Li A, Riddick G, Kotliarov Y, Baysan M, et al. 2013. Age-specific signatures of glioblastoma at the genomic, genetic, and epigenetic levels. PLOS ONE 8(4):e62982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bratic A, Larsson N-G. 2013. The role of mitochondria in aging. J. Clin. Invest 123(3):951–57 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Briggs JA, Li VC, Lee S, Woolf CJ, Klein A, Kirschner MW. 2017. Mouse embryonic stem cells can differentiate via multiple paths to the same state. eLife 6:e26945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Burns LT, Wente SR. 2014. From hypothesis to mechanism: uncovering nuclear pore complex links to gene expression. Mol. Cell. Biol 34(12):2114–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bushman DM, Kaeser GE, Siddoway B, Westra JW, Rivera RR, et al. 2015. Genomic mosaicism with increased amyloid precursor protein (APP) gene copy number in single neurons from sporadic Alzheimer’s disease brains. eLife 4:e05116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Caiazzo M, Dell’Anno MT, Dvoretskova E, Lazarevic D, Taverna S, et al. 2011. Direct generation of functional dopaminergic neurons from mouse and human fibroblasts. Nature 476(7359):224–27 [DOI] [PubMed] [Google Scholar]
  • 17.Caiazzo M, Giannelli S, Valente P, Lignani G, Carissimo A, et al. 2015. Direct conversion of fibroblasts into functional astrocytes by defined transcription factors. Stem Cell Rep. 4(1):25–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Capelson M, Hetzer MW. 2009. The role of nuclear pores in gene regulation, development and disease. EMBO Rep. 10(7):697–705 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L. 2009. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat. Biotechnol 27(3):275–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chambers SM, Studer L.2011. Cell fate plug and play: direct reprogramming and induced pluripotency. Cell 145(6):827–30 [DOI] [PubMed] [Google Scholar]
  • 21.Chow H-M, Herrup K. 2015. Genomic integrity and the ageing brain. Nat. Rev. Neurosci 16(11):672–84 [DOI] [PubMed] [Google Scholar]
  • 22.D’Angelo MA, Raices M, Panowski SH, Hetzer MW. 2009. Age-dependent deterioration of nuclear pore complexes causes a loss of nuclear integrity in postmitotic cells. Cell 136(2):284–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Davis RL, Weintraub H, Lassar AB. 1987. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell 51(6):987–1000 [DOI] [PubMed] [Google Scholar]
  • 24.Dong X, Milholland B, Vijg J. 2016. Evidence for a limit to human lifespan. Nature 538(7624):257–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Drouin-Ouellet J, Lau S, Brattås PL, Rylander Ottosson D, Pircs K, et al. 2017. REST suppression mediates neural conversion of adult human fibroblasts via microRNA-dependent and -independent pathways. EMBO Mol. Med. 9:1117–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Duan L, Bhattacharyya BJ, Belmadani A, Pan L, Miller RJ, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Eitan E, Hutchison ER, Mattson MP. 2014. Telomere shortening in neurological disorders: an abundance of unanswered questions. Trends Neurosci. 37(5):256–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Enge M,Arda HE,Mignardi M, Beausang J, Bottino R, et al. 2017. Single-cell analysis of human pancreas reveals transcriptional signatures of aging and somatic mutation patterns. Cell 171(2):321–330.e14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Eriksson PS, Perfilieva E, Björk-Eriksson T, Alborn AM, Nordborg C, et al. 1998. Neurogenesis in the adult human hippocampus. Nat. Med 4(11):1313–17 [DOI] [PubMed] [Google Scholar]
