前神经细胞重编程:方法、挑战与前景

封面


如何引用文章

全文:

开放存取 开放存取
受限制的访问 ##reader.subscriptionAccessGranted##
受限制的访问 订阅或者付费存取

详细

近几十年来,开发在in vitro、ex vivo和in vivo条件下对不同分化程度细胞进行重编程的技术手段,已成为生命科学领域的重要突破之一。近年来,关于通过直接重编程将分化细胞转化为特化神经元的实验数据逐年增加,包括在in vivo条件下由胶质细胞生成神经元的研究成果。未来,这类技术有望将再生医学的潜力提升到一个全新水平。尽管我们对细胞分化和表型可塑性机制的理解日益深入,且调控这些过程的手段不断丰富,但细胞重编程在临床中的实际应用仍面临诸多挑战。本文综述了细胞可塑性的概念,介绍了前神经细胞重编程的最新研究进展,包括直接和间接重编程方法,并分析了阻碍其转化为临床实践的主要难点。

全文:

INTRODUCTION

Recent advances in the technology and methods for reprogramming of cells of various differentiation stages represent one of the most important scientific and technological achievements of the last decades. Each year, an increasing number of experimental papers describe direct cellular reprogramming in vitro, ex vivo, and in vivo [1–4]. In the future, such technologies will potentially reach a higher level of regenerative medicine including neuroregeneration. Global trends toward lifespan extension and a growing population of elder people are leading to an increased incidence and prevalence of age-related diseases, including neurodegenerative diseases such as Alzheimer disease for which there is no effective cure so far [5]. So, many researchers and investors consider replacement of dysfunctional neurons by in situ reprogramming or by intracranial delivery of in vitro reprogrammed cells to be an extremely promising technology. The potential of using proneural reprogramming, e.g., for glial cells, is being evaluated in experimental models of traumatic spinal cord and brain injury [6, 7], stroke [8], retinal neuron regeneration [9, 10], Parkinson disease [11], and Huntington disease [12].

However, the rapid and widespread practical application of this technology remains an unrealistic task, despite continuously growing understanding of the mechanisms of cell differentiation and phenotypic plasticity, and the emergence of novel technological capabilities for cellular reprogramming, including in situ.

The aim of this review was to summarize the results of recent studies regarding direct and indirect proneural reprogramming and to elucidate the barriers to clinical implementation of these technologies.

DEVELOPMENT OF THE CENTRAL NERVOUS SYSTEM AT DIFFERENT ONTOGENETIC STAGES

The development of the nervous system, or neurulation, begins with the formation of the gastrula at the beginning of embryogenesis. The neural plate formed by a thickened portion of the ectoderm endures a ventromedial turnover to form neural folds and a groove that constitutes the neural tube during neurulation. Further differentiation of the neural tube leads to the development of the brain and spinal cord. The neural tube is formed from the neuroectoderm, so all multipotent progenitor cells in the central nervous system (CNS) are neuroepithelial or neural stem cells (NSCs) [13]. They can divide in two ways. Symmetrical division is needed to form a pool of NSCs, whereas asymmetrical one yields cells that retain NSC properties and postmitotic neuronal progenitor cells which cease to divide any longer [14]. Subsequently, most NSCs differentiate into radial glial cells (RGCs). RGCs are characterized by high expression of specific transcription factors: Pax6 (Paired box gene family), a tissue-specific coordinator of embryonic development of the nervous system and eyes; Sox2 (SRY-related HMG box family), one of the major regulators of pluripotency maintenance in stem cells; Ascl1/Mash1 (Achaete-scute family bHLH transcription factor 1/Mammalian achaete-scute homolog 1); Dlx (Distal-less homeobox genes) and NEUROD (neurogenic differentiation factors) gene families encoding proteins that regulate neuronal migration and differentiation. Cells expressing RGC markers [15] appear at days 8–10 of embryogenesis in rats [14] and at weeks 5–8 of prenatal development in humans [16]. The bodies of these cells are predominantly located in brain regions formed during forebrain development: subventricular zone (SVZ) in the lateral walls of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus [17]. RGCs can also divide symmetrically and asymmetrically to develop into intermediate progenitor cells (IPCs). They are the main source of all major CNS cells such as neurons, astrocytes, and oligodendrocytes, as well as highly specialized populations such as Müller cells which are the most abundant glial cells in the retina [18] and Bergmann gliocytes in the cerebellum [19]. In contrast to neurons, glial cells do not generate action potentials, but perform essential support functions. They regulate synaptogenesis, maintain synaptic plasticity and myelination of nerve fibers; for rapid conduction of nerve impulses, produce neuroprotective compounds and factors vital for proliferation, survival, migration and differentiation of neurons, provide neuronal trophism and brain tissue regeneration [20]. IPCs continuously migrate to the cortex during the embryogenesis to secure the formation of the neocortical histoarchitecture and the interneuron network. By the end of embryonic development, most RGCs not yet differentiated leave the ventricular zone and migrate to the cortex where they differentiate to astrocytes [13] (Fig. 1).

 

Fig. 1. Schematic representation of embryonic brain development. Neural stem cells (NSCs) in the subventricular zone (SVZ) divide symmetrically, generating a pool of radial glial cells (RGCs). Through both symmetric and asymmetric division, RGCs give rise to intermediate progenitor cells: neuronal intermediate progenitor cells (nIPCs), oligodendrocyte intermediate progenitor cells (oIPCs), and astrocyte intermediate progenitor cells (aIPCs). As differentiation progresses, these cells migrate from the neurogenic SVZ, contributing to neocortical histoarchitecture (indicated by the red dashed arrow) and maturing into neurons, oligodendrocytes, and astrocytes. During the earliest stages of embryogenesis, a niche of quiescent NSCs and RGCs (qNSCs/qRGCs) is established, which contribute to neurogenesis in the adult brain.

Рис. 1. Схема эмбрионального развития головного мозга. Стволовые нейроэпителиальные клетки (NSCs) в субвентрикулярной зоне (SVZ) делятся симметрично и формируют пул клеток радиальной глии (RGCs). В результате симметричного и асимметричного деления RGCs образуются промежуточные клетки-предшественники — нейрональные (Neuronal intermediate progenitor cells, nIPCs), олигодендроцитарные (Oligodendrocyte intermediate progenitor cells, oIPCs) и астроцитарные (Astrocyte intermediate progenitor cells, aIPCs). По мере дифференцировки клетки мигрируют из нейрогенной области SVZ, формируют гистоархитектонику неокортекса (обозначено красной пунктирной стрелкой) и становятся зрелыми нейронами, олигодендроцитами и астроцитами. На самых ранних этапах эмбриогенеза формируется ниша покоящихся NSCs и RGCs (Quiescent NSCs/RGCs, qNSCs/qRGCs), за счёт которых происходит нейрогенез во взрослом головном мозге.

 

However, the SVZ and SGZ remain and secure the pool of quiescent RGCs and NSCs [21]. These populations of multipotent cells maintain neurogenesis, the process of mature neurons differentiation from progenitor cells throughout the entire postnatal ontogenesis [22].

Neurogenesis in the fully formed brain is required to regenerate olfactory cells. They are sensory neurons located in the olfactory region of the nasal mucosa and first ones of the cranial nerve I [23]. Neurogenesis is vital for neuroplasticity maintenance, replacing neurons in the structures responsible for memory and other cognitive processes [24]. In adult rodents, the SVZ is primarily responsible for the formation of olfactory interneurons in the olfactory bulb (OB). It was also reported that small populations of neuroblasts might be able to migrate to the prefrontal cortex and striatum [25]. However, the canonical rostral SVZ-OB migratory stream and the less studied medial migratory stream directing neuroblasts to the prefrontal cortex appear to exist only for the short neonatal period in humans [26]. The SGZ is responsible for hippocampal neuroregeneration [22]. SGZ-derived neurons are involved in spatial and social memory functions in adults, provide the ability to distinguish similar events, and support stable encoding of information to consolidate short-term memory into long-term memory [27].

During postnatal neurogenesis, quiescent RGCs become activated under the influence of the surrounding glial cells and resume symmetric and asymmetric cell division to yield required progenitor cells. As they migrate to different parts of the brain, they differentiate into neuroblasts, then into immature neurons and finally into differentiated neurons. The spectrum of genes expressed in progenitor cells changes significantly throughout their development which make it possible to distinct the maturation stage of particular progenitor cells. However, it is not an easy task as multiple markers are common for different stages of progenitors’ development [28] (Fig. 2).

 

Fig. 2. Stages of neurogenesis in the adult brain of rodents and humans. Quiescent neural stem cells and radial glial cells (qNSCs/ qRGCs) become activated (aNSCs/aRGCs) in response to microenvironmental signals and resume asymmetric division. Unlike embryonic neurogenesis, postnatal neurogenesis does not involve the direct generation of new neurons from radial glial cells (RGCs). Instead, a sequential differentiation process occurs, giving rise to neuronal progenitor cells (NPCs), neuroblasts, immature neurons, and finally, functionally active, mature neurons that integrate into the existing neuronal network. The lower part of the figure (below the arrow) indicates key genes expressed at different stages of cell differentiation during neurogenesis.

Рис. 2. Этапы нейрогенеза во взрослом мозге грызунов и человека. Покоящиеся стволовые нейроэпителиальные клетки и клетки радиальной глии (qNSCs/qRGCs) под воздействием сигналов от микроокружения активируются (aNSCs/aRGCs) и возобновляют асимметричное деление. При постнатальном нейрогенезе новые нейроны не образуются напрямую из клеток радиальной глии (RGCs), что отличает его от эмбрионального нейрогенеза. Происходит последовательная дифференцировка в нейрональные клетки-предшественники (NPCs), нейробласты, незрелые и дифференцированные нейроны, которые функционально активны и способны встраиваться в существующую нейронную сеть. В нижней части рисунка, под стрелкой, обозначены основные гены, экспрессирующиеся на разных стадиях дифференцировки клеток при нейрогенезе.

 

REGENERATIVE POTENTIAL OF THE ADULT BRAIN

Being multipotent cells, RGCs simultaneously express markers of various differentiation stages of progenitors—PAX6, DLX, ASCL1, and NEUROD1— and mature astrocyte-specific proteins, including glial fibrillary acidic protein (GFAP), astrocytic glutamate transporter (GLAST), and brain lipid binding protein (BLBP) [29]. In adult mice, neurogenesis is regulated by Pax6, Dlx, Ascl1 and Neurod1 genes just as in embryogenesis [30]. However, as RGCs divide, neurons are formed both as primary cells and from progenitor cells during embryonic development. In the postnatal brain, new neurons might only be formed from intermediate progenitors and due to the secretory context of the mature glia microenvironment, primarily astrocytes and ependymocytes [31]. As a result, RGCs are prone to gliogenesis rather than neurogenesis. The formation of new functional neurons from these progenitor cells, require the much higher expression of proneural factors (Pax6, Dlx, Ascl1, Neurod1, etc.) than it is needed for gliogenesis. Expression of proneural factors, as well as the transition of progenitor cells from a quiescent to an activated state, are strictly regulated processes. These physiological mechanisms presumably occur to prevent accelerated exhaustion of the NSC and RGC pool in the neurogenic niches. At the same time, a depletion of the pool of stem and progenitor cells in neurogenic zones progresses with age and negatively regulates neurogenesis. The fewer quiescent NSCs and RGCs remain, the lower the potential of their activation and the efficiency of differentiation into mature neurons [32]. Kalamakis et al. [33] showed that the number of NSCs in the SVZ in C57Bl/6-TLX-CreERT2YFP mice, in which NSC marker proteins was visualized in vivo via fluorescent imaging was significantly reduced in 7-month-old (m.o.) mice compared to 2 m.o. ones. There also was no differences found between the animals of 7 m.o. and 22 m.o. groups. Similar changes were observed in the subpopulation of activated NSCs, which were stimulated to resume the division for further neurogenesis initiation. Their number was significantly reduced at 7 m.o. compared to one of 2 m.o. mice [33]. Inflammaging, the age-related pro-inflammatory background plays a critical role in suppressing progenitor cell activation as well. In this study, it was found that the pro-inflammatory cytokines IL-33 and IFN-γ along with the chemokine CXCL10, whose upregulated expression is often observed with aging, inhibit the Wnt-dependent pathway, which is one of the main mechanisms maintaining neurogenesis processes [33].

Notably, that the new neurons of the SVZ and SGZ origin are markedly different despite the similarity of neurogenesis processes in these neurogenic regions. In contrast to the SVZ, NSCs and RGCs in the dentate gyrus are located deep in the brain parenchyma and surrounded by mature neurons and glial cells. This layer is far from the ventricular walls and cerebrospinal fluid flow, but close to blood vessels [34, 35]. As mentioned above, in humans, new neurons are not likely to appear in the olfactory bulb at postnatal period. NSCs and RGCs in this region differentiate predominantely into oligodendrocytes [25]. The published data on the duration of hippocampal SGZ neurogenesis are controversial. Some researchers found differentiating immature neurons in children aged 7–13 years, but did not observe them in adults [36]. Others reported the presence of RGCs with neurogenic potential and progenitor cells of more differentiation stage in middle-aged and older subjects [37, 38]. However, it is quite reasonable to assume that neurogenesis in the SGZ continues effectively even at the late postnatal period due to the close proximity of this neurogenic niche of the SGZ to mature hippocampal structures. Differences in the data are likely depend on the spatial and secretory context of the microenvironment as well as its epigenetic changes that make progenitor cells prone to acquire a particular phenotype in different brain regions.

The neurogenic potential of the progenitor niche is maintained throughout the entire life. NSCs and RGCs proliferation and differentiation into neurons are strictly regulated to happen only when needed. Some non-genetic diseases and CNS injuries are associated with the death of a significant number of neurons at once which results in cognitive impairment, neurological deficits, and a consequent reduction of life quality and longevity. Such events occur instantly: traumatic injury, ischemic stroke caused by thrombosis and disruption of brain blood flow, and hemorrhagic stroke, which involves bleeding in the brain tissue, between its membranes, or within the ventricles [39]. In a mouse model of transient ischemic attack, it was showed that stroke might activate neurogenesis, migration, and differentiation of neuroblasts, and the treatment with the pro-inflammatory cytokine IL-1β, which enhances the acute inflammatory response, promoted the survival of new neurons in these mice [40]. In addition, the differentiation of progenitor cells into glia might activate angiogenesis processes, which are also required for neuroregeneration [41]. Activation of quiescent progenitor cells and initiation of their differentiation were also reported in traumatic brain injury, both in the SGZ and in single quiescent cells of the cerebral cortex, where they were apparently in a quiescent, immature state [42, 43]. The effectiveness of neurogenesis in traumatic brain injury and stroke of various origins depends on a variety of factors, including the location and volume of brain tissue damage, age of the patient, comorbidities, their monitoring and treatment. For example, the suppression of hippocampal neurogenesis was reported in neuropsychiatric disorders such as mixed anxiety-depressive disorders [44].

A decline in the number of functioning neurons in certain brain areas might be gradual and take a long time to become clinically apparent. This is primarily common in age-related neurodegenerative disorders such as Alzheimer disease and Parkinson disease. Manifesting as mild cognitive impairment, at the age of 60 years and older and gradually progresses, leading to dementia in such patients [45]. Neurodegenerative diseases that are more of the genetic origin include Huntington disease [46], frontotemporal dementia, and amyotrophic lateral sclerosis. However, age is also a risk factor for their clinical manifestation. The depletion of the pool of progenitor cells in the neurogenic niches and the reduced effectiveness of neurogenesis due to aging are important features of these diseases. However, the brain adapts to a gradual reduction in the number of properly functioning neurons over time. As a result, more progenitor cells have to differentiate into mature neurons to maintain stable cognitive function. However, this is prevented by the pro-inflammatory state of inflammaging, a higher activation threshold for quiescent NSCs and RGCs, and a lack of neurotrophic factors caused by aging and senescence of glial and resident immune cells such as astrocytes and microglia [38, 47, 48].

Therefore, despite the enormous adaptive regenerative capacity of the brain, a wide range of CNS disorders leads to the extensive neuronal loss that cannot be restored by the body’s own resources. However, it is likely that treatment approaches designed to stimulate current neurogenic potential or to replace lost neurons with new ones derived in vitro or in situ would have significant clinical efficacy.

CELLULAR REPROGRAMMING: DEFINITIONS AND MECHANISMS

Metaplasia is the process whereby differentiated cells and tissues acquire characteristic of other differentiated cells and tissues. It was described and discussed by Rudolf Virchow and his successors in the 19th century [49, 50]. In modern cell biology and histology, metaplasia is described as a potentially reversible change in the phenotype of differentiated epithelial tissue, i.e., the appearance of features physiologically typical of another type of epithelial tissue [51]. The corresponding intracellular processes are called transdifferentiation or cellular metaplasia [52]. Abnormal and usually adverse stimuli make differentiated cells adapt by changing their phenotype and so might initiate or accelerate metaplastic processes [53]. Such stimuli may include harmful external chemical agents: acids or bases that mediate pH changes [54]; tobacco or alcohol use [55–57]; elevated levels of hormones or hormone-like substances such as bisphenol A and its derivatives [58, 59]; viral infections; chronic inflammation; oxidative stress [60–62]. There is still no consensus on whether cells and tissues can revert to the normal type of differentiation for their location after the harmful stimulus is removed and how long adverse environmental conditions should persist to result in dysplasia and increase the risk of malignancy [63]. It is not possible to determine the exact prevalence or incidence of any type of tissue metaplasia in the general population because most population studies include only a few of the most common diseases. For example, intestinal metaplasia of the esophageal epithelium, also known as Barrett esophagus, often accompanies gastroesophageal reflux disease. Constant reflux of gastric acid into the esophagus initiates the replacement of the squamous epithelium of the esophagus to single-layered cylindrical epithelium. Previously, patients diagnosed with Barrett esophagus were thought to have a 30 to 125 times higher relative risk of esophageal adenocarcinoma than the general population with an average annual risk of malignancy of 0.5% (range: 0.1%–3.5%). However, subsequent studies estimated a lower annual risk of adenocarcinoma ranging from 0.12% to 0.18% [64]. Metaplastic lesions of various types in some locations may regress after the irritating stimuli cease to exist. However, in the case of Barrett esophagus epithelial cells remain a phenotype of prismatic intestinal epithelium and do not revert to squamous epithelial phenotype even with effective treatment [65].