  • 30.Fang EF, Lautrup S, Hou Y, Demarest TG, Croteau DL, et al. 2017. NAD+ in aging: molecular mechanisms and translational implications. Trends Mol. Med. 23(10):899–916 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fang EF, Scheibye-Knudsen M, Chua KF, Mattson MP, Croteau DL, Bohr VA. 2016. Nuclear DNA damage signalling to mitochondria in ageing. Nat. Rev. Mol. Cell Biol. 17(5):308–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Freibaum BD, Lu Y, Lopez-Gonzalez R, Kim NC, Almeida S, et al. 2015. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 525(7567):129–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Frobel J, Hemeda H, Lenz M, Abagnale G, Joussen S,et al. 2014. Epigenetic rejuvenation of mesenchymal stromal cells derived from induced pluripotent stem cells. Stem Cell Rep. 3(3):414–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fumagalli M, Rossiello F, Clerici M, Barozzi S, Cittaro D, et al. 2012. Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat. Cell Biol. 14(4):355–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gladyshev TV, Gladyshev VN. 2016A disease or not a disease? Aging as a pathology. Trends Mol. Med. 22(12):995–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gladyshev VN. 2016. Aging: progressive decline in fitness due to the rising deleteriome adjusted by genetic, environmental, and stochastic processes. Aging Cell 15(4):594–602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Gonzalo S, Kreienkamp R. 2015. DNA repair defects and genome instability in Hutchinson–Gilford Progeria Syndrome. Curr. Opin. Cell Biol. 34:75–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Graf T, Enver T 2009. Forcing cells to change lineages. Nature 462(7273):587–94 [DOI] [PubMed] [Google Scholar]
  • 39.Grima JC, Daigle JG, Arbez N, Cunningham KC, Zhang K, et al. 2017. Mutant Huntingtin disrupts the nuclear pore complex. Neuron 94(1):93–107.e6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hanawalt PC, Spivak G.2008. Transcription-coupled DNA repair: two decades of progress and surprises. Nat. Rev. Mol. Cell Biol. 9(12):958–70 [DOI] [PubMed] [Google Scholar]
  • 41.Hansson J, Rafiee MR, Reiland S, Polo JM, Gehring J, et al. 2012. Highly coordinated proteome dynamics during reprogramming of somatic cells to pluripotency. Cell Rep. 2(6):1579–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Harries LW, Hernandez D, Henley W, Wood AR, Holly AC, et al. 2011. Human aging is characterized by focused changes in gene expression and deregulation of alternative splicing. Aging Cell 10(5):868–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Heinrich C, Bergami M, Gascón S, Lepier A, Viganò F, et al. 2014. Sox2-mediated conversion of NG2 glia into induced neurons in the injured adult cerebral cortex. Stem Cell Rep. 3(6):1000–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hernández-Vega A, Braun M, Scharrel L, Jahnel M, Wegmann S, et al. 2017. Local nucleation of microtubule bundles through tubulin concentration into a condensed tau phase. Cell Rep. 20(10):2304–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Hoelz A, Debler EW, Blobel G. 2011. The structure of the nuclear pore complex. Annu. Rev. Biochem 80:613–43 [DOI] [PubMed] [Google Scholar]
  • 46.Horvath S 2013. DNA methylation age of human tissues and cell types. Genome Biol. 14(10):R115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hu B-Y, Du Z-W, Zhang S-C. 2009. Differentiation of human oligodendrocytes from pluripotent stem cells. Nat. Protoc 4(11):1614–22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hu W, Qiu B, Guan W, Wang Q, Wang M, et al. 2015. Direct conversion of normal and Alzheimer’s disease human fibroblasts into neuronal cells by small molecules. Cell Stem Cell 17(2):204–12 [DOI] [PubMed] [Google Scholar]
  • 49.Huang P, Zhang L, Gao Y, He Z, Yao D, et al. 2014. Direct reprogramming of human fibroblasts to functional and expandable hepatocytes. Cell Stem Cell 14(3):370–84 [DOI] [PubMed] [Google Scholar]
  • 50.Huh CJ, Zhang B, Victor M, Dahiya S, Batista LF, et al. 2016. Maintenance of age in human neurons generated by microRNA-based neuronal conversion of fibroblasts. eLife 5:e18648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Ibarra A, Benner C, Tyagi S, Cool J, Hetzer MW. 2016. Nucleoporin-mediated regulation of cell identity genes. Genes Dev. 30(20):2253–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ieda M, Fu J-D, Delgado-Olguin P, Vedantham V, Hayashi Y, et al. 2010. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142(3):375–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Inak G, Lorenz C, Lisowski P, Zink A, Mlody B, Prigione A. 2017. Concise review: induced pluripotent stem cell-based drug discovery for mitochondrial disease. Stem Cells 35(7):1655–62 [DOI] [PubMed] [Google Scholar]
  • 54.Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, et al. 2012. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 482(7384):216–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jaarsma D, van der Pluijm I, de Waard MC, Haasdijk ED, Brandt R, et al. 2011. Age-related neuronal degeneration: complementary roles of nucleotide excision repair and transcription-coupled repair in preventing neuropathology. PLOS Genet. 7(12):e1002405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Jovičić A, Mertens J, Boeynaems S, Bogaert E, Chai N,et al. 2015. Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nat. Neurosci 18(9):1226–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jovičić A, Paul JW, Gitler AD. 2016. Nuclear transport dysfunction: a common theme in amyotrophic lateral sclerosis and frontotemporal dementia. J. Neurochem 138(S1):134–44 [DOI] [PubMed] [Google Scholar]
  • 58.Kang E, Wang X, Tippner-Hedges R, Ma H, Folmes CDL, et al. 2016. Age-related accumulation of somatic mitochondrial DNA mutations in adult-derived human iPSCs. Cell Stem Cell 18(5):625–36 [DOI] [PubMed] [Google Scholar]
  • 59.Karch CM, Goate AM. 2015. Alzheimer’s disease risk genes and mechanisms of disease pathogenesis. Biol. Psychiatry 77(1):43–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Karow M, Sánchez R, Schichor C, Masserdotti G, Ortega F, et al. 2012. Reprogramming of pericyte-derived cells of the adult human brain into induced neuronal cells. Cell Stem Cell 11(4):471–76 [DOI] [PubMed] [Google Scholar]
  • 61.Kim D, Kim C-H, Moon J-I, Chung Y-G, Chang M-Y, et al. 2009. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell 4(6):472–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kim HJ, Taylor JP. 2017. Lost in transportation: nucleocytoplasmic transport defects in ALS and other neurodegenerative diseases. Neuron 96(2):285–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kim Y, Zheng X, Ansari Z, Bunnell MC, Herdy JR, et al. 2018. Mitochondrial aging defects emerge in directly reprogrammed human neurons due to their metabolic profile. Cell Rep. 23(9):2550–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Koch P, Breuer P, Peitz M, Jungverdorben J, Kesavan J, et al. 2011. Excitation-induced ataxin-3 aggregation in neurons from patients with Machado-Joseph disease. Nature 480:543–46 [DOI] [PubMed] [Google Scholar]
  • 65.Koch P, Optiz T, Steinbeck JA, Ladewig J, Brüstle O. 2009. A rosette-type, self-renewing human ES cell-derived neural stem cell with potential for in vitro instruction and synaptic integration. PNAS 106:3225–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Koch P, Tamboli IY, Mertens J, Wunderlich P, Ladewig J, et al. 2012. Presenilin-1 L166P mutant human pluripotent stem cell-derived neurons exhibit partial loss of γ-secretase activity in endogenous amyloid-β generation. Am. J. Pathol. 180(6):2404–16 [DOI] [PubMed] [Google Scholar]
  • 67.Krencik R, Weick JP, Liu Y, Zhang Z-J, Zhang S-C. 2011. Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nat. Biotechnol 29(6):528–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kriks S, Shim J-W, Piao J, Ganat YM, Wakeman DR, et al. 2011. Dopamine neurons derived from human ES cells efficiently engraft in animal models of Parkinson’s disease. Nature 480:547–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ladewig J, Mertens J, Kesavan J, Doerr J, Poppe D, et al. 2012. Small molecules enable highly efficient neuronal conversion of human fibroblasts. Nat. Methods 9:575–78 [DOI] [PubMed] [Google Scholar]