In human tissues, metaplasia is mainly considered for several epithelial types and is categorized as squamous, intestinal, or acinar–ductal metaplasia [63]. The processes occurring in the human tissues are related to direct metaplasia, in which phenotypic changes in cells are not accompanied by the stage of differentiation decrease. Such phenotype shifts are the result of the activation and/or deactivation of genes in combinations that are not typical of a particular type of epithelium under physiological conditions. The same mechanisms underlie direct reprogramming in vivo, or so called induced metaplasia. The absence of dedifferentiation stage is a key difference between direct reprogramming and the approaches of induced pluripotent stem cells (iPSCs) yielding and further regulation of their differentiation to obtain cells with the desired phenotype [66]. In terms of their morphofunctional and molecular biological properties, iPSCs are closest to embryonic stem cells, which have the lowest degree of differentiation [67]. Most somatic cells can be reprogrammed into iPSCs, such as keratinocytes [68], melanocytes [69], adipose tissue-derived stem cells and adipocytes [67], neural progenitor cells (NPCs) [70], etc. However, fibroblasts are most commonly used to obtain human iPSC populations [71, 72]. Hundreds of protocols for the differentiation of iPSCs into various cell types have been developed and performed with varying efficacy. They are based on the overexpression of transcription factors that are only active in poorly differentiated cells, or the co-cultivation with proteins and small molecules typical of pluripotent cells. As a result, the processed cells lose their signs of differentiation and revert to the stem cell state. After that, the in vitro iPSCs undergo all differentiation stages that cells of the target phenotype do under physiological conditions in the body by regulating the expression of different genes or by culturing with specific proteins and low-molecular compounds typical of the microenvironment of the target cells.

Neurons can be obtained via both the direct reprogramming and dedifferentiation into iPSCs from non-neuroectodermal cells, such as fibroblasts and peripheral blood monocytes. Whether this process can be considered as an example of mesenchymal–epithelial transition remains a matter of discussion [73, 74]. Embryonic ecto-, meso-, or endodermal cytogenesis appears to be a limiting but not prohibitive factor for reprogramming into cells of another germ layer. This may be due to the loss of unique epigenetic properties by the cells subjected to controlled exposure during culture, more in iPSC obtaining and less in direct reprogramming protocols, especially at late passages (≥10) [75]. However, several papers reported that some epigenetic changes are still preserved, which might keep cells to be prone to more successful reprogramming within the tissue of origin [67, 76]. This also applies to neurons. As shown by Hargus et al. [77], iPSCs derived from fetal NSCs and dermal fibroblasts differ significantly in the levels of expression of neuronal markers. iPSCs derived from low-differentiated neuroectodermal cells were significantly closer to neurons in the spectrum of expressed genes, and demonstrated significantly higher survival rates after the intracranial injection into the cerebral cortex of mice [77].

Differentiation and proper function of cells, especially neurons, highly depends on the microenvironment, including the changes in its spatiotemporal and secretory context over time. It defines the unique epigenetic changes of cells in a particular type of differentiated tissue [78]. The loss of these modifications, the need to extract cells from the body for in vitro reprogramming, and the reinjection of the reprogramed cell population are factors that make iPSCs potentially less practical for widespread clinical use than direct in situ reprogramming methods [79]. Although the use of iPSCs is not yet brought in practice, studies are underway using extracellular vesicles isolated from iPSCs to stimulate neurogenesis or neuroregeneration [80]. However, this is a complicated technological task that has not yet been tested in a clinical setting.

PRONEURAL REPROGRAMMING: METHODOLOGY AND TECHNOLOGICAL APPROACHES

Viral vectors carrying plasmids with genes of classical OSKM reprogramming factors (Oct4, Sox2, Klf4 and с-Myc) in different combinations is one of the most effective and widely used techniques to induce the transition of differentiated cells to a pluripotent state. Lentiviral or retroviral delivery of plasmids is characterized by their integration into the cell genome, which potentially should secure stable expression of genes required for iPSC induction [66, 81]. However, such viral integration into the cell genome might lead to insertional mutagenesis and unwanted transgene reactivation in differentiated cells [82]. An alternative way is the use of non-integrating viral vectors such as adenovirus, adeno-associated virus (AAV), or Sendai paramyxovirus [83]. The development of malignant neoplasms was reported in only a few experimental studies with AVV vector-based gene delivery systems in newborn mice and in dogs with hemophilia. The risk of insertional mutagenesis can be minimized by improving reprogramming protocols and selecting the optimal dose of viral particles [82]. Non-integrating viral vectors are gradually removed from the proliferating and maturing iPSCs. It allows achieving the reprogramming goals without permanent OSKM integration into the cell genome. OSKM factors can also be delivered into cells using non-viral vectors such as transposons [84], episomal plasmids [85], mRNA [86], etc. In addition, pluripotency can be induced using siRNA [87] and chemical compounds [88–90] both without OSKM. Such approaches increase the efficiency of dedifferentiation and subsequent reprogramming. As mentioned above, the effectiveness of reprogramming highly depends on the ecto- , meso-, or endodermal origin of the cells transfected into iPSCs, as well as the basal expression level of the genes used to induce pluripotency. Mouse and human iPSCs can be derived from NSCs using only Oct4 and Klf4, or even Oct4 alone, because the basal expression of Sox2, c-Myc and some other genes is sufficiently high in NSCs and does not require stimulation [70, 91, 92].

The same approaches and techniques are frequently used for direct reprogramming except for the dedifferentiation step. Using various combinations of transcription factors, small molecules, mRNA, miRNA, and other inducers, it is potentially possible to derive cells of any differentiation stage, except embryonic stem cells, from any cell. Currently, this technique is used to derive progenitor cells at different stages of embryonic and adult neurogenesis such as NSCs [93, 94], intermediate progenitors [95, 96], and RGCs [97] from mouse and human fibroblasts and monocytes in vitro.

Protocols for direct in vitro and in vivo reprogramming of various cells into specialized neurons (induced neurons, iNs) like glutamatergic, serotonergic, acetylcholinergic, motor ones and others are described in detail and systematized [98, 99]. For example, human fibroblasts were transdifferentiated in vitro into cells that demonstrated morphological features, gene expression, protein secretion , and electrophysiological properties typical of dopaminergic neurons. This result was achieved using a combination of several lentiviral vectors carrying the hAscl1, hNurr1, hLmx1a, and hmiR-124 genes [100]. Dopaminergic neurons were derived from human fetal fibroblasts without using viral vectors by culturing the cells in the presence of small molecules and growth factors such as Valproic acid; aromatic heterocyclic compounds including Repsox (transforming growth factor beta receptor 1 (TGFβR1) inhibitor), kenpaullon (glycogen synthase kinase 3β inhibitor), forskolin (adenylate cyclase activator), purmorphamine (Sonic hedgehog (Shh) pathway activator); proteins such as Wnt1, Wnt5a, fibroblast growth factors, and the protein encoded by the Shh gene [101]. Human embryonic lung fibroblasts from 7–9 weeks of development were successfully reprogrammed in vitro into cortical glutamatergic neurons using a lentiviral vector carrying the Fezf2, Ctip2, Ngn2, and NeuroD1 genes. Reprogrammed cells were identified as iNs, namely cortical neurons, based on electrophysiological activity and levels of the neuronal markers SOX5, FEZF2, CTIP2, OTX1, TBR1, and SATB2. The generated iNs formed synaptic contacts when co-cultured with cells from a primary culture of human cortical neurons, as confirmed by the detection of formed dendritic spines and the presence of the postsynaptic structural protein PSD95 associated with excitatory potential generation [102].

Induced neurons can be obtained either directly or by pre-generating iPSCs both via in vitro delivering a vector carrying target genes into somatic cells. The resulting reprogrammed cells are being inoculated directly into the target area to integrate. In gene therapy, viral vector-based agents could require local or systemic administration depending on the disease (Fig. 3). Only animal models have been brought in trials of in vivo cell reprogramming so far. However, the advantages and disadvantages of both local and systemic administration of the viral vector have been under investigation.

 

Fig. 3. Schematic representation of cellular reprogramming in vitro and in vivo: TFs, transcription factors; pAAV-transgene, viral vector carrying target genes; iPSCs, induced pluripotent stem cells.

Рис. 3. Схематичное изображение клеточного репрограммирования в условиях in vitro и in vivo: TFs — факторы транскрипции (Transcription factors); pAAV-transgene — вирусный вектор с тaргетными генами; iPSCs — индуцированные плюрипотентные клетки.

 

Transdifferentiated iNs of different subpopulations as well as cells with a lower level of differentiation could be obtained from somatic cells as well as from progenitor and glial cells by direct reprogramming. For example, Rivetti di Val Cervo et al. [103] obtained induced dopaminergic neurons from human astrocytes both in vitro and in vivo in a mouse model of Parkinson disease [103]. The modelling was performed via an injection of 6-hydroxydopamine into one hemisphere of mice brain to cause death of dopaminergic neurons in the ventral midbrain of the ipsilateral hemisphere, as well as denervation and reactive gliosis. A viral vector containing a plasmid with Neurod1, Ascl1, and Lmx1a genes, as well as the miRNA miR218, which can only be expressed by GFAP-positive cells, was injected transcranially into the ipsilateral striatum. Todd et al. [9] performed in vivo reprogramming of Müller glia cells into neuron-like retinal cells in mice by intravitreal inoculation of a lentiviral vector with a plasmid carrying Atoh1:Ascl1 genes [9].

In addition to direct vector delivery to a specific brain region, intrathecal and intraventricular injections of viral vectors-based agents carrying transcription factors genes were quite effective [104]. The possibility of systemic delivery of the plasmid on AAV-vectors and reaching the CNS was also evaluated. An intravenous administration of viral particles that could be activated only by interaction with specific markers for radial glia and astrocytes (GFAP) and neurons (synaptophysin) resulted in transgene expression, monitored via GFP fluorescence, was detected only within the CNS, albeit in a small number of target cells [105].

There is a wide range of approaches and methods available for the direct proneural reprogramming. This type of reprogramming is widely investigated, improved and applied as protocols for preclinical experimental studies. AAV vectors as gene delivery systems offer most available high short-term safety and stability compared with other viral and non-viral systems for reprogramming. However, clinical trials of therapeutic systems based on direct neuronal reprogramming are still not possible due to some serious limitations.

LIMITATIONS OF IN VIVO CLINICAL USE OF DIRECT PRONEURAL REPROGRAMMING

Gene therapy, including those based on AAV vectors, is already used in clinical practice. Currently, 26 viral vector-based agents are approved for in vivo use by the U.S. Food and Drug Administration (FDA) and other regulatory agencies [104]. All of these agents insert functional nucleotide sequences into the DNA of patient’s cells to replace defective ones in rare genetic diseases, including those affecting the nervous system. For example, Solgensma (onasemnogene abeparvovec) uses AAV serotype 9 capsids with plasmids containing a copy of the survival of motor neuron (SMN) gene, which dysfunction leads to spinal muscular atrophy manifestation [106]. Similarly, Upstaza (eladocagene exuparvovec1) has been developed for gene therapy of aromatic L-amino acid decarboxylase (AADC) deficiency. The absence of AADC disrupts the synthesis of serotonin and dopamine and results in death in early childhood [107]. However, none of the approved agents affect the degree of cell differentiation and cell phenotype. Despite a growing understanding of mechanisms of proneural reprogramming and the development of new protocols, including suitable for in situ use, there are still some fundamental issues to be resolved before clinical trials would become feasible.

Many papers described the in vivo direct proneural reprogramming using transgenic mouse lines with cells expressing Cre-loxP or Flp-FRT recombinases. These recombinases trigger expression of plasmid-embedded target genes [108]. Activation of the recombinases also induces the synthesis of the fluorescent GFP, which make it possible to detect cells expressing target genes and synthesizing the necessary proteins using in vivo imaging systems [109]. However, it is not always successful even in mice [110]. Similarly, in situ expression of direct reprogramming factors could theoretically be initiated in humans. However, it is not currently possible to assess the efficiency of reprogramming in vivo using available imaging systems designed for cell lines and small organisms such as mice.

In addition, there are still no completely safe methods for the delivery of gene constructs. As mentioned above, the risk of malignancies after AAV vector-based gene therapy is quite low. However, adverse reactions are common with both local and systemic administration of such agents. Preclinical studies in mice, rats, and primates displayed damage to neurons in dorsal root ganglia (DRG), accompanied by ataxia and proprioceptive sensory impairment [111–113]. A case report describes dorsal root gangliopathy following intravenous administration of AAV-miR-SOD1 to two patients for the treatment of amyotrophic lateral sclerosis caused by a mutation in the superoxide dismutase 1 gene. One patient reported tingling in the hands and severe shooting pain in the left foot after drug administration, which correlated with electrophysiologic changes and DRG contrast enhancement on magnetic resonance imaging. The second patient had previously received immunosuppressive therapy and showed no clinical manifestations of DRG damage [114]. Toxic liver damage of varying severity may occur in 60% of cases during systemic gene therapy with AAV vector-based agents [115]. Thrombotic microangiopathy (TMA) is also a common complication (8.2%; 0.5%-75.0%), which can lead to death in the short term, and 1-year survival after therapy with relief of TMA symptoms varies from 16% to 80% [116, 117]. Such adverse reactions are frequently associated with a systemic immune response to the administration of large numbers of viral particles. The vast majority of people are infected with a variety of different viruses during their lifetime, requiring careful multi-step testing for antibodies to AAV capsids prior to treatment. If a patient has antibodies to certain types of viruses, the adaptive immune response is activated before the agent will be able to induce any therapeutic effect [118].

Available viral vector-based gene therapy agents are primarily used in early childhood to treat severe congenital diseases. Up to six months of age, infants may have antibodies that are transplacentally transferred from the mother. However, the older the patient, the higher the possibility of having antibodies to adenoviral capsids [119]. However, the administration of large numbers of viral copies inevitably activates innate and adaptive immunity even in the absence of such antibodies prior to therapy. It correlates with the development of side effects as well. Advances in gene therapy regimens may reduce the incidence and severity of side effects, including by reducing the number of viral copies in a dose of the agent and increasing the number of doses administered [120]. However, this approach significantly increases the financial cost of treatment. Intensive immunosuppressive therapy is still required to achieve the necessary therapeutic effect on the underlying disease. At the same time, immunosuppressive agents such as corticosteroids, sirolimus, tacrolimus, complement inhibitors, and various monoclonal antibodies [114, 117, 121] cause a variety of systemic side effects themselves and have many contraindications for use. Given the high incidence of chronic diseases in the older population and the need for treatment to control the symptoms and progression of medical conditions, it is unlikely that the use of AAV vector-based agents for proneuronal reprogramming of glial cells will be approved for clinical trials in the near future, particularly in patients with neurodegenerative diseases.

Notably, it is not currently possible to precisely and reproducibly direct differentiation towards the desired subtype of differentiated cells. The use of Sox2, one of the key transcription factors in reprogramming, was reported to obtain low differentiated RGCs, NPCs, and neuroblasts in brain and spinal cord tissues, including in vivo [53, 74–76]. Although the outcome of reprogramming depends on the number of copies of vector administered, it is difficult to say with high certainty which specific cell population will be obtained in situ and in what quantity because different differentiation stages express many identical markers [28]. The division cycle of neural stem cells is extremely short in embryogenesis and lasts 10–18 hours [122], whereas the maturation of a neuron from NSCs or NPCs can take up to two weeks in the adult brain under physiological conditions [123, 124]. Therefore, the formation rate of mature specialized neurons from glial cells in situ and the effectiveness of their functional integration to restore lost neuronal connections will be extremely difficult to predict and regulate in pathological conditions, especially in neurodegenerative diseases.

Most experiments reported so called leaky expression, which is detectable expression of target genes by non-target cells [125]. When transcription factors with activity limited to the initial stages of cell differentiation are administered externally, a complex cascade of processes involving DNA unfolding and interaction with enhancers and promoters of target genes appears to be initiated in cells that need to be reprogrammed. In addition, Katsuda et al. [126] reported hepatobiliary metaplasia in mice after systemic administration of an AAV vector carrying the genes of Sox4 and Sox9 transcription factors [126]. Similar changes occur with toxic damage to the organ and can lead to adenocarcinoma [127]. Spontaneous metaplasia of glial cells has not been described in mammals, and the long-term consequences of their reprogramming into neurons have not been studied in vivo. Given the uncertain probability of metaplasia transition to dysplasia with subsequent malignancy, especially in older patients with comorbidities and an age-related increased risk of cancer, the potential side effects outweigh the potential therapeutic benefit at the current stage of research and development of AAV vector technologies.

Epigenetic changes inevitably acquired by people throughout their life might have a significant impact on the effectiveness of reprogramming as well. Some single-cell transcriptomic studies of iPSCs derived from commercial cell lines of mature fibroblasts reported the expression of molecular genetic signatures typical of NSCs [128]. However, direct reprogramming has shown that transdifferentiated iNs derived from cells of young and old people retain transcriptional signatures of aging due to unique epigenetic changes [129]. Therefore, given the influence of aging and the associated inflammaging that inhibits neurogenesis, there is a high probability that even successful direct reprogramming of astrocytes into neurons in situ in humans with age-related neurodegenerative diseases may be therapeutically ineffective due to the senescent phenotype of glial cells.

Many of the methodological and therapeutic issues described are likely to be resolved in the near future. However, it is necessary to integrate new neurons not only in close proximity to neurogenic zones such as the hippocampus, but also at a great distance from them in order to achieve a significant and lasting therapeutic effect in the treatment of various CNS diseases. Neurogenesis is a complex process that is tightly regulated by the internal genetic programs of progenitor cells and mature neurons, as well as by external microenvironmental factors. As neurons differentiate, they lose the ability to extend axons due to selective inhibition of migration factors and integrins that were highly expressed during embryogenesis [130]. Attempts to use bone marrow stem cells and embryonic CNS cells to treat spinal cord injury have been successful in rodent experiments [131], but clinical trials have not reported any significant efficacy [132]. Some other clinical trials assessed an effect of intracranial injection of embryonic dopaminergic neurons into the basal ganglia region of patients with Parkinson disease. Despite the high survival rate of the implanted cells, clinical improvement was either absent or unstable [133, 134]. It is clear that targeting a specific pathway will not be sufficient to fully regenerate damaged axons in the adult mammalian CNS, and manipulating a large number of genes at once is an extremely complex task with a high risk of adverse consequences. Therefore, effective proneural reprogramming is still not possible, especially in older patients with comorbidities and a senescent cell phenotype.

CONCLUSION

The integration of multi-step and mutually regulated genomic, epigenomic, and biochemical pathways involved in cell plasticity processes, provides the basis for adaptation and normal function of tissues, organs, and systems. Numerous signaling pathways with similar functions form a complex, self-sustaining, and self-regulating system that ensures the stability of the entire organism when one or more pathways are blocked. The short- and long-term consequences of external interference with the histogenetic signatures of cells, especially when elements of such a complex structure as the human brain are affected, are still poorly understood. In addition, there are a number of fundamental barriers that need to be overcome, such as a lack of ability to ensure the growth of axons to CNS regions that are significantly distant from neurogenic zones. Further studies are warranted to better understand the fundamental cellular and molecular biological processes underlying cellplasticity and transdifferentiation under physiological and pathological conditions. In addition, available gene therapy methods need to be continually improved to secure the safety and efficacy of direct reprogramming approaches.

ADDITIONAL INFORMATION

Author's contributions. A.V. Sentyabreva: literature review and analysis, illustration creation, manuscript writing and editing. The author approved the manuscript (final version for publication) and agreed to take responsibility for all aspects of the work, ensuring proper investigation and resolution of any issues related to accuracy and integrity.

Acknowledgements. The author expresses gratitude to Dr. Sci. (Biology) Anna M. Kosyreva, for her consultative assistance in editing the review.

Funding sources. None declared.

Disclosure of interests. The author declares no relationships, activities, or competing interests with third parties (commercial or non-commercial) that could be affected by the content of this article within the past three years.

 

1 The product is not approved in the Russian Federation

×

作者简介

Alexandra V. Sentyabreva

Petrovsky National Research Centre of Surgery

编辑信件的主要联系方式.
Email: alexandraasentyabreva@gmail.com
ORCID iD: 0000-0001-5064-219X
SPIN 代码: 6966-9959
俄罗斯联邦, Moscow

参考

  1. Jorstad NL, Wilken MS, Grimes WN, et al. Stimulation of functional neuronal regeneration from Müller glia in adult mice. Nature. 2017;548(7665):103–107. doi: 10.1038/nature23283
  2. Qian L, Huang Y, Spencer CI, et al. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature. 2012;485(7400):593–598. doi: 10.1038/nature11044
  3. Ieda M, Fu JD, Delgado-Olguin P, et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell. 2010;142(3):375–386. doi: 10.1016/j.cell.2010.07.002
  4. Cavelti-Weder C, Zumsteg A, Li W, Zhou Q. Reprogramming of pancreatic acinar cells to functional beta-cells by in vivo transduction of a polycistronic construct containing Pdx1, Ngn3, MafA in mice. Curr Protoc Stem Cell Biol. 2017;40:4A.10.1–4A.10.12. doi: 10.1002/cpsc.21
  5. Kosyreva AM, Sentyabreva AV, Tsvetkov IS, Makarova OV. Alzheimer’s disease and inflammaging. Brain Sci. 2022;12(9):1237. doi: 10.3390/brainsci12091237
  6. Tai W, Wu W, Wang LL, et al. In vivo reprogramming of NG2 glia enables adult neurogenesis and functional recovery following spinal cord injury. Cell Stem Cell. 2021;28(5):923–937.e4. doi: 10.1016/j.stem.2021.02.009
  7. Tang Y, Wu Q, Gao M, et al. Restoration of visual function and cortical connectivity after ischemic injury through neurod1-mediated gene therapy. Front Cell Dev Biol. 2021;9:720078. doi: 10.3389/fcell.2021.720078
  8. Chen YC, Ma NX, Pei ZF, et al. A NeuroD1 AAV-based gene therapy for functional brain repair after ischemic injury through in vivo astrocyte-to-neuron conversion. Mol Ther. 2020;28(1):217–234. doi: 10.1016/j.ymthe.2019.09.003
  9. Todd L, Hooper MJ, Haugan AK, et al. Efficient stimulation of retinal regeneration from Müller glia in adult mice using combinations of proneural bHLH transcription factors. Cell Rep. 2021;37(3):109857. doi: 10.1016/j.celrep.2021.109857
  10. Le N, Vu TD, Palazzo I, et al. Robust reprogramming of glia into neurons by inhibition of Notch signaling and nuclear factor I (NFI) factors in adult mammalian retina. Sci Adv. 2024;10(28):eadn2091. doi: 10.1126/sciadv.adn2091
  11. Qian H, Kang X, Hu J, et al. Reversing a model of Parkinson’s disease with in situ converted nigral neurons. Nature. 2020;582(7813):550–556. doi: 10.1038/s41586-020-2388-4
  12. Wu Z, Parry M, Hou XY, et al. Gene therapy conversion of striatal astrocytes into GABAergic neurons in mouse models of Huntington’s disease. Nat Commun. 2020;11(1):1105. doi: 10.1038/s41467-020-14855-3
  13. Kriegstein A, Alvarez-Buylla A. The glial nature of embryonic and adult neural stem cells. Annu Rev Neurosci. 2009;32:149–184. doi: 10.1146/annurev.neuro.051508.135600
  14. Malatesta P, Appolloni I, Calzolari F. Radial glia and neural stem cells. Cell Tissue Res. 2008;331(1):165–178. doi: 10.1007/s00441-007-0481-8
  15. Miranda-Negrón Y, García-Arrarás JE. Radial glia and radial glia-like cells: Their role in neurogenesis and regeneration. Front Neurosci. 2022;16:1006037. doi: 10.3389/fnins.2022.1006037
  16. Howard BM, Mo Z, Filipovic R, et al. Radial glia cells in the developing human brain. Neuroscientist. 2008;14(5):459–473. doi: 10.1177/1073858407313512
  17. Stiles J, Jernigan TL. The basics of brain development. Neuropsychol Rev. 2010;20(4):327–348. doi: 10.1007/s11065-010-9148-4
  18. Kobat SG, Turgut B. Importance of Müller cells. Beyoglu Eye J. 2020;5(2):59–63. doi: 10.14744/bej.2020.28290
  19. Yamada K, Watanabe M. Cytodifferentiation of Bergmann glia and its relationship with Purkinje cells. Anat Sci Int. 2002;77(2):94–108. doi: 10.1046/j.0022-7722.2002.00021.x
  20. Allen NJ, Lyons DA. Glia as architects of central nervous system formation and function. Science. 2018;362(6411):181–185. doi: 10.1126/science.aat0473
  21. Gage FH, Temple S. Neural stem cells: generating and regenerating the brain. Neuron. 2013;80(3):588–601. doi: 10.1016/j.neuron.2013.10.037
  22. Ming GL, Song H. Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron. 2011;70(4):687–702. doi: 10.1016/j.neuron.2011.05.001
  23. Purves D, Augustine GJ, Fitzpatrick D. Neuroscience. 6th ed. New York Oxford: Oxford University Press; 2018.
  24. Jurkowski MP, Bettio L, K Woo EK, et al. Beyond the hippocampus and the SVZ: adult neurogenesis throughout the brain. Front Cell Neurosci. 2020;14:576444. doi: 10.3389/fncel.2020.576444
  25. Ernst A, Alkass K, Bernard S, et al. Neurogenesis in the striatum of the adult human brain. Cell. 2014;156(5):1072–1083. doi: 10.1016/j.cell.2014.01.044
  26. Sanai N, Nguyen T, Ihrie RA, et al. Corridors of migrating neurons in human brain and their decline during infancy. Nature. 2011;478(7369):382–386. doi: 10.1038/nature10487
  27. Semënov MV. Adult hippocampal neurogenesis is a developmental process involved in cognitive development. Front Neurosci. 2019;13:159. doi: 10.3389/fnins.2019.00159
  28. Franzén O, Gan LM, Björkegren JLM. PanglaoDB: a web server for exploration of mouse and human single-cell RNA sequencing data. Database (Oxford). 2019;2019:baz046. doi: 10.1093/database/baz046
  29. Mori T, Buffo A, Götz M. The novel roles of glial cells revisited: the contribution of radial glia and astrocytes to neurogenesis. Curr Top Dev Biol. 2005;69:67–99. doi: 10.1016/S0070-2153(05)69004-7
  30. Ninkovic J, Steiner-Mezzadri A, Jawerka M, et al. The BAF complex interacts with Pax6 in adult neural progenitors to establish a neurogenic cross-regulatory transcriptional network. Cell Stem Cell. 2013;13(4):403–418. doi: 10.1016/j.stem.2013.07.002
  31. Ortega F, Gascón S, Masserdotti G, et al. Oligodendrogliogenic and neurogenic adult subependymal zone neural stem cells constitute distinct lineages and exhibit differential responsiveness to Wnt signalling. Nat Cell Biol. 2013;15(6):602–613. doi: 10.1038/ncb2736
  32. Bast L, Calzolari F, Strasser MK, et al. Increasing neural stem cell division asymmetry and quiescence are predicted to contribute to the age-related decline in neurogenesis. Cell Rep. 2018;25(12):3231–3240.e8. doi: 10.1016/j.celrep.2018.11.088
  33. Kalamakis G, Brüne D, Ravichandran S, et al. Quiescence modulates stem cell maintenance and regenerative capacity in the aging brain. Cell. 2019;176(6):1407–1419.e14. doi: 10.1016/j.cell.2019.01.040
  34. Fuentealba LC, Obernier K, Alvarez-Buylla A. Adult neural stem cells bridge their niche. Cell Stem Cell. 2012;10(6):698–708. doi: 10.1016/j.stem.2012.05.012
  35. Baklaushev VP, Yusubalieva GM, Samoilova EM, et al. Resident neural stem cell niches and regeneration: the splendors and miseries of adult neurogenesis. Russ J Dev Biol. 2022;53:159–179. doi: 10.1134/S1062360422030080
  36. Sorrells SF, Paredes MF, Cebrian-Silla A, et al. Human hippocampal neurogenesis drops sharply in children to undetectable levels in adults. Nature. 2018;555(7696):377–381. doi: 10.1038/nature25975
  37. Zhou Y, Su Y, Li S, et al. Molecular landscapes of human hippocampal immature neurons across lifespan. Nature. 2022;607(7919):527–533. doi: 10.1038/s41586-022-04912-w
  38. Moreno-Jiménez EP, Flor-García M, Terreros-Roncal J, et al. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat Med. 2019;25(4):554–560. doi: 10.1038/s41591-019-0375-9
  39. Ojaghihaghighi S, Vahdati SS, Mikaeilpour A, Ramouz A. Comparison of neurological clinical manifestation in patients with hemorrhagic and ischemic stroke. World J Emerg Med. 2017;8(1):34–38. doi: 10.5847/wjem.j.1920-8642.2017.01.006
  40. Salmeron KE, Maniskas ME, Edwards DN, et al. Interleukin 1 alpha administration is neuroprotective and neuro-restorative following experimental ischemic stroke. J Neuroinflammation. 2019;16(1):222. doi: 10.1186/s12974-019-1599-9
  41. Grinchevskaya LR, Salikhova DI, Silachev DN, Goldshtein DV. Neural and glial regulation of angiogenesis in CNS in ischemic stroke. Bull Exp Biol Med. 2024;177(4):528–533. doi: 10.1007/s10517-024-06219-4
  42. Zheng W, ZhuGe Q, Zhong M, et al. Neurogenesis in adult human brain after traumatic brain injury. J Neurotrauma. 2013;30(22):1872–1880. doi: 10.1089/neu.2010.1579
  43. Bielefeld P, Martirosyan A, Martín-Suárez S, et al. Traumatic brain injury promotes neurogenesis at the cost of astrogliogenesis in the adult hippocampus of male mice. Nat Commun. 2024;15(1):5222. doi: 10.1038/s41467-024-49299-6
  44. Wu A, Zhang J. Neuroinflammation, memory, and depression: new approaches to hippocampal neurogenesis. J Neuroinflammation. 2023;20(1):283. doi: 10.1186/s12974-023-02964-x
  45. 2022 Alzheimer’s disease facts and figures. Alzheimers Dement. 2022;18(4):700–789. doi: 10.1002/alz.12638
  46. Schwab L, Richetin K, Barker R, Déglon N. Formation of hippocampal mHTT aggregates leads to impaired spatial memory, hippocampal activation and adult neurogenesis. Neurobiol Dis. 2017;102:105–112. doi: 10.1016/j.nbd.2017.03.005
  47. Rahman AA, Amruta N, Pinteaux E, Bix GJ. Neurogenesis after stroke: a therapeutic perspective. Transl Stroke Res. 2021;12(1):1–14. doi: 10.1007/s12975-020-00841-w
  48. Sentyabreva AV, Miroshnichenko EA, Melnikova EA, et al. Morphofunctional changes in brain and peripheral blood in adult and aged Wistar rats with AlCl3-induced neurodegeneration. Biomedicines. 2023;11(9):2336. doi: 10.3390/biomedicines11092336
  49. Virchow R. Cellular pathology. As based upon physiological and pathological histology. Lecture XVI--Atheromatous affection of arteries. 1858. Nutr Rev. 1989;47(1):23–25. doi: 10.1111/j.1753-4887.1989.tb02747.x
  50. Virchow R. Ueber Metaplasie. Archiv f. pathol. Anat. 1984;97:410–430. doi: 10.1007/BF02430434
  51. Kumar V, Abbas AK, Aster JC. Robbins & Cotran pathologic basis of disease. 10th ed. Elsevier Health Sciences; 2020.
  52. Slack JM, Tosh D. Transdifferentiation and metaplasia — switching cell types. Curr Opin Genet Dev. 2001;11(5):581–586. doi: 10.1016/S0959-437X(00)00236-7
  53. Slack JMW. Metaplasia and transdifferentiation: from pure biology to the clinic. Nat Rev Mol Cell Biol. 2007;8(5):369–378. doi: 10.1038/nrm2146
  54. Jenkins GJS, D’Souza FR, Suzen SH, et al. Deoxycholic acid at neutral and acid pH, is genotoxic to oesophageal cells through the induction of ROS: the potential role of anti-oxidants in Barrett’s oesophagus. Carcinogenesis. 2007;28(1):136–142. doi: 10.1093/carcin/bgl147
  55. Gray MR, Donnelly RJ, Kingsnorth AN. The role of smoking and alcohol in metaplasia and cancer risk in Barrett’s columnar lined oesophagus. Gut. 1993;34(6):727–731. doi: 10.1136/gut.34.6.727
  56. Peters EJ, Morice R, Benner SE, et al. Squamous metaplasia of the bronchial mucosa and its relationship to smoking. Chest. 1993;103(5):1429–1432. doi: 10.1378/chest.103.5.1429
  57. Sangani RG, Deepak V, Ghio AJ, et al. Peribronchiolar metaplasia: a marker of cigarette smoke-induced small airway injury in a rural cohort. Clin Pathol. 2023;16:2632010X231209878. doi: 10.1177/2632010X231209878
  58. Ogura Y, Ishii K, Kanda H, et al. Bisphenol A induces permanent squamous change in mouse prostatic epithelium. Differentiation. 2007;75(8):745–756. doi: 10.1111/j.1432-0436.2007.00177.x
  59. Amin MAS, Sonpol HMA, Gouda RHE, Aboregela AM. Bisphenol A enhances apoptosis, fibrosis, and biochemical fluctuations in the liver of adult male rats with possible regression after recovery. Anat Rec (Hoboken). 2023;306(1):213–225. doi: 10.1002/ar.25032
  60. Holtzman MJ, Battaile JT, Patel AC. Immunogenetic programs for viral induction of mucous cell metaplasia. Am J Respir Cell Mol Biol. 2006;35(1):29–39. doi: 10.1165/rcmb.2006-0092SF
  61. Lee JS, Oh TY, Ahn BO, et al. Involvement of oxidative stress in experimentally induced reflux esophagitis and Barrett’s esophagus: clue for the chemoprevention of esophageal carcinoma by antioxidants. Mutat Res. 2001;480–481:189–200. doi: 10.1016/S0027-5107(01)00199-3
  62. Feng C, Luo Y, Nian Y, et al. Diallyl disulfide suppresses the inflammation and apoptosis resistance induced by DCA through ROS and the NF-κB signaling pathway in human Barrett’s epithelial cells. Inflammation. 2017;40(3):818–831. doi: 10.1007/s10753-017-0526-4
  63. Giroux V, Rustgi AK. Metaplasia: tissue injury adaptation and a precursor to the dysplasia–cancer sequence. Nat Rev Cancer. 2017;17(10):594–604. doi: 10.1038/nrc.2017.68
  64. Holmberg D, Ness-Jensen E, Mattsson F, et al. Risk of oesophageal adenocarcinoma in individuals with Barrett’s oesophagus. Eur J Cancer. 2017;75:41–46. doi: 10.1016/j.ejca.2016.12.037
  65. King TC, editor. Front Matter. In: Elsevier’s Integrated Pathology. Saint Louis: Mosby; 2007. https://archive.org/details/elseviersintegra0000king
  66. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676. doi: 10.1016/j.cell.2006.07.024
  67. Liang G, Zhang Y. Embryonic stem cell and induced pluripotent stem cell: an epigenetic perspective. Cell Res. 2013;23(1):49–69. doi: 10.1038/cr.2012.175
  68. Aasen T, Raya A, Barrero MJ, et al. Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nat Biotechnol. 2008;26(11):1276–1284. doi: 10.1038/nbt.1503
  69. Utikal J, Maherali N, Kulalert W, Hochedlinger K. Sox2 is dispensable for the reprogramming of melanocytes and melanoma cells into induced pluripotent stem cells. J Cell Sci. 2009;122(Pt 19):3502–3510. doi: 10.1242/jcs.054783
  70. Kim JB, Greber B, Araúzo-Bravo MJ, et al. Direct reprogramming of human neural stem cells by OCT4. Nature. 2009;461(7264):649–653. doi: 10.1038/nature08436
  71. Huang P, Sun L, Zhang L, Hui L. Conversion of fibroblasts to hepatocytes in vitro. Methods in Mol Biol. 2019;1905:93–101. doi: 10.1007/978-1-4939-8961-4_9
  72. Du J, Liu X, Wong CWY, et al. Direct cellular reprogramming and transdifferentiation of fibroblasts on wound healing — Fantasy or reality? Chronic Dis Transl Med. 2023;9(3):191–199. doi: 10.1002/cdt3.77
  73. He S, Chen J, Zhang Y, et al. Sequential EMT-MET induces neuronal conversion through Sox2. Cell Discov. 2017;3:17017. doi: 10.1038/celldisc.2017.17
  74. Singh S, Howell D, Trivedi N, et al. Zeb1 controls neuron differentiation and germinal zone exit by a mesenchymal-epithelial-like transition. ELife. 2016;5:e12717. doi: 10.7554/eLife.12717
  75. Hu S, Zhao MT, Jahanbani F, et al. Effects of cellular origin on differentiation of human induced pluripotent stem cell–derived endothelial cells. JCI Insight. 2016;1(8):e85558. doi: 10.1172/jci.insight.85558
  76. Scesa G, Adami R, Bottai D. iPSC preparation and epigenetic memory: does the tissue origin matter? Cells. 2021;10(6):1470. doi: 10.3390/cells10061470
  77. Hargus G, Ehrlich M, Araúzo-Bravo MJ, et al. Origin-dependent neural cell identities in differentiated human iPSCs in vitro and after transplantation into the mouse brain. Cell Rep. 2014;8(6):1697–1703. doi: 10.1016/j.celrep.2014.08.014
  78. Stone NR, Gifford CA, Thomas R, et al. Context-specific transcription factor functions regulate epigenomic and transcriptional dynamics during cardiac reprogramming. Cell Stem Cell. 2019;25(1):87–102.e9. doi: 10.1016/j.stem.2019.06.012
  79. Wang H, Yang Y, Liu J, Qian L. Direct cell reprogramming: approaches, mechanisms and progress. Nat Rev Mol Cell Biol. 2021;22(6):410–424. doi: 10.1038/s41580-021-00335-z
  80. Belousova E, Salikhova D, Maksimov Y, et al. Proposed mechanisms of cell therapy for Alzheimer’s disease. Int J Mol Sci. 2024;25(22):12378. doi: 10.3390/ijms252212378
  81. Yu J, Vodyanik MA, Smuga-Otto K, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318(5858):1917–1920. doi: 10.1126/science.1151526
  82. Ay C, Reinisch A. Gene therapy: principles, challenges and use in clinical practice. Wien Klin Wochenschr. 2024. doi: 10.1007/s00508-024-02368-8
  83. Haridhasapavalan KK, Borgohain MP, Dey C, et al. An insight into non-integrative gene delivery approaches to generate transgene-free induced pluripotent stem cells. Gene. 2019;686:146–159. doi: 10.1016/j.gene.2018.11.069
  84. Woltjen K, Michael IP, Mohseni P, et al. piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature. 2009;458(7239):766–770. doi: 10.1038/nature07863
  85. Chao J, Feng L, Ye P, et al. Therapeutic development for Canavan disease using patient iPSCs introduced with the wild-type ASPA gene. iScience. 2022;25(6):104391. doi: 10.1016/j.isci.2022.104391
  86. Warren L, Manos PD, Ahfeldt T, et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells using synthetic modified mRNA. Cell Stem Cell. 2010;7(5):618–630. doi: 10.1016/j.stem.2010.08.012
  87. Miyoshi N, Ishii H, Nagano H, et al. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell. 2011;8(6):633–638. doi: 10.1016/j.stem.2011.05.001
  88. Hou P, Li Y, Zhang X, et al. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science. 2013;341(6146):651–654. doi: 10.1126/science.1239278
  89. Zhao T, Fu Y, Zhu J, et al. Single-cell RNA-Seq reveals dynamic early embryonic-like programs during chemical reprogramming. Cell Stem Cell. 2018;23(1):31–45.e7. doi: 10.1016/j.stem.2018.05.025
  90. Chen G, Guo Y, Li C, et al. Small molecules that promote self-renewal of stem cells and somatic cell reprogramming. Stem Cell Rev Rep. 2020;16(3):511–523. doi: 10.1007/s12015-020-09965-w
  91. Kim JB, Zaehres H, Wu G, et al. Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors. Nature. 2008;454(7204):646–650. doi: 10.1038/nature07061
  92. Kim JB, Sebastiano V, Wu G, et al. Oct4-induced pluripotency in adult neural stem cells. Cell. 2009;136(3):411–419. doi: 10.1016/j.cell.2009.01.023
  93. Ring KL, Tong LM, Balestra ME, et al. Direct reprogramming of mouse and human fibroblasts into multipotent neural stem cells with a single factor. Cell Stem Cell. 2012;11(1):100–109. doi: 10.1016/j.stem.2012.05.018
  94. Thier MC, Hommerding O, Panten J, et al. Identification of embryonic neural plate border stem cells and their generation by direct reprogramming from adult human blood cells. Cell Stem Cell. 2019;24(1):166–182.e13. doi: 10.1016/j.stem.2018.11.015
  95. Xu XL, Yang JP, Fu LN, et al. Direct reprogramming of porcine fibroblasts to neural progenitor cells. Protein Cell. 2014;5(1):4–7. doi: 10.1007/s13238-013-0015-y
  96. Ahlfors JE, Azimi A, El-Ayoubi R, et al. Examining the fundamental biology of a novel population of directly reprogrammed human neural precursor cells. Stem Cell Res Ther. 2019;10(1):166. doi: 10.1186/s13287-019-1255-4
  97. Beattie R, Hippenmeyer S. Mechanisms of radial glia progenitor cell lineage progression. FEBS Lett. 2017;591(24):3993–4008. doi: 10.1002/1873-3468.12906
  98. El Wazan L, Urrutia-Cabrera D, Wong RCB. Using transcription factors for direct reprogramming of neurons in vitro. World J Stem Cells. 2019;11(7):431–444. doi: 10.4252/wjsc.v11.i7.431
  99. Samoilova EM, Belopasov VV, Baklaushev VP. Transcription factors of direct neuronal reprogramming in ontogenesis and ex vivo. Mol Biol. 2021;55:645–669. doi: 10.1134/S0026893321040087
  100. Jiang H, Xu Z, Zhong P, et al. Cell cycle and p53 gate the direct conversion of human fibroblasts to dopaminergic neurons. Nat Commun. 2015;6:10100. doi: 10.1038/ncomms10100
  101. Qin H, Zhao AD, Sun ML, et al. Direct conversion of human fibroblasts into dopaminergic neuron-like cells using small molecules and protein factors. Mil Med Res. 2020;7(1):52. doi: 10.1186/s40779-020-00284-2
  102. Miskinyte G, Devaraju K, Grønning Hansen M, et al. Direct conversion of human fibroblasts to functional excitatory cortical neurons integrating into human neural networks. Stem Cell Res Ther. 2017;8(1):207. doi: 10.1186/s13287-017-0658-3
  103. Rivetti Di Val Cervo P, Romanov RA, Spigolon G, et al. Induction of functional dopamine neurons from human astrocytes in vitro and mouse astrocytes in a Parkinson’s disease model. Nat Biotechnol. 2017;35(5):444–452. doi: 10.1038/nbt.3835
  104. Wang JH, Gessler DJ, Zhan W, et al. Adeno-associated virus as a delivery vector for gene therapy of human diseases. Signal Transduct Target Ther. 2024;9(1):78. doi: 10.1038/s41392-024-01780-w
  105. Dashkoff J, Lerner EP, Truong N, et al. Tailored transgene expression to specific cell types in the central nervous system after peripheral injection with AAV9. Mol Ther Methods Clin Dev. 2016;3:16081. doi: 10.1038/mtm.2016.81
  106. Strauss KA, Farrar MA, Muntoni F, et al. Onasemnogene abeparvovec for presymptomatic infants with two copies of SMN2 at risk for spinal muscular atrophy type 1: the Phase III SPR1NT trial. Nat Med. 2022;28(7):1381–1389. doi: 10.1038/s41591-022-01866-4
  107. Keam SJ. Eladocagene Exuparvovec: first approval. Drugs. 2022;82(13):1427–1432. doi: 10.1007/s40265-022-01775-3
  108. Hubbard EJA. FLP/FRT and Cre/lox recombination technology in C. elegans. Methods. 2014;68(3):417–424. doi: 10.1016/j.ymeth.2014.05.007
  109. Fischer KB, Collins HK, Callaway EM. Sources of off-target expression from recombinase-dependent AAV vectors and mitigation with cross-over insensitive ATG-out vectors. Proc Natl Acad Sci U S A. 2019;116(52):27001–27010. doi: 10.1073/pnas.1915974116
  110. Xie Y, Zhou J, Wang LL, et al. New AAV tools fail to detect Neurod1-mediated neuronal conversion of Müller glia and astrocytes in vivo. EBioMedicine. 2023;90:104531. doi: 10.1016/j.ebiom.2023.104531
  111. Hinderer C, Katz N, Buza EL, et al. Severe toxicity in nonhuman primates and piglets following high-dose intravenous administration of an Adeno-associated virus vector expressing human SMN. Hum Gene Ther. 2018;29(3):285–298. doi: 10.1089/hum.2018.015
  112. Fader KA, Pardo ID, Kovi RC, et al. Circulating neurofilament light chain as a promising biomarker of AAV-induced dorsal root ganglia toxicity in nonclinical toxicology species. Mol Ther Methods Clin Dev. 2022;25:264–277. doi: 10.1016/j.omtm.2022.03.017
  113. Buss N, Lanigan L, Zeller J, et al. Characterization of AAV-mediated dorsal root ganglionopathy. Mol Ther Methods Clin Dev. 2022;24:342–354. doi: 10.1016/j.omtm.2022.01.013
  114. Mueller C, Berry JD, McKenna-Yasek DM, et al. SOD1 Suppression with Adeno-associated virus and microRNA in familial ALS. N Engl J Med. 2020;383(2):151–158. doi: 10.1056/NEJMoa2005056
  115. Delire B, De Martin E, Meunier L, et al. Immunotherapy and gene therapy: new challenges in the diagnosis and management of drug-induced liver injury. Front Pharmacol. 2022;12:786174. doi: 10.3389/fphar.2021.786174
  116. Jodele S, Mizuno K, Sabulski A, Vinks AA. Adopting model-informed precision-dosing for Eculizumab in transplant associated-thrombotic microangiopathy to gene therapies. Clin Pharmacol Ther. 2023;114(3):511–514. doi: 10.1002/cpt.2966
  117. Jodele S, Dandoy CE, Lane A, et al. Complement blockade for TA-TMA: lessons learned from a large pediatric cohort treated with eculizumab. Blood. 2020;135(13):1049–1057. doi: 10.1182/blood.2019004218
  118. Falese L, Sandza K, Yates B, et al. Strategy to detect pre-existing immunity to AAV gene therapy. Gene Ther. 2017;24(12):768–778. doi: 10.1038/gt.2017.95
  119. Gross DA, Tedesco N, Leborgne C, Ronzitti G. Overcoming the challenges imposed by humoral immunity to AAV vectors to achieve safe and efficient gene transfer in seropositive patients. Front Immunol. 2022;13:857276. doi: 10.3389/fimmu.2022.857276
  120. Kishimoto TK, Samulski RJ. Addressing high dose AAV toxicity — ‘one and done’ or ‘slower and lower’? Expert Opin Biol Ther. 2022;22(9):1067–1071. doi: 10.1080/14712598.2022.2060737
  121. Servais L, Horton R, Saade D, et al. 261st ENMC International Workshop: Management of safety issues arising following AAV gene therapy. 17th–19th June 2022, Hoofddorp, The Netherlands. Neuromuscul Disord. 2023;33(11):884–896. doi: 10.1016/j.nmd.2023.09.008
  122. Liu L, Michowski W, Kolodziejczyk A, Sicinski P. The cell cycle in stem cell proliferation, pluripotency and differentiation. Nat Cell Biol. 2019;21(9):1060–1067. doi: 10.1038/s41556-019-0384-4
  123. Bonaguidi MA, Wheeler MA, Shapiro JS, et al. In vivo clonal analysis reveals self-renewing and multipotent adult neural stem cell characteristics. Cell. 2011;145(7):1142–1155. doi: 10.1016/j.cell.2011.05.024
  124. Lugert S, Vogt M, Tchorz JS, et al. Homeostatic neurogenesis in the adult hippocampus does not involve amplification of Ascl1(high) intermediate progenitors. Nat Commun. 2012;3:670. doi: 10.1038/ncomms1670
  125. Jung H, Kim SW, Kim M, et al. Noninvasive optical activation of Flp recombinase for genetic manipulation in deep mouse brain regions. Nat Commun. 2019;10(1):314. doi: 10.1038/s41467-018-08282-8
  126. Katsuda T, Sussman JH, Ito K, et al. Cellular reprogramming in vivo initiated by SOX4 pioneer factor activity. Nat Commun. 2024;15(1):1761. doi: 10.1038/s41467-024-45939-z
  127. Barsouk A, Thandra KC, Saginala K, et al. Chemical risk factors of primary liver cancer: an update. Hepat Med. 2021;12:179–188. doi: 10.2147/HMER.S278070
  128. Spathopoulou A, Podlesnic M, De Gaetano L, et al. Single-cell profiling of reprogrammed human neural stem cells unveils high similarity to neural progenitors in the developing central nervous system. Stem Cell Rev Rep. 2024;20(5):1325–1339. doi: 10.1007/s12015-024-10698-3
  129. Mertens J, Paquola ACM, Ku M, et al. Directly reprogrammed human neurons retain aging-associated transcriptomic signatures and reveal age-related nucleocytoplasmic defects. Cell Stem Cell. 2015;17(6):705–718. doi: 10.1016/j.stem.2015.09.001
  130. Andrews MR, Czvitkovich S, Dassie E, et al. Alpha9 integrin promotes neurite outgrowth on tenascin-C and enhances sensory axon regeneration. J Neurosci. 2009;29(17):5546–5557. doi: 10.1523/JNEUROSCI.0759-09.2009
  131. Sasaki M, Radtke C, Tan AM, et al. BDNF-hypersecreting human mesenchymal stem cells promote functional recovery, axonal sprouting, and protection of corticospinal neurons after spinal cord injury. J Neurosci. 2009;29(47):14932–14941. doi: 10.1523/JNEUROSCI.2769-09.2009
  132. Urdzíková L, Jendelová P, Glogarová K et al. Transplantation of bone marrow stem cells as well as mobilization by granulocyte-colony stimulating factor promotes recovery after spinal cord injury in rats. J Neurotrauma. 2006;23(9):1379–1391. doi: 10.1089/neu.2006.23.1379
  133. Freed CR, Zhou W, Breeze RE. Dopamine cell transplantation for Parkinson’s disease: the importance of controlled clinical trials. Neurotherapeutics. 2011;8(4):549–561. doi: 10.1007/s13311-011-0082-9
  134. Sonntag KC, Song B, Lee N, et al. Pluripotent stem cell-based therapy for Parkinson’s disease: current status and future prospects. Prog Neurobiol. 2018;168:1–20. doi: 10.1016/j.pneurobio.2018.04.005

补充文件

附件文件
动作
1. JATS XML
2. Fig. 1. Schematic representation of embryonic brain development. Neural stem cells (NSCs) in the subventricular zone (SVZ) divide symmetrically, generating a pool of radial glial cells (RGCs). Through both symmetric and asymmetric division, RGCs give rise to intermediate progenitor cells: neuronal intermediate progenitor cells (nIPCs), oligodendrocyte intermediate progenitor cells (oIPCs), and astrocyte intermediate progenitor cells (aIPCs). As differentiation progresses, these cells migrate from the neurogenic SVZ, contributing to neocortical histoarchitecture (indicated by the red dashed arrow) and maturing into neurons, oligodendrocytes, and astrocytes. During the earliest stages of embryogenesis, a niche of quiescent NSCs and RGCs (qNSCs/qRGCs) is established, which contribute to neurogenesis in the adult brain.

下载 (222KB)
3. Fig. 2. Stages of neurogenesis in the adult brain of rodents and humans. Quiescent neural stem cells and radial glial cells (qNSCs/ qRGCs) become activated (aNSCs/aRGCs) in response to microenvironmental signals and resume asymmetric division. Unlike embryonic neurogenesis, postnatal neurogenesis does not involve the direct generation of new neurons from radial glial cells (RGCs). Instead, a sequential differentiation process occurs, giving rise to neuronal progenitor cells (NPCs), neuroblasts, immature neurons, and finally, functionally active, mature neurons that integrate into the existing neuronal network. The lower part of the figure (below the arrow) indicates key genes expressed at different stages of cell differentiation during neurogenesis.

下载 (230KB)
4. Fig. 3. Schematic representation of cellular reprogramming in vitro and in vivo: TFs, transcription factors; pAAV-transgene, viral vector carrying target genes; iPSCs, induced pluripotent stem cells.

下载 (265KB)

版权所有 © Eco-Vector, 2024

Creative Commons License
此作品已接受知识共享署名-非商业性使用-禁止演绎 4.0国际许可协议的许可。

Периодический печатный журнал зарегистрирован как СМИ Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор): 0110212 от 08.02.1993.
Сетевое издание зарегистрировано как СМИ Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор): ЭЛ № ФС 77 - 84733 от 10.02.2023.