  • 70.Laiosa CV, Stadtfeld M, Xie H, de Andres-Aguayo L, Graf T. 2006. Reprogramming of committed T cell progenitors to macrophages and dendritic cells by C/EBP α and PU.1 transcription factors. Immunity 25(5):731–44 [DOI] [PubMed] [Google Scholar]
  • 71.Lancaster MA, Knoblich JA. 2014. Organogenesis in a dish: modeling development and disease using organoid technologies. Science 345(6194):1247125. [DOI] [PubMed] [Google Scholar]
  • 72.Lapasset L, Milhavet O, Prieur A, Besnard E, Babled A, et al. 2011. Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state. Genes Dev. 25(21):2248–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lau S, Rylander Ottosson D, Jakobsson J, Parmar M. 2014. Direct neural conversion from human fibroblasts using self-regulating and nonintegrating viral vectors. Cell Rep. 9(5):1673–80 [DOI] [PubMed] [Google Scholar]
  • 74.Li X, Zuo X, Jing J, Ma Y, Wang J, et al. 2015. Small-molecule-driven direct reprogramming of mouse fibroblasts into functional neurons. Cell Stem Cell 17(2):195–203 [DOI] [PubMed] [Google Scholar]
  • 75.Lindahl T, Barnes DE. 2000. Repair of endogenous DNA damage. Cold Spring Harb. Symp. Quant. Biol. 65:127–33 [DOI] [PubMed] [Google Scholar]
  • 76.Liu H, Zhang S-C. 2011. Specification of neuronal and glial subtypes from human pluripotent stem cells. Cell. Mol. Life Sci. 68(24):3995–4008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Liu M-L, Zang T, Zhang C-L. 2016. Direct lineage reprogramming reveals disease-specific phenotypes of motor neurons from human ALS patients. Cell Rep. 14(1):115–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Liu M-L, Zang T, Zou Y, Chang JC, Gibson JR, et al. 2013. Small molecules enable neurogenin 2 to efficiently convert human fibroblasts into cholinergic neurons. Nat. Commun 4:2183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lo Sardo V, Ferguson W, Erikson GA, Topol EJ, Baldwin KK, Torkamani A. 2016. Influence of donor age on induced pluripotent stem cells. Nat. Biotechnol 35(1):69–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Lodato MA, Rodin RE, Bohrson CL, Coulter ME, Barton AR, et al. 2017. Aging and neurodegeneration are associated with increased mutations in single human neurons. Science 359:555–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. 2013. The hallmarks of aging. Cell 153(6):1194–217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lu T, Pan Y, Kao S-Y, Li C, Kohane I, et al. 2004. Gene regulation and DNA damage in the ageing human brain. Nature 429(6994):883–91 [DOI] [PubMed] [Google Scholar]
  • 83.Madabhushi R, Gao F, Pfenning AR, Pan L, Yamakawa S, et al. 2015. Activity-induced DNA breaks govern the expression of neuronal early-response genes. Cell 161(7):1592–605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Marion RM, Strati K, Li H, Tejera A, Schoeftner S, et al. 2009. Telomeres acquire embryonic stem cell characteristics in induced pluripotent stem cells. Cell Stem Cell 4(2):141–54 [DOI] [PubMed] [Google Scholar]
  • 85.Marro S, Pang ZP, Yang N, Tsai M-C, Qu K, et al. 2011. Direct lineage conversion of terminally differentiated hepatocytes to functional neurons. Cell Stem Cell 9(4):374–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Mattis VB, Svendsen SP, Ebert A, Svendsen CN, King AR, et al. (HD iPSC Consort.) 2012. Induced pluripotent stem cells from patients with Huntington’s disease show CAG-repeat-expansion-associated phenotypes. Cell Stem Cell 11(2):264–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.McConnell MJ, Lindberg MR, Brennand KJ, Piper JC, Voet T, et al. 2013. Mosaic copy number variation in human neurons. Science 342(6158):632–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.McKinnon PJ. 2017. Genome integrity and disease prevention in the nervous system. Genes Dev. 31(12):1180–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Meissner A, Mikkelsen TS, Gu H, Wernig M, Hanna J, et al. 2008. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 454(7205):766–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Mertens J, Marchetto MC, Bardy C, Gage FH. 2016. Evaluating cell reprogramming, differentiation and conversion technologies in neuroscience. Nat. Rev. Neurosci 17(7):424–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Mertens J, Paquola ACM, Ku M, Hatch E, Böhnke L, et al. 2015. Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects. Cell Stem Cell 17(6):705–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Mertens J, Stüber K, Poppe D, Doerr J, Ladewig J, et al. 2013. Embryonic stem cell-based modeling of tau pathology in human neurons. Am. J. Pathol 182(5):1769–79 [DOI] [PubMed] [Google Scholar]
  • 93.Mertens J, Stüber K, Wunderlich P, Ladewig J, Kesavan JC, et al. 2013. APP processing in human pluripotent stem cell-derived neurons is resistant to NSAID-based γ-secretase modulation. Stem Cell Rep. 1:491–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Miller JD, Ganat YM, Kishinevsky S, Bowman RL, Liu B, et al. 2013. Human iPSC-based modeling of late-onset disease via progerin-induced aging. Cell Stem Cell 13(6):691–705 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Mong J, Panman L, Alekseenko Z, Kee N, Stanton LW, et al. 2014. Transcription factor-induced lineage programming of noradrenaline and motor neurons from embryonic stem cells. Stem Cells 32(3):609–22 [DOI] [PubMed] [Google Scholar]
  • 96.Murry CE, Keller G. 2008. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 132(4):661–80 [DOI] [PubMed] [Google Scholar]
  • 97.Nekrasov ED, Vigont VA, Klyushnikov SA, Lebedeva OS, Vassina EM, et al. 2016. Manifestation of Huntington’s disease pathology in human induced pluripotent stem cell-derived neurons. Mol. Neurodegener. 11:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Nguyen HN, Byers B, Cord B, Shcheglovitov A, Byrne J, et al. 2011. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 8(3):267–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Nicholas CR, Chen J, Tang Y, Southwell DG, Chalmers N, et al. 2013. Functional maturation of hPSC-derived forebrain interneurons requires an extended timeline and mimics human neural development. Cell Stem Cell 12(5):573–86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Nishimura T, Kaneko S, Kawana-Tachikawa A, Tajima Y, Goto H,et al. 2013. Generation of rejuvenated antigen-specific T cells by reprogramming to pluripotency and redifferentiation. Cell Stem Cell 12(1):114–26 [DOI] [PubMed] [Google Scholar]
  • 101.Ocampo A, Reddy P, Martinez-Redondo P, Platero-Luengo A, Hatanaka F, et al. 2016. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 167:1719–33.e12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Okita K, Yamanaka S. 2011. Induced pluripotent stem cells: opportunities and challenges. Philos. Trans. R. Soc. B 366(1575):2198–207 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pang ZP, Yang N, Vierbuchen T, Ostermeier A, Fuentes DR, et al. 2011. Induction of human neuronal cells by defined transcription factors. Nature 476:220–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Deleted in proof
  • 105.Pereira M, Pfisterer U, Rylander D, Torper O, Lau S, et al. 2014. Highly efficient generation of induced neurons from human fibroblasts that survive transplantation into the adult rat brain. Sci. Rep 4(1):6330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Pfisterer U, Kirkeby A, Torper O, Wood J, Nelander J, et al. 2011. Direct conversion of human fibroblasts to dopaminergic neurons. PNAS 108(25):10343–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Prigione A, Hossini AM, Lichtner B, Serin A, Fauler B,et al. 2011. Mitochondrial-associated cell death mechanisms are reset to an embryonic-like state in aged donor-derived iPS cells harboring chromosomal aberrations. PLOS ONE 6(11):e27352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Puzzo D, Gulisano W, Palmeri A, Arancio O. 2015. Rodent models for Alzheimer’s disease drug discovery. Expert Opin. Drug Discov. 10(7):703–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Rakovic A, Shurkewitsch K, Seibler P, Grünewald A, Zanon A, et al. 2013. Phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1)-dependent ubiquitination of endogenous Parkin attenuates mitophagy: study in human primary fibroblasts and induced pluripotent stem cell-derived neurons. J. Biol. Chem 288(4):2223–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Rando TA, Chang HY. 2012. Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell 148:46–57 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Reinhardt P, Schmid B, Burbulla LF, Schöndorf DC, Wagner L, et al. 2013. Genetic correction of a LRRK2 mutation in human iPSCs links Parkinsonian neurodegeneration to ERK-dependent changes in gene expression. Cell Stem Cell 12(3):354–67 [DOI] [PubMed] [Google Scholar]
  • 112.Rivetti di Val Cervo P, Romanov RA, Spigolon G, Masini D, Martín-Montañez E, et al. 2017. Induction of functional dopamine neurons from human astrocytes in vitro and mouse astrocytes in a Parkinson’s disease model. Nat. Biotechnol 35(5):444–52 [DOI] [PubMed] [Google Scholar]
  • 113.Savas JN, Toyama BH, Xu T, Yates JR, Hetzer MW. 2012. Extremely long-lived nuclear pore proteins in the rat brain. Science 335(6071):942–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Scheibye-Knudsen M, Mitchell SJ, Fang EF, Iyama T, Ward T,et al. 2014. A high-fat diet and NAD+ activate Sirt1 to rescue premature aging in Cockayne syndrome. Cell Metab. 20(5):840–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Scheibye-Knudsen M, Tseng A, Borch Jensen M, Scheibye-Alsing K, Fang EF, et al. 2016. Cockayne syndrome group A and B proteins converge on transcription-linked resolution of non-B DNA. PNAS 113(44):12502–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Seibler P, Graziotto J, Jeong H, Simunovic F, Klein C, Krainc D. 2011. Mitochondrial Parkin recruitment is impaired in neurons derived from mutant PINK1 induced pluripotent stem cells. J. Neurosci 31(16):5970–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Sepe S, Milanese C, Gabriels S, Derks KWJ, Payan-Gomez C, et al. 2016. Inefficient DNA repair is an aging-related modifier of Parkinson’s disease. Cell Rep. 15(9):1866–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Shi KY, Mori E, Nizami ZF, Lin Y, Kato M, et al. 2017. Toxic PRn poly-dipeptides encoded by the C9orf72 repeat expansion block nuclear import and export. PNAS 114(7):E1111–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Smith DK, Yang J, Liu M-L, Zhang C-L. 2016. Small molecules modulate chromatin accessibility to promote NEUROG2-mediated fibroblast-to-neuron reprogramming. Stem Cell Rep. 7:955–69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Son EY, Ichida JK, Wainger BJ, Toma JS, Rafuse VF, et al. 2011. Conversion of mouse and human fibroblasts into functional spinal motor neurons. Cell Stem Cell 9:205–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Studer L, Vera E, Cornacchia D. 2015. Programming and reprogramming cellular age in the era of induced pluripotency. Cell Stem Cell 16(6):591–600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Suberbielle E, Sanchez PE, Kravitz AV, Wang X, Ho K, et al. 2013. Physiologic brain activity causes DNA double-strand breaks in neurons, with exacerbation by amyloid-β. Nat. Neurosci 16(5):613–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Sugiura M, Kasama Y, Araki R, Hoki Y, Sunayama M, et al. 2014. Induced pluripotent stem cell generation-associated point mutations arise during the initial stages of the conversion of these cells. Stem Cell Rep. 2(1):52–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Suhr ST, Chang E-A, Rodriguez RM, Wang K, Ross PJ, et al. 2009. Telomere dynamics in human cells reprogrammed to pluripotency. PLOS ONE 4(12):e8124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Suhr ST, Chang E-A, Tjong J, Alcasid N, Perkins GA, et al. 2010. Mitochondrial rejuvenation after induced pluripotency. PLOS ONE 5(11):e14095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Tabar V, Studer L.2014. Pluripotent stem cells in regenerative medicine: challenges and recent progress. Nat. Rev. Genet 15(2):82–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, et al. 2007. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131(5):861–72 [DOI] [PubMed] [Google Scholar]
  • 128.Tang Y, Liu M-L, Zang T, Zhang C-L. 2017. Direct reprogramming rather than iPSC-based reprogramming maintains aging hallmarks in human motor neurons. Front. Mol. Neurosci 10:359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Teschendorff AE, West J, Beck S. 2013. Age-associated epigenetic drift: implications, and a case of epigenetic thrift? Hum. Mol. Genet 22:R7–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Theka I, Caiazzo M, Dvoretskova E, Leo D, Ungaro F, et al. 2013. Rapid generation of functional dopaminergic neurons from human induced pluripotent stem cells through a single-step procedure using cell lineage transcription factors. Stem Cells Transl. Med. 2(6):473–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Their M, Wörsdörfer P, Lakes YB, Gorris R, Herms S, et al. 2012. Direct conversion of fibroblasts into stably expandable neural stem cells. Cell Stem Cell 10(4):473–79 [DOI] [PubMed] [Google Scholar]
  • 132.Thoma EC,Wischmeyer E, Offen N,Maurus K, Sirén A-L, et al. 2012. Ectopic expression of neurogenin 2 alone is sufficient to induce differentiation of embryonic stem cells into mature neurons. PLOS ONE 7(6):e38651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Toda T, Hsu JY, Linker SB, Hu L, Schafer ST, et al. 2017. Nup153 interacts with Sox2 to enable bimodal gene regulation and maintenance of neural progenitor cells. Cell Stem Cell 21:618–634.e7 [DOI] [PubMed] [Google Scholar]
  • 134.Torper O, Pfisterer U, Wolf DA, Pereira M, Lau S,et al. 2013. Generation of induced neurons via direct conversion in vivo. PNAS 110(17):7038–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Toyama BH, Hetzer MW. 2013. Protein homeostasis: live long, won’t prosper. Nat. Rev. Mol. Cell Biol. 14(1):55–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Toyama BH, Savas JN, Park SK, Harris MS, Ingolia NT, et al. 2013. Identification of long-lived proteins reveals exceptional stability of essential cellular structures. Cell 154(5):971–82 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Treutlein B, Lee QY, Camp JG, Mall M, Koh W, et al. 2016. Dissecting direct reprogramming from fibroblast to neuron using single-cell RNA-seq. Nature 534(7607):391–95 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Vadodaria KC, Marchetto MC, Mertens J, Gage FH. 2016. Generating human serotonergic neurons in vitro: methodological advances. BioEssays 38(11):1123–29 [DOI] [PubMed] [Google Scholar]
  • 139.Vadodaria KC, Mertens J, Paquola A, Bardy C, Li X, et al. 2016. Generation of functional human serotonergic neurons from fibroblasts. Mol. Psychiatry 21(1):49–61 [DOI] [PubMed] [Google Scholar]
  • 140.Vera E, Bosco N, Studer L. 2016. Generating late-onset human iPSC-based disease models by inducing neuronal age-related phenotypes through telomerase manipulation. Cell Rep. 17(4):1184–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Vermeij WP, Dollé MET, Reiling E, Jaarsma D, Payan-Gomez C, et al. 2016. Restricted diet delays accelerated ageing and genomic stress in DNA-repair-deficient mice. Nature 537(7620):427–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Victor MB, Richner M, Hermanstyne TO, Ransdell JL, Sobieski C, et al. 2014. Generation of human striatal neurons by microRNA-dependent direct conversion of fibroblasts. Neuron 84(2):311–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Victor MB, Richner M, Olsen HE, Lee SW, Monteys AM, et al. 2018. Striatal neurons directly converted from Huntington’s disease patient fibroblasts recapitulate age-associated disease phenotypes. Nat. Neurosci 21:341–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Südhof TC, Wernig M. 2010. Direct conversion of fibroblasts to functional neurons by defined factors. Nature 463(7284):1035–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Wapinski OL, Lee QY, Chen AC, Li R, Corces MR, et al. 2017. Rapid chromatin switch in the direct reprogramming of fibroblasts to neurons. Cell Rep. 20(13):3236–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Wapinski OL, Vierbuchen T, Qu K, Lee QY, Chanda S, et al. 2013. Hierarchical mechanisms for direct reprogramming of fibroblasts to neurons. Cell 155(3):621–35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Warren L, Manos PD, Ahfeldt T, Loh Y-H, Li H, et al. 2010. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7(5):618–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Woodruff G, Young JE, Martinez FJ, Buen F, Gore A, et al. 2013. The presenilin-1 ΔE9 mutation results in reduced γ-secretase activity, but not total loss of PS1 function, in isogenic human stem cells. Cell Rep. 5(4):974–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Xu Z, Jiang H, Zhong P, Yan Z, Chen S, Feng J. 2016. Direct conversion of human fibroblasts to induced serotonergic neurons. Mol. Psychiatry 21(1):62–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Xue Y, Ouyang K, Huang J, Zhou Y, Ouyang H, et al. 2013. Direct conversion of fibroblasts to neurons by reprogramming PTB-regulated microRNA circuits. Cell 152:82–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, et al. 2011. Modeling familial Alzheimer’s disease with induced pluripotent stem cells. Hum. Mol. Genet 20(23):4530–39 [DOI] [PubMed] [Google Scholar]
  • 152.Yang N, Chanda S, Marro S, Ng YH, Janas JA, et al. 2017. Generation of pure GABAergic neurons by transcription factor programming. Nat. Methods 14(6):621–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Yang N, Zuchero JB, Ahlenius H, Marro S, Ng YH, et al. 2013. Generation of oligodendroglial cells by direct lineage conversion. Nat. Biotechnol 31(5):434–39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Yang Y, Jiao J, Gao R, Le R, Kou X, et al. 2015. Enhanced rejuvenation in induced pluripotent stem cell-derived neurons compared with directly converted neurons from an aged mouse. Stem Cells Dev. 24(23):2767–77 [DOI] [PubMed] [Google Scholar]
  • 155.Yang Y, Jiao J, Gao R, Yao H, Sun X-F, Gao S. 2013. Direct conversion of adipocyte progenitors into functional neurons. Cell. Reprogramming 15(6):484–89 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, et al. 2011. MicroRNA-mediated conversion of human fibroblasts to neurons. Nature 476(7359):228–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Zhang K, Donnelly CJ, Haeusler AR, Grima JC, Machamer JB, et al. 2015. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature 525(7567):56–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zhang K, Liu G-H, Yi F, Montserrat N, Hishida T, et al. 2014. Direct conversion of human fibroblasts into retinal pigment epithelium-like cells by defined factors. Protein Cell 5(1):48–58 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Zeng H, Guo M, Martins-Taylor K, Wang X, Zhang Z, et al. 2010. Specification of region-specific neurons including forebrain glutamatergic neurons from human induced pluripotent stem cells. PLOS ONE 5(7):e11853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Zhao J, He H, Zhou K, Ren Y, Shi Z, et al. 2012. Neuronal transcription factors induce conversion of human glioma cells to neurons and inhibit tumorigenesis. PLOS ONE 7(7):e41506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Zheng SC, Widschwendter M, Teschendorff AE. 2016. Epigenetic drift, epigenetic clocks and cancer risk. Epigenomics 8(5):705–19 [DOI] [PubMed] [Google Scholar]
  • 162.Zhou T, Benda C, Duzinger S, Huang Y, Li X, et al. 2011. Generation of induced pluripotent stem cells from urine. J. Am. Soc. Nephrol 22(7):1221–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Zhu S, Russ HA, Wang X, Zhang M, Ma T, et al. 2016. Human pancreatic beta-like cells converted from fibroblasts. Nat. Commun 7:10080. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES