Review

Modeling rare pediatric neurogenetic disorders with IPSCs

  • Intensive research has been performed to identify the pathological mechanisms of many pediatric neurogenetic disorders and to identify potential therapeutic targets. Although research into many pediatric neurological disorders has provided tremendous insight into the mechanisms of disease, effective treatments remain elusive. A significant impediment to progress has been a lack of thorough disease models. Transgenic/knockout animal models have been very valuable in determining the mechanisms of many neurogenetic disorders; however, these models cannot always mimic human-specific pathology and can be inadequate in representing human pathogenesis. This can be especially true for diseases of the nervous system. Alternatively, human patient-derived nervous tissue can be dangerous to acquire and difficult to propagate. The development of patient-derived induced pluripotent stem cells (IPSCs) has given researchers a fresh means of modeling these disorders with renewable human cells that can be used to generate neurons and glia. IPSCs are somatic cells that are reprogrammed back to a pluripotent stage, which can provide an unlimited source of human cells possessing patient-specific genetic mutations. Their potential to be differentiated into any cell type enables them to be a flexible platform to investigate neurogenetic disease. Of course, efficient methods for differentiating IPSCs into homogeneous populations of somatic cells must be established to provide the “disease-in-a-dish” systems. We will discuss the current methods for generating IPSC-derived neural cells to model pediatric neurogenetic disorders, as well as provide examples of the disorders that have been studied that include several neurodevelopmental and neurodegenerative disorders (Rett syndrome, spinal muscular atrophy, hereditary spastic paraplegias, and leukodystrophies). In addition, we provide examples on how patient-specific neural cells can be used in therapeutic development with high-throughput drug screening platforms or with correction via genome editing.

    Citation: Jaemin Kim, David Nonis, Maria Gabriela Otero, Tyler Mark Pierson. Modeling rare pediatric neurogenetic disorders with IPSCs[J]. AIMS Cell and Tissue Engineering, 2019, 3(1): 1-25. doi: 10.3934/celltissue.2019.1.1

    Related Papers:

    [1] Xin Yun, Myung Hwan Chun . The impact of personalized recommendation on purchase intention under the background of big data. Big Data and Information Analytics, 2024, 8(0): 80-108. doi: 10.3934/bdia.2024005
    [2] Bill Huajian Yang . Modeling path-dependent state transitions by a recurrent neural network. Big Data and Information Analytics, 2022, 7(0): 1-12. doi: 10.3934/bdia.2022001
    [3] Antonio N. Bojanic . Accounting for the Trump factor in modeling the COVID-19 epidemic: the case of Louisiana. Big Data and Information Analytics, 2021, 6(0): 74-85. doi: 10.3934/bdia.2021006
    [4] Xing Tan, Giri Kumar Tayi .  CERTONTO: TOWARDS AN ONTOLOGICAL REPRESENTATION OF FAIR TRADE CERTIFICATION STANDARDS. Big Data and Information Analytics, 2017, 2(3&4): 255-264. doi: 10.3934/bdia.2017022
    [5] David E. Bernholdt, Mark R. Cianciosa, David L. Green, Kody J.H. Law, Alexander Litvinenko, Jin M. Park . Comparing theory based and higher-order reduced models for fusion simulation data. Big Data and Information Analytics, 2018, 3(2): 41-53. doi: 10.3934/bdia.2018006
    [6] Zhouchen Lin . A Review on Low-Rank Models in Data Analysis. Big Data and Information Analytics, 2016, 1(2): 139-161. doi: 10.3934/bdia.2016001
    [7] Xiaoying Chen, Chong Zhang, Zonglin Shi, Weidong Xiao . Spatio-temporal Keywords Queries in HBase. Big Data and Information Analytics, 2016, 1(1): 81-91. doi: 10.3934/bdia.2016.1.81
    [8] Dongyang Yang, Wei Xu . Statistical modeling on human microbiome sequencing data. Big Data and Information Analytics, 2019, 4(1): 1-12. doi: 10.3934/bdia.2019001
    [9] Weidong Bao, Wenhua Xiao, Haoran Ji, Chao Chen, Xiaomin Zhu, Jianhong Wu . Towards big data processing in clouds: An online cost-minimization approach. Big Data and Information Analytics, 2016, 1(1): 15-29. doi: 10.3934/bdia.2016.1.15
    [10] Tieliang Gong, Qian Zhao, Deyu Meng, Zongben Xu . Why Curriculum Learning & Self-paced Learning Work in Big/Noisy Data: A Theoretical Perspective. Big Data and Information Analytics, 2016, 1(1): 111-127. doi: 10.3934/bdia.2016.1.111
  • Intensive research has been performed to identify the pathological mechanisms of many pediatric neurogenetic disorders and to identify potential therapeutic targets. Although research into many pediatric neurological disorders has provided tremendous insight into the mechanisms of disease, effective treatments remain elusive. A significant impediment to progress has been a lack of thorough disease models. Transgenic/knockout animal models have been very valuable in determining the mechanisms of many neurogenetic disorders; however, these models cannot always mimic human-specific pathology and can be inadequate in representing human pathogenesis. This can be especially true for diseases of the nervous system. Alternatively, human patient-derived nervous tissue can be dangerous to acquire and difficult to propagate. The development of patient-derived induced pluripotent stem cells (IPSCs) has given researchers a fresh means of modeling these disorders with renewable human cells that can be used to generate neurons and glia. IPSCs are somatic cells that are reprogrammed back to a pluripotent stage, which can provide an unlimited source of human cells possessing patient-specific genetic mutations. Their potential to be differentiated into any cell type enables them to be a flexible platform to investigate neurogenetic disease. Of course, efficient methods for differentiating IPSCs into homogeneous populations of somatic cells must be established to provide the “disease-in-a-dish” systems. We will discuss the current methods for generating IPSC-derived neural cells to model pediatric neurogenetic disorders, as well as provide examples of the disorders that have been studied that include several neurodevelopmental and neurodegenerative disorders (Rett syndrome, spinal muscular atrophy, hereditary spastic paraplegias, and leukodystrophies). In addition, we provide examples on how patient-specific neural cells can be used in therapeutic development with high-throughput drug screening platforms or with correction via genome editing.


    1. Introduction

    Modern farming systems used worldwide to satisfy the increasing demand for food require the extensive use of chemical fertilizers, which are not only very costly but produce many environmental and human health problems [1]. Also, consumers have become more aware of different food production systems and food safety and quality [2]. In recent years, new production methods to ensure food quality have been investigated, addressing a key area within the European political agenda [3]. Due to general environmental awareness, these new methods are often in line with the ideology of green politics, and many countries have implemented these methods and have included them within their legislation.

    Currently, there are many studies that have analysed increased crop production following the use of several bacterial strains. And, there is also a significant number of studies that not only show increased total yield productions, but also the nutritional benefits achieved by the application of biofertilizers on plants and the role of bacteria in improving the quality of crops [4].

    Recently, the plant-associated microorganisms which cause beneficial effects have been defined as "Plant Probiotic" [5,6]. Plant probiotics are microorganisms that confer benefits to the health of plants when administered in specific amounts [7]. In addition, they belong to the bacterial group "Plant Growth Promoting Rhizobacteria" (PGPR) which are able to colonize plant roots [8,9], promote plant growth through different direct and indirect mechanisms [1,10], improve plant nutritional content [11,12] and increase crop quality [13,14].

    However, it is known that in order to achieve benefits to plants, the selection of efficient and specific PGPR is required. Therefore, the complex interactions between plants, bacteria and environmental factors need to be understood. Environment factors such as inorganic mineral content and plant exudates are associated with the growth and establishment of bacteria [15,16]. Thus, the microbial communities associated with plants depend on ecosystem development [16]. Understanding how microbial communities colonize plant roots [17] and the interactions and mechanisms of biofertilizers is essential if plant quality is to be improved [18]. Another key factor is to ensure the non-pathogenicity of the microorganisms used in biofertilization processes, with the aim to avoid problems to human health or the environment [19].

    Among current food guidelines, one of the new initiatives is based on the consumption of vegetables and fruits rich in bioactive substances. These substances are able to express anticarcinogenic effects and prevent various diseases and health disorders [20]. Consequently, the beneficial effects of bioactive substances on human health have been analysed in several studies [5,21]. Moreover, the high consumption of these types of foods is recommended for a healthy diet. For example, berries provide significant health benefits owing to the high level of vitamins, minerals and antioxidants they contain [20]. In addition, berries are readily available and can be consumed in a wide range of forms, such as fresh or frozen, and can be found in food products like jam and yogurt [22]. On the other hand, horticultural crops provide humans with a large variety of indispensable vitamins, such as A and C, and elements such as potassium [23]. According to Ramsay et al. [24], consumption of a variety of fruits and vegetables is linked to overall diet quality.

    This review focuses on improved crop quality after the application of plant probiotic bacteria as enhancers of the nutritional value of fruits and horticultural crops.


    2. Impact of Bacterial Probiotics on Plant Nutrient Content

    Vitamins are among the nutrients required for many physiological functions essential to life. Despite being presented in minute amounts on food, vitamins prevent specific deficiency syndromes which can affect people when there is an absence or a reduction of their contents [25]. Moreover, vitamin deficiency in humans can produce several diseases such as ocular surface abnormalities [26] or neurodegenerative problems [27]. Due to the importance of vitamins, one of the proposals presented by the World Health Organization (WHO) is to improve the content of essential vitamins in food in order to decrease worldwide malnutrition [28]. Therefore, scientists are searching for different ways to increase the total content of these types of substances in crops. Here, some of the studies in which bacterial plant probiotic inoculation has been used to increase the vitamin content of fruits and horticultural crops (Table 1) are presented.

    Table 1. Plant probiotic bacteria used to enhance the quality of fruit and horticultural crops.
    Bacterial strain Fruit or horticultural crop Crop effects caused Reference
    Phyllobacterium sp. PEPV15 Fragraria x ananassa Enhancement of vitamin C [5]
    Pseudomonas BA-8, Bacillus OSU-142 and Bacillus M-3 Fragraria x ananassa Enhancement of vitamin C [30]
    Paenibacillus polymyxa Fragraria x ananassa Enhancement of vitamin C [31]
    Pseudomonas sp. 19Fv1T Lycopersicon esculentum Enhancement of vitamin C [32]
    Bacillus megaterium and Bacillus amyloliquefaciens Lycopersicon esculentum Enhancement of vitamin C [33]
    Bacillus amyloliquefaciens FZB2 and B. amyloliquefaciens FZB42 Lycopersicon esculentum Enhancement of vitamin C [34]
    Pseudomonas sp. 5Vm1K and AMF Fragaria × ananassa Enhancement of vitamin B and C [36]
    Pseudomonas putida 41, Azotobacter chroococcum5, Azospirillum lipoferum OF and AMF Lycopersicon esculentum Increased antioxidant activity [52]
    Bacillus licheniformis Lycopersicon esculentum Improved total flavonoids content [14]
    Pseudomonas putida 41, Azotobacterchroococcum5, Azospirillum lipoferum OF Ocimum basilicum Increased antioxidant activity [42]
    Pseudomonas sp., Bacillus lentus and Azospirillum brasilens. Ocimum basilicum Increased antioxidant activity and chlorophyll leaf content [43]
    Rhizobium leguminosarum PETP01 Capsicum annuum Increased antioxidant activity [44]
    Pseudomonas fluorescens N21.4 Rubus sp. Increased flavonoids concentration [13]
    Rhizobium sp. PEPV12 Spinacia oleracea. Increase chlorophyll content [9]
    Paenibacillus polymyxa RC14 Brassica oleracea var capitata cv Yalova 1. N, P, K, S, Fe, and Cu content increase [12]
    Bacillus subtilis BA-142, Bacillus megaeorium-GC subgroup A. MFD-2, Acinetobacter baumannii CD-1 and Pantoea agglomerans FF Lycopersicon esculentum L. and Cucumis sativus L. N, P, Mg, Ca, Na, K, Cu, Mn, Fe and Zn content increase in both fruit [114]
    Bacillus subtilis EY2, Bacillus atrophaeus EY6, Bacillus spharicus GC subgroup B EY30, Staphylococcus kloosii EY37, and Kocuriaerythromyxa EY43 Fragaria x ananassa. N, P, K, Ca, Mg, S, Mn, Cu and Fe [115]
    Pseudomonas fluorescens and Bradyrhizobium sp. Origanum majorana Increase the amount of essentials oils [65]
    Bacillus megaterium, Pantoea agglomerans and Bacillus subtilis Brassica oleracea Increase chlorophyll plant content [116]
    Pseudomonas fluorescens N21.4 Rubus sp. var Lochness Increase and stabilize total flavonoid content during different season [79]
     | Show Table
    DownLoad: CSV

    Vitamin C is the vitamin with the highest recommended daily dose and L-Ascorbic acid (AsA) is derived from L-threo-hex-2-enono-1, 4-lactone. Some mammals cannot synthesize AsA due to the lack of functionality of the gene that encodes the enzyme which catalyses its biosynthesis [29]. The results obtained by Flores-Félix et al. [5] showed that vitamin C levels were significantly higher (79% compare to the control) in strawberries obtained from plants inoculated with the strain Phyllobacterium sp. PEPV15. Moreover, Pirlak et al. [30] compare the results obtained, depending on the type of inoculation, and report that the highest levels of vitamin C in strawberry were obtained by applying root and foliar application in comparison with the uninoculated treatment were higher. The strains used in these experiments were Pseudomonas BA-8, Bacillus OSU-142 and Bacillus M-3. Erturk et al. [31] also showed high levels of vitamin C content in strawberry fruits after plants were inoculated with Paenibacillus polymyxa RC05.

    Increased levels of vitamin C after bacterial treatment have also been described for vegetables. For instance, Bona et al. [30] shows that inoculation with the strain Pseudomonas sp. 19Fv1T not only enhances yield but also positively affects the concentration of vitamin C in tomato fruits in comparison with the control treatment. Additionally, Shen et al. [33] shows that vermicompost combined with plant probiotic Bacillus megaterium and Bacillus amyloliquefaciens also increases tomato yield and vitamin C contents. According to Gül et al. [34], the highest levels of vitamin C content in tomato fruits were obtained after the inoculation of two bacterial strains Bacillus amyloliquefaciens (FZB2 and FZB42) in different treatments.

    Berry crops are regarded as a good source of vitamins in addition to their anticarcinogenic and antimutagenic properties [22,35]. In this sense, Bona et al. [36] analyses the improvement of vitamin B9 and vitamin C content in strawberry fruits after the inoculation with arbuscular mycorrhizal fungi (AMF) and different strains of plant growth promoting bacteria (PGPB). They showed significant differences in ascorbic acid levels compared to the control after the inoculation with Pseudomonas sp. 5Vm1K, a mixture of AMF, and a co-inoculation formed by AMF and Pseudomonas sp. 5Vm1K. However, the vitamin B9 content was only significant higher after double co-inoculation with AMF and PGPB.

    Moreover, in the literature it is known that some vitamins also have antioxidants effects, being substances that interfere with normal oxidation processes, such as oxygen scavengers, promoting a more favourable anaerobic environment [37]. Thus, plant antioxidants are one of the most active food compounds [38] and are present in vegetables like tomato, carrot, cabbage, red beets and fruits as cocoa, prunes, red grapes and citrus fruits [39]. These substances can be classified into two different groups according to their mode of action: chemical substances which interrupt free radical chain propagation, and oxygen scavengers and chelators which bind to ions involved in the formation of free radicals [39].

    Here, we present some of the studies published on fruit and horticultural crops that have shown a significant increase in antioxidant activity after being inoculated with plant probiotic bacteria (Table 1).

    Tomato, one of the most cultivate vegetables in the world, is among the vegetables and horticultural used in these types of studies [40]. Moreover, tomatoes are regarded as an excellent source of antioxidant compounds [32,41]. Ochoa-Velasco et al. [14] compared the different levels of total antioxidant content in tomato fruits after biofertilization with Bacillus licheniformis and different doses of a nitrogen fertilizer. In general terms, they describe an improvement in the quality of tomato fruits with respect to total hydrophilic antioxidant compounds (vitamin C and total phenols) after inoculation with B. licheniformes and the reduction of the nitrogen doses.

    The bacterial effects on antioxidant activity have also been analysed in basil (Ocimum basilicum L.). This medicinal plant is used worldwide in cooking, and its antioxidant activity has been investigated after different treatments. The inoculation of basil with a mixture of Pseudomonas putida 41, Azotobacter chroococcum 5, Azospirillum lipoferum OF strains presented the highest level of antioxidant activity [42] as compared to the control treatment. In addition, another study involving basil reported that, under water stress, inoculation with three bacterial species (Pseudomonas sp., Bacillus lentus and Azospirillum brasilens) also increased antioxidant activity (Catalase and guaiacol peroxidise activity), as well as a significant change in leaf chlorophyll content [43].

    Peppers (Capsicum annuum L.) are also considered as an excellent dietary source of antioxidant compounds, in addition to being an economically important agricultural crop. In the study published by Silva et al. [44], pepper extracts were tested against 1, 1-Diphenyl-2-picrylhydrazy, superoxide (O2 •−), and nitric oxide (NO) radicals. The results showed a significant difference between the extract from the fruits of inoculated plants and the extracts from uninoculated plants. Also, higher antioxidant activity was found in peppers from plants inoculated with the strain Rhizobium leguminosarum PETP01.

    Antioxidant activity has also been analysed in fruits. And, strawberries have been found to be a good source of antioxidants. The anthocyanin content is in part responsible for the antioxidant power in berries [45]. Lingua et al. [46] reported that the co-inoculation of plant growth promoting bacterial strains and mycorrhizal fungi on strawberry plants produced the best levels of anthocyanin content in strawberries.

    Carotenoids are natural pigments which belong to the second group of antioxidants. They are involved in indispensable roles in photoprotection, stress and photosynthesis, in addition to acting as precursors of vitamin A [47,48]. Some carotenoids compounds have been reported to prevent humans from developing certain cancers and cardiovascular diseases. These compounds also protect plants against photooxidative damage and contribute to maintaining the plants overall structure.

    Lycopene is a red carotenoid that is an intermediate in the biosynthesis of other carotenoids, such as β-carotene [49], and is an effective free radical scavenger. Also, its presence in the human diet is correlated with a reduction in the incidence of cancer [3,50]. Lycopene is involved in the reddening of the tomato due to its function in the differentiation of chloroplasts and chromoplasts. In addition, lycopene is important in the final nutritional quality of this plant product [51]. With regard to the importance of carotenoids, during the past years scientists have being analyzing the way to improve their contents in crops by using fertilizers based on bacterial plant probiotics. Ordookhani et al. [52] concluded that the inoculation of tomato plants with Pseudomonas putida 41, Azotobacter chroococcum 5, Azospirillum lipoferum OF and a mixture of arbuscular mycorrhiza fungi (Glomus lipoferum, Glomus mossea and Glomus etunicatum) increased lycopene antioxidant levels. Moreover a positive correlation between lycopene and fruit potassium content was found in tomato plants after inoculation.

    In order to increase crop yield, large amounts of N and P are required and excessive amounts are applied to the soil, causing significant environmental pollution as a result of nitrate leaching [53]. To reduce these adverse effects, PGPR bacteria can be used as an alternative to chemical fertilizers as they can improve plant growth through plant nutrient uptake and thus improving crop production [54].

    PGPRs have direct mechanisms such as nitrogen fixation, phosphate solubilization, phytohormones production, among others, that facilitate the availability and absorption of nutrients [55,56].

    Plant growth promoting bacteria are used in numerous studies which also have such mechanisms as nitrogen fixing bacteria [57]. Consequently, the increase of N content may produce a greater uptake of nutrients present in the soil by the plant and an increase of the plant nutrient element (PNE) contents, improving the quality of the crops [58].

    Many studies use PGPR microorganisms to improve the quality of various crops by increasing the PNE content. For example, the increase in Ca content produces a more rigid peel when fruit is ripe. On the other hand, an increase in K content leads to an increase in the translocation of carbohydrates from the leaves to the fruit and, consequently, a higher fruit volume is obtained [59]. Some studies have evaluated the nutritional content of fruits and crops after inoculation with PGPR bacteria.

    Broccoli is a vegetable rich in dietary fiber, nutrients and several phytochemicals with proposed health-related properties [11]. Yildirim et al. [58] inoculated broccoli roots with the PGPR bacteria (Bacillus cereus, Brevibacillus reuszeri and Rhizobium rubi) to evaluate if they had any additional effect on crop growth and nutrient uptake. The results obtained from the inoculated plants showed an increase in chlorophyll, N, K, Ca, S, P, Mg, Fe, Mn, Zn and Cu contents. In addition, crop yield increased by 17.0%, 20.2% and 24.3% and the chlorophyll content by 14.7%, 14.0% and 13.7% in treatments inoculated with Bacillus cereus, Rhizobium rubi and Brevibacillus reuszeri, respectively.

    As mentioned above, tomato fruit is one of the most consumed vegetables in the world, either fresh or processed [60]. Sharafzadeh [61] performed a greenhouse test, inoculating tomato plants with different combinations of PGPRs such as Pseudomonas, Azotobacter and Azosprillum. The result obtained from 5 out of the 7 inoculated treatments showed an increase in the nutrient content of N, P, K, Ca and Mg.

    Vayssières et al. [62] obtained similar yields of banana crops in two experiments: a treatment with a nitrogen fertilizer (100%) and another treatment involving inoculation with PGPB and lower nitrogen fertilizer conditions (33%). The results showed that the inoculations with Azospirillum brasilense Sp7 and Bacillus sphericus UPMB10 improved fruit yield and fruit quality under the reduced fertilizer conditions. The nutrients which were shown to be increased in the treatments with the PGPB were N, P, K, Ca and Mg. Thus, it was concluded that these strains could be used as biofertilizers to improve banana production and quality.


    3. Effects of Plant Probiotics Bacteria on Flavonoids, Organic Acids and Volatile Compounds Levels

    Plants usually produce organic compounds as root exudates. These compounds are usually metabolites from different metabolic routes such as flavonoids, organic acids, sterols and volatile compounds [63]. They have diverse functions in plants like providing protection against pathogens [64,65,66,67] or abiotic stresses [68], as well as increasing the nutrient content of plants [69] and therefore their quality. The concentration of these metabolites in plant tissues varies depending on their biosynthesis, storage and degradation processes [63]. Furthermore, some of these compounds can be beneficial to human health when consumed in large quantities, thus they are important not only for the food industry but also for pharmaceutics companies [70].

    Flavonoids are the largest group of phenolic compounds that are abundantly present in plants, and are one of the major groups of plant secondary metabolites [71]. They are considered as having a wide range of biochemical and pharmacological properties. Recently, the analysis of flavonoid content in plants has received considerable attention due to fact that these substances possess biological properties, such as chemoprevention [72], and its role in the protection against diseases [73,74]. In addition, they are known to present antifungical, antibacterial and antiviral effects [75]. Considering all of the beneficial effects of flavonoids on human health, several research projects have been mentioned in the literature where scientists attempt to enhance flavonoid content by bacterial inoculation of crops with special economic interest.

    Since fruits are regarded as a good source of flavonoids, Basu and Maier [76] analysed the flavonoid content in different species of berries crops, showing that all of their antioxidant activity had the potential to improve human health. Blackberry is an important type of berry, which provides beneficial health effects [77]. García-Seco et al. [78] shows that the inoculation of blackberry plants with the strain Pseudomonas fluorescens N21.4 improves flavonoid content by 22%, as compared to the control treatment, during a specific period of the year (from July through October). Taking these results into account, Ramos-Solano et al. [79] analysed the effect of the bacterial probiotic inoculation on plant quality throughout the year and reports that the strain N21.4 tends to increase and stabilize total flavonoid content in blackberry crops, even when environmental conditions are adverse. Moreover, García-Seco et al. [13] reported that the blackberry root treatment with Pseudomonas fluorescens N21.4 not only increased the expression of flavonoids biosynthetic genes, but also flavonoid concentration in fruits. In the research published by Ghorbanpour et al. [80], several Pseudomonas fluorescens strains were also analysed with the intention of determining their role on plant growth and flavonoids production in other type of crops. They showed that the inoculation with the strain Pseudomonas fluorescens Ap14 induced a significantly higher flavonoid content in comparison with the uninoculated control treatment. Apart from enhancing the berries overall quality, bacterial probiotics have been reported to delay fungal growth [81].

    Beneficial effects in horticultural crops have also been shown. Silva et al. [44] showed that aqueous extracts of Capsicum annuum leaves, after the bacterial inoculation with the rhizobium strain TVP08, presented a significant acetylcholinesterease (AchE) inhibitory activity (with an important relevance in the treatment of Alzheimer's disease). This result could be explained by the presence of flavonoids.

    Moreover, improved total flavonoid content has been described in fruit and horticultural species, and also in other types of crops such as buckwheat (Fagopyrum esculentum Moench). Singh et al. [82] showed that buckwheat inoculated with Azospirillum spp. and Azotobacter spp. produces increased concentrations of flavonoid and phenolic contents.

    Organic acids play an important role in plant responses to nutrient stress, and their content in plants depends of their nutritional status [83]. Organic acids (glutamic, tartaric, quinic, malonic, malic, shikimic, a-ketoglutaric, pyruvic, citric, succinic and fumaric acids) are widely distributed in fruits (melon, grape, peach, orange, lemon) and vegetables (green and red pepper, tomato, lettuce and lamb's lettuce) and influence the nutritional state, quality food and organoleptic properties like aroma, color and flavor [69].

    Typically, roots exudates contain many organic acids such as acetate, malate, aconitate, lactate, fumarate, isocitrate, oxalate, citrate and succinate. These compounds are the primary anion components and make up a complex mixture of metal cations in solution; this in turn allows for the displacement of anions from the soil matrix and the mobilization of micronutrients like Zn and Cu into the rhizosphere [84].

    Citrate and malate are involved in nutrient acquisition, and form potent complexes with Fe in soil and induce the dissolution of unavailable insoluble ferric oxyhydroxides. Moreover, they are the primary components released into the rhizosphere under P deficiency [84]. It has been shown that some PGPR can promote plant growth by capturing nutrients, since these bacteria produce an increase of organic acids in the roots of plants. For example, in strawberry crops the application of Bacillus and Pseudomonas strains produce an increase in plant biomass and nutrient content by the production of organic acids from bacteria [85], and in the same way in raspberry by the application of Bacillus [86], and in tomato, cucumber and sweet pepper by the application of Pseudomonas [87].

    Fatty acids are among the organic compounds produced by root exudates and these compounds are mainly used for energy storage [88]. Some of them are not produced by the human body and must be introduced into the diet by eating vegetables (potato, cabbages, wheal and chive) and fruits (lemon, strawberry and orange) [89]. The importance of consuming these compounds is based on their biological properties which has been shown to have positive effects against cardiovascular diseases, arthritis and triglyceride levels. Cucurbit, strawberry and tomato are a good source of bioactive compounds and have an excellent nutritional composition, containing fatty acids among other compounds with antioxidants effects [90]. Some plant oils commonly used for human consumption have unsaturated fatty acids like linolenic, linoleic and oleic acid and saturated ones such us palmitic and stearic [88,91]. Also, pumpkin seed oil has high nutritional value due to containing fatty acids like oleic, linolenid, palmitic and stearic. The application of phosphate solubilizing bacteria (Pseudomonas putida and Bacillus lentus) and nitrogen fixing bacteria (Azotobacter sp. and Azospirillum sp.) in pumpkin plants has been lead to an increase in oil, seed and fruit yield, especially the fatty acid content [92].

    On the other hand, plants sterols are essential bioactive components of cell membranes which are present in high concentrations in vegetable oils [93]. Furthermore, the presence of these organic compounds in plants is related to protective effects against abiotic stresses [68]. These organic compounds are usually released in plant root exudates; the most abundant are campestrol, stigmasterol and sitoesterol and are found in the fruits and vegetables that make up the human diet [94]. The consumption of plants sterols may have some health benefits, like reducing the risk of cardiovascular diseases and protecting against different types of cancer [91]. It has been reported in the literature that the application of PGPR can produce an increment in the levels of sterols in plants. Silva et al. [95] inoculated two strains of Rhizobium (TVP08 and PEPT01) in pepper (Capsicum annuum L) and evaluated their effect on sterols. The inoculation of Rhizobium produced a positive effect on the ripening of the pepper fruit, in addition to an improvement in several primary and secondary metabolites, which improved the nutritional value of the plant.

    Other organic substances which are common components of plants are volatile compounds. These can be found in small amounts in plants and are related to flowers, fruits, leaves, roots and vegetative tissues [96]. These compounds can act as a plant defense response against pathogens and predators [66]. Plants have control mechanisms that regulate the substances and the moment of the volatiles compounds synthesis. Their production can be influenced by the interaction with environmental microorganisms [96]. The application of rhizobacteria that promote plant growth can trigger an increase in the amount of volatile compounds, improving their qualities for the biological control of diseases [67]. In this respect, it has been shown that Bacillus [67,97,98] and Pseudomonas strains [90] can produce the inhibition of plant pathogens by the increment of volatile organic compounds in plants.

    Essential oils are among the group of volatile compounds and are intensely aromatic compounds biosynthesized by plants [99]. It is known that soil microorganisms can modify the secondary metabolic routes of plants, influencing the synthesis of essential oils that are of great importance for the food and pharmaceutical industries [46]. The most volatile compounds contained in Origanum majorana L. are essential oils and have an important economic interest because of their use as flavoring, fragrances, fungicides and insecticides. Some authors have determined the effects of root colonization by PGPRs on the composition and amount of essential oils in different crops, and the inoculation of Origanum majorana with P. fluorescens and Bradyrhizobium sp. [65] and the inoculation of peppermint (Mentha piperita) with P. fluorescens [100] produces an increase in the total essential oil content without modifying its composition.

    Additionally, the production of antimicrobial compounds by plant probiotic bacteria should be highlighted. Many studies have reported that some PGPR strains are also able to produce these types of molecules, which have been shown to be effective against pest and pathogens and effective as biocontrol agents [101]. According to Zhen et al. [102], several Bacillus strains have an important effect on suppressing pathogens and promoting plant growth due to antagonistic relationships and the enhancement of the host's nutritional content and growth [103]. Antibiosis processes and an induction of systematic resistance (ISR) have also been reported to have effects on plant health [104]. The production of several organic acids, such as citric and ascorbic, which are strong antimicrobial agents, has been reported to be efficient at fighting against pathogens in vegetables and fruits due to a reduction in pH and anion accumulation [64]. Therefore, based on their mode of action and effects, many strains can also be used as biocontrol agents, which greatly enhance food quality and growth [102].


    4. Situation, Characteristics and Problems Concerning the Current Commercialization of Plant Probiotic Bacterial Products

    In addition to the many bacterial inoculants that have already been used to treat legume crops, the number of commercial products for other types of crops is increasing every year [1]. However the development and commercialization of new inoculants and formulas require governmental approval and must fulfill several requirements that are not always general and well established. Moreover, they depend on where the new product is registered [105].

    In spite of being a worldwide process, one that is increasing year by year, there are no specific standards established for the creation and development of green products that can be used to reduce the amount of chemical fertilizers used, according to the new politics initiatives and programs (e.g. HORIZON 2020) [1]. Thus, the laws vary among countries and to date there is no international agreement regarding the quality control of biofertilizers [105]. Many countries have developed governmental policies for the use of biofertilizers, however, specifications to regulate such new products have not yet been established [3]. In this context, collaborations between researchers and industries are a key factor for establishing the basis of a worldwide biofertilizer market [1].

    Suitable inoculants should be safe and nontoxic for environment and humans [106,107,108]. Herrmann et al. [109] shows that 64% of the commercial biofertilizers analyzed contained one or several contaminant strains, as well as human pathogens. Findings such as these highlight the need for better quality control of the inoculants that make up biofertilizers.

    Moreover, in spite of the successful results so far achieved in the numerous studies involving plant growth promotion under controlled laboratories and greenhouses conditions, there have been variations within the results obtained in field trails. These differences can be explained by the abiotic and biotic conditions that can influence bacteria in the soil, as quite often bacterial inoculants are unable to survive in some specific niches [110]. Thus, according to Quin et al. [16] microbial plant communities fluctuate and depend on the state of the surrounding ecosystem.

    Inoculant formulas should include the most effective plant growth promoting bacterial strains [111] and be designed to provide a high number of viable cells. Also, the presence of these strains should not become rapidly diminished [108]. The bacteria need to be able to survive in a broad spectrum of conditions, since crops are grown under a multiplicity of environmental and difficult conditions [107]. Thus, over the years, researchers have been working to develop new bacterial products active in a wide variety of weather conditions that can cause a difference in the benefits provided by the bacterial inoculants [112]. In this sense, according to Lesueur et al. [108], there are four types of products depending on the carrier material used: granules, liquids, slurries and powders. Herrmann and Lesueur [113] established the importance of a suitable carrier as a key factor to ensure bacterial protection against environmental conditions, microorganism survival and their establishment and action after the inoculation of crops.

    Nowadays, despite all of the requirements and specifications, a large variety of bacterial inoculants, based on plant probiotic bacteria that enhances the quality of fruit and horticultural crops, are commercialized worldwide. This part of the review focuses on presenting some of the best known companies or sold products (Table 2). Nevertheless, the effects on food quality by the majority of commercialized bacterial products on sale today are not detailed or analyzed yet.

    Table 2. Commercial quality enhancing products used on fruit and horticultural crops.
    Product Company Crop type Effect caused
    Pseudo-Guard Bio Fungicide ® BioFix Horticultural crops Enhanced food quality
    Ajay Azospirillum® Ajay Bio-tech Vegetables, cash and fruit crops Enhanced chlorophyll content and enhanced vitamin, auxin, gibberellin and cytokinin production
    Ajay Azo® Ajay Bio-tech Vegetable and horticulture crops Enhanced auxin production
    Ajay Meal® Ajay Bio-tech Fruit crops and vegetables Enhanced overall quality
    Inogro® Flozyme corporation Horticultural crops Enhanced plant nutritional content
    Azotobacter Culture® Guajart State Fertilizers & Chemicals LTD Banana, cash and horticultural crops Enhanced plant nutritional content
    Azospirillum Culture ® Guajart State Fertilizers & Chemicals LTD Banana, cash and horticultural crops Enhance nutritional plant content
    Prabha Zinc® Prabhat Fertilizer & Chemical Works Horticultural crops Enhanced zinc content
    Bonasol® Abiosa Tomato, pepper and chilli crops Enhanced plant nutritional content
    EM-1 Microbial inoculants® TeraGanix All types of vegetables and trees Improved product quality
    Bio-TilisTM Agrovergel Horticultural crops Enhanced crop quality
     | Show Table
    DownLoad: CSV

    Due to the beneficial effects of fruit and vegetable crops on human health, the company "Guajart State Fertilizers & Chemical LTD" commercializes Azotobacter and Azospirillum culture®, a mixture of bacterial strains, to enhance the nutrient plant content in cash and horticultural crops.

    Among the biofertilizers, useful for the production of horticultural crops such as tomatoes, Inogro® is produced and commercialized by the company "Flozyme Corporation" (USA). Inogro® is a mix of more than 30 bacterial species that not only enhance plant growth promotion and crop production, but also improves plant nutrient content.

    One of the best results found in the improvement of tomato and pepper yields is achieved using Bonasol®, sold and produced by the company "Abiosa". This product is based on a mixture of bacterial and arbuscular mycorrhizal fungi. Bonasol® improves phosphorus and potassium availability to the plants and enhances the plants' final nutritional state.

    India is one of the countries whose government is making a large effort to try to increase the application of biofertilizers in their current farming system, and this has led to the development of companies such as Ajay Bio-tech (India) LDT. The company "Ajay Bio-tech (India) LTD" is one of the most important in India [105], and offers different quality enhancing products (Ajay Azo®, Ajay Azospirillum® and Ajay Meal®) used on many fruit and horticultural crops. These products are based on nitrogen-fixing or phosphate, and potassium-solubilising bacteria single or combined. Many of these strains enhance the uptake of indispensable nutrients by the plant, and overall plant nutrient content.

    Thus, it is very important that the use of biofertilizers and the changes in traditional farming systems are approved by farmers worldwide. This is a key step for encouraging the commercialization and production of green products and will help to secure the foundations of a solid biofertilizer market [105].


    5. Conclusions and Future Research Perspectives

    Unlike chemical fertilizers, the application of plant probiotic bacteria as green products in the field enhances the nutritional quality of fruits and vegetables. Therefore, the reduced use of chemical fertilizers in favor of products based on probiotic bacteria is a good alternative, not only to reduce the overuse of chemical fertilizer but also to ensure food quality. It has been reported on many occasions how the application of green products enhances crop production, reduces pathogen and pest attacks and increases plant nutrient content. However, to improve the use of biofertilizers many changes need to be made. This includes establishing more effective quality control systems that ensure commercial products are safely made and with nontoxic bacteria. As regards green products need to be designed using suitable carriers that allow these products to be successful in a broad spectrum of environmental conditions.

    In this review, we have described several cases of plant probiotic bacteria that when used as biofertilizers act to promoter plant growth, produce increases in yield and improve some parameters associated with food quality. According to some recent studies, bacterial plant probiotics are able to establish a relationship with fruit and horticultural crops and as a result of this interaction some plant compounds, which are beneficial to human health, have an increased production. On one hand, as outlined above, the market for bacterial fertilizers is continuously growing, but the effects of these commercialized bacteria on food quality parameters are not detailed. Therefore, more studies on the effect of improved food quality caused by different types of bacteria are necessary. This would allow for the better selection of plant probiotic bacteria with potential applications in agricultural practices orientated toward the production of high quality fruits and vegetables.

    On the other hand, it has been reported how the application of plant probiotic bacteria improve the quality of fruit and vegetables by increasing vitamins, flavonoids and antioxidants content, among other benefits. However, there are other functional food compounds that can also be potentially improved by bacterial inoculants. In the future, new research approaches such as metabolomic studies, comparing fruits and vegetables grown with and without the application of plant probiotic bacteria, may reveal additional beneficial effects on the quality of food crops from the applications of these types of bacteria.


    Acknowledgments

    The authors are grateful for the funding received by the Regional Government of Castile and Leon and the Spanish Ministry of Economics and Competitiveness (JCyL SA169U14 and MINECO; AGL2015-70510-R). AJG was funded by the FPU predoctoral fellowship from the Spanish Government. The authors also thank Emma Keck for the English language editing.


    Conflict of Interest

    The authors declare that they have no conflict of interest.




    [1] Bürk K (2017) Friedreich Ataxia: current status and future prospects. Cerebellum Ataxias.
    [2] Leonard H, Cobb S, Downs J (2016) Clinical and biological progress over 50 years in Rett syndrome. Nat Rev Neurol 13: 37–51.
    [3] Auburger G, Klinkenberg M, Drost J, et al. (2012) Primary Skin Fibroblasts as a Model of Parkinson's Disease. Mol Neurobiol 46: 20–27. doi: 10.1007/s12035-012-8245-1
    [4] Petters RM, Sommer JR (2000) Transgenic animals as models for human disease. Transgenic Res 9: 347–351. doi: 10.1023/A:1008926303533
    [5] Hargus G, Ehrlich M, Hallmann AL, et al. (2014) Human stem cell models of neurodegeneration: A novel approach to study mechanisms of disease development. Acta Neuropathol 127: 151–173. doi: 10.1007/s00401-013-1222-6
    [6] Ilic D, Ogilvie C (2017) Concise Review: Human Embryonic Stem Cells-What Have We Done? What Are We Doing? Where Are We Going? Stem Cells 35: 17–25.
    [7] Randolph LN, Jiang Y, Lian X (2017) Stem Cell Engineering and Differentiation for Disease Modeling and Cell-based Therapies. AIMS Cell Tissue Eng 1: 140–157. doi: 10.3934/celltissue.2017.2.140
    [8] Takahashi K, Yamanaka S (2006) Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell 126: 663–676. doi: 10.1016/j.cell.2006.07.024
    [9] Takahashi K, Tanabe K, Ohnuki M, et al. (2007) Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell 131: 861–872. doi: 10.1016/j.cell.2007.11.019
    [10] Poon A, Zhang Y, Chandrasekaran A, et al. (2017) Modeling neurodegenerative diseases with patient-derived induced pluripotent cells: Possibilities and challenges. N Biotechnol 39: 190–198. doi: 10.1016/j.nbt.2017.05.009
    [11] Tabar V, Studer L (2014) Pluripotent stem cells in regenerative medicine: Challenges and recent progress. Na Rev Genet 15: 82–92. doi: 10.1038/nrg3563
    [12] Mertens J, Marchetto MC, Bardy C, et al. (2016) Evaluating cell reprogramming, differentiation and conversion technologies in neuroscience. Nat Rev Neurosci 17: 424–437. doi: 10.1038/nrn.2016.46
    [13] Lancaster M, Renner M, Martin C, et al. (2013) Cerebral organoids model human brain development and microcephaly. Nature 501: 373–379. doi: 10.1038/nature12517
    [14] Pasca AM, Sloan SA, Clarke LE, et al. (2015) Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat Methods 12: 671–678. doi: 10.1038/nmeth.3415
    [15] Soldner F, Laganière J, Cheng AW, et al. (2011) Generation of Isogenic Pluripotent Stem Cells Differing Exclusively at Two Early Onset Parkinson Point Mutations. Cell 146: 318–331. doi: 10.1016/j.cell.2011.06.019
    [16] Bassett AR (2017) Editing the genome of hiPSC with CRISPR/Cas9: disease models. Mamm Genome 28: 348–364. doi: 10.1007/s00335-017-9684-9
    [17] Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. (1998) Embryonic stem cell lines derived from human blastocysts. Science 282: 1145–1147.
    [18] Wilmut I, Schnieke AE, McWhir J, et al. (1997) Viable offspring derived from fetal and adult mammalian cells. Nature 385: 810–813. doi: 10.1038/385810a0
    [19] Cowan CA, Atienza J, Melton DA, et al. (2005) Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells. Science 309: 1369–1373. doi: 10.1126/science.1116447
    [20] Yu J, Vodyanik MA, Smuga-Otto K, et al. (2007) Induced Pluripotent Stem Cell Lines Derived from Human Somatic Cells. Science 318: 1917–1920. doi: 10.1126/science.1151526
    [21] Okita K, Matsumura Y, Sato Y, et al. (2011) A more efficient method to generate integration-free human iPS cells. Nat Methods 8: 409. doi: 10.1038/nmeth.1591
    [22] Daley GQ (2014) Dissecting reprogramming, differentiation and conversion with network biology. Leuk Suppl 3: S3–S4. doi: 10.1038/leusup.2014.2
    [23] Zhou W, Freed CR (2009) Adenoviral Gene Delivery Can Reprogram Human Fibroblasts to Induced Pluripotent Stem Cells. Stem Cells 27: 2667–2674. doi: 10.1002/stem.201
    [24] Zhou Y, Zeng F (2013) Integration-free Methods for Generating Induced Pluripotent Stem Cells. Genom Proteom Bioinform 11: 284–287. doi: 10.1016/j.gpb.2013.09.008
    [25] Nakanishi M, Otsu M (2012) Development of Sendai Virus Vectors and their Potential Applications in Gene Therapy and Regenerative Medicine. Curr Gene Ther 12: 410–416. doi: 10.2174/156652312802762518
    [26] Kim D, Kim CH, Moon JI, et al. (2009) Generation of Human Induced Pluripotent Stem Cells by Direct Delivery of Reprogramming Proteins. Cell Stem Cell 4: 472–476. doi: 10.1016/j.stem.2009.05.005
    [27] Okita K, Yamanaka S (2011) Induced pluripotent stem cells: opportunities and challenges. Philos Trans R Soc B Biol Sci 366: 2198–2207. doi: 10.1098/rstb.2011.0016
    [28] Paridaen JT, Huttner WB (2014) Neurogenesis during development of the vertebrate central nervous system. EMBO Rep 15: 351–364. doi: 10.1002/embr.201438447
    [29] Chandrasekaran A, Avci HX, Ochalek A, et al. (2017) Comparison of 2D and 3D neural induction methods for the generation of neural progenitor cells from human induced pluripotent stem cells. Stem Cell Res 25: 139–151. doi: 10.1016/j.scr.2017.10.010
    [30] Chuang JH (2015) Neural differentiation from embryonic stem cells in vitro : An overview of the signaling pathways. World J Stem Cells 7: 437. doi: 10.4252/wjsc.v7.i2.437
    [31] Chambers SM, Fasano CA, Papapetrou EP, et al. (2009) Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol 27: 275–280. doi: 10.1038/nbt.1529
    [32] Qi Y, Zhang XJ, Renier N, et al. (2017) Combined small-molecule inhibition accelerates the derivation of functional cortical neurons from human pluripotent stem cells. Nat Biotechnol 35: 154–163. doi: 10.1038/nbt.3777
    [33] Kim DS, Jung SJ, Lee JS, et al. (2017) Rapid generation of OPC-like cells from human pluripotent stem cells for treating spinal cord injury. Exp Mole Med 49: e361. doi: 10.1038/emm.2017.106
    [34] Wang S, Bates J, Li X, et al. (2013) Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell 12: 252–264. doi: 10.1016/j.stem.2012.12.002
    [35] Xu XH, Zhong Z (2013) Disease modeling and drug screening for neurological diseases using human induced pluripotent stem cells. Acta Pharmacol Sin 34: 755–764. doi: 10.1038/aps.2013.63
    [36] Lojewski X, Staropoli JF, Biswas-legrand S, et al. (2014) Human iPSC models of neuronal ceroid lipofuscinosis capture distinct effects of TPP1 and CLN3 mutations on the endocytic pathway. Hum Mol Genet 23: 2005–2022. doi: 10.1093/hmg/ddt596
    [37] Espuny-Camacho I, Michelsen KA, Linaro D, et al. (2018) Human Pluripotent Stem-Cell-Derived Cortical Neurons Integrate Functionally into the Lesioned Adult Murine Visual Cortex in an Area-Specific Way. Cell Rep 23: 2732–2743. doi: 10.1016/j.celrep.2018.04.094
    [38] Dwyer ND, Chen B, Chou SJ, et al. (2016) Neural Stem Cells to Cerebral Cortex: Emerging Mechanisms Regulating Progenitor Behavior and Productivity. J Neurosci 36: 11394–11401. doi: 10.1523/JNEUROSCI.2359-16.2016
    [39] Rigamonti A, Repetti GG, Sun C, et al. (2016) Large-Scale Production of Mature Neurons from Human Pluripotent Stem Cells in a Three-Dimensional Suspension Culture System. Stem cell Rep 6: 993–1008. doi: 10.1016/j.stemcr.2016.05.010
    [40] Gunhanlar N, Shpak G, van der Kroeg M, et al. (2018) A simplified protocol for differentiation of electrophysiologically mature neuronal networks from human induced pluripotent stem cells. Mol Psychiatry 23: 1336–1344. doi: 10.1038/mp.2017.56
    [41] Alcamo EA, Chirivella L, Dautzenberg M, et al. (2008) Satb2 regulates callosal projection neuron identity in the developing cerebral cortex. Neuron 57: 364–377. doi: 10.1016/j.neuron.2007.12.012
    [42] Britanova O, de Juan Romero C, Cheung A, et al. (2008) Satb2 is a postmitotic determinant for upper-layer neuron specification in the neocortex. Neuron 57: 378–392. doi: 10.1016/j.neuron.2007.12.028
    [43] Shi Y, Kirwan P, Livesey FJ (2012) Directed differentiation of human pluripotent stem cells to cerebral cortex neurons and neural networks. Nat Protoc 7: 1836–1846. doi: 10.1038/nprot.2012.116
    [44] Chen B, Wang SS, Hattox AM, et al. (2008) The Fezf2-Ctip2 genetic pathway regulates the fate choice of subcortical projection neurons in the developing cerebral cortex. Proc Nat Acad Sci 105: 11382–11387. doi: 10.1073/pnas.0804918105
    [45] Espuny-Camacho I, Michelsen KA, Gall D, et al. (2013) Pyramidal neurons derived from human pluripotent stem cells integrate efficiently into mouse brain circuits in vivo. Neuron 77: 440–456. doi: 10.1016/j.neuron.2012.12.011
    [46] Elkabetz Y, Panagiotakos G, Al Shamy G, et al. (2008) Human ES cell-derived neural rosettes reveal a functionally distinct early neural stem cell stage. Genes Dev 22: 152–165. doi: 10.1101/gad.1616208
    [47] Paşca SP, Portmann T, Voineagu I, et al. (2011) Using iPSC-derived neurons to uncover cellular phenotypes associated with Timothy syndrome. Nat Med 17: 1657–1662. doi: 10.1038/nm.2576
    [48] Davis-Dusenbery BN, Williams LA, Klim JR, et al. (2014) How to make spinal motor neurons. Development 141: 491–501. doi: 10.1242/dev.097410
    [49] Santiago C, Bashaw GJ (2014) Transcription factors and effectors that regulate neuronal morphology. Development 141: 4667–4680. doi: 10.1242/dev.110817
    [50] Stifani N (2014) Motor neurons and the generation of spinal motor neuron diversity. Front Cell Neurosci 8: 293.
    [51] Du ZW, Chen H, Liu H, et al. (2015) Generation and expansion of highly pure motor neuron progenitors from human pluripotent stem cells. Nat Commun 6: 6626. doi: 10.1038/ncomms7626
    [52] Hester ME, Murtha MJ, Song S, et al. (2011) Rapid and efficient generation of functional motor neurons from human pluripotent stem cells using gene delivered transcription factor codes. Mol Ther 19: 1905–1912. doi: 10.1038/mt.2011.135
    [53] Di Giorgio FP, Boulting GL, Bobrowicz S, et al. (2008) Human Embryonic Stem Cell-Derived Motor Neurons Are Sensitive to the Toxic Effect of Glial Cells Carrying an ALS-Causing Mutation. Cell Stem Cell 3: 637–648. doi: 10.1016/j.stem.2008.09.017
    [54] Amoroso MW, Croft GF, Williams DJ, et al. (2013) Accelerated high-yield generation of limb-innervating motor neurons from human stem cells. J Neurosci 33: 574–586. doi: 10.1523/JNEUROSCI.0906-12.2013
    [55] Karumbayaram S, Novitch BG, Patterson M, et al. (2009) Directed differentiation of human-induced pluripotent stem cells generates active motor neurons. Stem Cells 27: 806–811. doi: 10.1002/stem.31
    [56] Toma JS, Shettar BC, Chipman PH, et al. (2015) Motoneurons derived from induced pluripotent stem cells develop mature phenotypes typical of endogenous spinal motoneurons. J Neurosci 35: 1291–1306. doi: 10.1523/JNEUROSCI.2126-14.2015
    [57] Pepper JP, Wang TV, Hennes V, et al. (2017) Human induced pluripotent stem cell–derived motor neuron transplant for neuromuscular atrophy in a mouse model of sciatic nerve injury. JAMA Facial Plast Surg 19: 197. doi: 10.1001/jamafacial.2016.1544
    [58] Chandrasekaran A, Avci HX, Leist M, et al. (2016) Astrocyte differentiation of human pluripotent stem cells: New tools for neurological disorder research. Front Cell Neurosci 10: 215.
    [59] Santos R, Vadodaria KC, Jaeger BN, et al. (2017) Differentiation of inflammation-responsive astrocytes from glial progenitors generated from human induced pluripotent stem cells. Stem Cell Rep 8: 1757–1769. doi: 10.1016/j.stemcr.2017.05.011
    [60] TCW J, Wang M, Pimenova AA, et al. (2017) An Efficient Platform for Astrocyte Differentiation from Human Induced Pluripotent Stem Cells. Stem Cell Rep 9: 600–614. doi: 10.1016/j.stemcr.2017.06.018
    [61] Liu H, Zhang SC (2011) Specification of neuronal and glial subtypes from human pluripotent stem cells. Cell Mol Life Sci 68: 3995–4008. doi: 10.1007/s00018-011-0770-y
    [62] Krencik R, Weick JP, Liu Y, et al. (2011) Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nat Biotechnol 29: 528–534. doi: 10.1038/nbt.1877
    [63] Jiang P, Chen C, Liu XB, et al. (2016) Human iPSC-Derived Immature Astroglia Promote Oligodendrogenesis by Increasing TIMP-1 Secretion. Cell Rep 15: 1303–1315. doi: 10.1016/j.celrep.2016.04.011
    [64] Nistor GI, Totoiu MO, Haque N, et al. (2005) Human embryonic stem cells differentiate into oligodendrocytes in high purity and myelinate after spinal cord transplantation. Glia 49: 385–396. doi: 10.1002/glia.20127
    [65] Hu BY, Du ZW, Zhang SC (2009) Differentiation of human oligodendrocytes from pluripotent stem cells. Nat Protoc 4: 1614–1622. doi: 10.1038/nprot.2009.186
    [66] Goldman SA, Kuypers NJ (2015) How to make an oligodendrocyte. Development 142: 3983–3995. doi: 10.1242/dev.126409
    [67] Doerr J, Böckenhoff A, Ewald B, et al. (2015) Arylsulfatase A Overexpressing Human iPSC-derived Neural Cells Reduce CNS Sulfatide Storage in a Mouse Model of Metachromatic Leukodystrophy. Mol Ther 23: 1519–1531. doi: 10.1038/mt.2015.106
    [68] Pouya A, Satarian L, Kiani S, et al. (2011) Human induced pluripotent stem cells differentiation into oligodendrocyte progenitors and transplantation in a rat model of optic chiasm demyelination. PLoS One 6: e27925. doi: 10.1371/journal.pone.0027925
    [69] Lippmann ES, Azarin SM, Kay JE, et al. (2012) Derivation of blood-brain barrier endothelial cells from human pluripotent stem cells. Na Biotechnol 30: 783–791. doi: 10.1038/nbt.2247
    [70] Appelt-Menzel A, Cubukova A, Günther K, et al. (2017) Establishment of a human blood-brain barrier co-culture model mimicking the neurovascular unit using induced pluri- and multipotent stem cells. Stem Cell Rep 8: 894–906. doi: 10.1016/j.stemcr.2017.02.021
    [71] Busskamp V, Lewis NE, Guye P, et al. (2014) Rapid neurogenesis through transcriptional activation in human stem cells. Mol Syst Biol 10: 760–760. doi: 10.15252/msb.20145508
    [72] Zhang Y, Pak CH, Han Y, et al. (2013) Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78: 785–798. doi: 10.1016/j.neuron.2013.05.029
    [73] Wang C, Ward ME, Chen R, et al. (2017) Scalable Production of iPSC-Derived Human Neurons to Identify Tau-Lowering Compounds by High-Content Screening. Stem Cell Rep 9: 1221–1233. doi: 10.1016/j.stemcr.2017.08.019
    [74] Fernandopulle MS, Prestil R, Grunseich C, et al. (2018) Transcription Factor–Mediated Differentiation of Human iPSCs into Neurons. Curr Protoc Cell Biol 79: e51. doi: 10.1002/cpcb.51
    [75] Li X, Tao Y, Bradley R, et al. (2018) Fast Generation of Functional Subtype Astrocytes from Human Pluripotent Stem Cells. Stem Cell Rep 11: 998–1008. doi: 10.1016/j.stemcr.2018.08.019
    [76] Ehrlich M, Mozafari S, Glatza M, et al. (2017) Rapid and efficient generation of oligodendrocytes from human induced pluripotent stem cells using transcription factors. Proc Natl Acade Sci U S A 114: E2243–E2252. doi: 10.1073/pnas.1614412114
    [77] García-León JA, Kumar M, Boon R, et al. (2018) SOX10 Single Transcription Factor-Based Fast and Efficient Generation of Oligodendrocytes from Human Pluripotent Stem Cells. Stem Cell Rep 10: 655–672. doi: 10.1016/j.stemcr.2017.12.014
    [78] Ardhanareeswaran K, Mariani J, Coppola G, et al. (2017) Human induced pluripotent stem cells for modelling neurodevelopmental disorders. Nat Rev Neurol 13: 265–278. doi: 10.1038/nrneurol.2017.45
    [79] Shovlin S, Tropea D (2018) Transcriptome level analysis in Rett syndrome using human samples from different tissues. Orphanet J Rare Dis 13: 113. doi: 10.1186/s13023-018-0857-8
    [80] Chahrour M, Zoghbi HY (2007) The Story of Rett Syndrome: From Clinic to Neurobiology. Neuron 56: 422–437. doi: 10.1016/j.neuron.2007.10.001
    [81] Huidobro C, Fernandez AF, Fraga MF (2013) The role of genetics in the establishment and maintenance of the epigenome. Cell Mol Life Sci 70: 1543–1573. doi: 10.1007/s00018-013-1296-2
    [82] Kishi N, Macklis JD (2004) MECP2 is progressively expressed in post-migratory neurons and is involved in neuronal maturation rather than cell fate decisions. Mol Cell Neurosci 27: 306–321. doi: 10.1016/j.mcn.2004.07.006
    [83] Damen D, Heumann R (2013) MeCP2 phosphorylation in the brain: From transcription to behavior. Biol Chem 394: 1595–1605.
    [84] Hendrich B, Bird A (1998) Identification and Characterization of a Family of Mammalian Methyl-CpG Binding Proteins. Mol Cell Biol 18: 6538–6547. doi: 10.1128/MCB.18.11.6538
    [85] Gold WA, Krishnarajy R, Ellaway C, et al. (2018) Rett Syndrome: A Genetic Update and Clinical Review Focusing on Comorbidities. ACS Chem Neurosci 9: 167–176. doi: 10.1021/acschemneuro.7b00346
    [86] Ausió J (2018) Role of MeCP2 in neurological disorders: current status and future perspectives. Epigenomics 10: 5–8. doi: 10.2217/epi-2017-0128
    [87] Linda K, Fiuza C, Nadif Kasri N (2018) The promise of induced pluripotent stem cells for neurodevelopmental disorders. Prog Neuropsychopharmacol Biol Psychiatry 84: 382–391. doi: 10.1016/j.pnpbp.2017.11.009
    [88] Marchetto MC, Carromeu C, Acab A, et al. (2010) A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell 143: 527–539. doi: 10.1016/j.cell.2010.10.016
    [89] Kim KY, Hysolli E, Park IH (2011) Neuronal maturation defect in induced pluripotent stem cells from patients with Rett syndrome. Proc Natl Acad Sci U S A 108: 14169–14174. doi: 10.1073/pnas.1018979108
    [90] Farra N, Zhang WB, Pasceri P, et al. (2012) Rett syndrome induced pluripotent stem cell-derived neurons reveal novel neurophysiological alterations. Mol Psychiatry 17: 1261–1271. doi: 10.1038/mp.2011.180
    [91] Cheung AY, Horvath LM, Grafodatskaya D, et al. (2011) Isolation of MECP2-null Rett Syndrome patient hiPS cells and isogenic controls through X-chromosome inactivation. Hum Mol Genet 20: 2103–2115. doi: 10.1093/hmg/ddr093
    [92] Djuric U, Cheung AYL, Zhang W, et al. (2015) MECP2e1 isoform mutation affects the form and function of neurons derived from Rett syndrome patient iPS cells. Neurobiol Dis 76: 37–45. doi: 10.1016/j.nbd.2015.01.001
    [93] Lioy DT, Garg SK, Monaghan CE, et al. (2011) A role for glia in the progression of Rett's syndrome. Nature 475: 497–500. doi: 10.1038/nature10214
    [94] Williams EC, Zhong X, Mohamed A, et al. (2014) Mutant astrocytes differentiated from Rett syndrome patients-specific iPSCs have adverse effects on wildtype neurons. Hum Mol Genet 23: 2968–2980. doi: 10.1093/hmg/ddu008
    [95] Pearn JH (1978) Incidence, prevalence, and gene frequency studies of chronic childhood spinal muscular atrophy. J Med Genet 15: 409–413. doi: 10.1136/jmg.15.6.409
    [96] Ebert AD, Svendsen CN (2010) Stem cell model of spinal muscular atrophy. Arch Neurol 67: 665–669.
    [97] Sareen D, Ebert AD, Heins BM, et al. (2012) Inhibition of apoptosis blocks human motor neuron cell death in a stem cell model of spinal muscular atrophy. PLoS One 7: e39113. doi: 10.1371/journal.pone.0039113
    [98] Liu Q, Fischer U, Wang F, et al. (1997) The spinal muscular atrophy disease gene product, SMN, and its associated protein SIP1 are in a complex with spliceosomal snRNP proteins. Cell 90: 1013–1021. doi: 10.1016/S0092-8674(00)80367-0
    [99] Fischer U, Liu Q, Dreyfuss G (1997) The SMN-SIP1 complex has an essential role in spliceosomal snRNP biogenesis. Cell 90: 1023–1029. doi: 10.1016/S0092-8674(00)80368-2
    [100] Rossoll W, Jablonka S, Andreassi C, et al. (2003) Smn, the spinal muscular atrophy-determining gene product, modulates axon growth and localization of β-actin mRNA in growth cones of motoneurons. J Cell Biol 163: 801–812. doi: 10.1083/jcb.200304128
    [101] Farrar MA, Kiernan MC (2015) The Genetics of Spinal Muscular Atrophy: Progress and Challenges. Neurotherapeutics 12: 290–302. doi: 10.1007/s13311-014-0314-x
    [102] Corti S, Nizzardo M, Simone C, et al. (2012) Genetic correction of human induced pluripotent stem cells from patients with spinal muscular atrophy. Sci Transl Med 4: 165ra162.
    [103] Yoshida M, Kitaoka S, Egawa N, et al. (2015) Modeling the early phenotype at the neuromuscular junction of spinal muscular atrophy using patient-derived iPSCs. Stem Cell Rep 4: 561–568. doi: 10.1016/j.stemcr.2015.02.010
    [104] Mcgivern JV, Patitucci TN, Nord JA, et al. (2013) Spinal muscular atrophy astrocytes exhibit abnormal calcium regulation and reduced growth factor production. Glia 61: 1418–1428. doi: 10.1002/glia.22522
    [105] Zhou C, Feng Z, Ko CP (2016) Defects in Motoneuron-Astrocyte Interactions in Spinal Muscular Atrophy. J Neurosci 36: 2543–2553. doi: 10.1523/JNEUROSCI.3534-15.2016
    [106] Almad AA, Maragakis NJ (2012) Glia: An emerging target for neurological disease therapy. Stem Cell Res Ther 3: 37. doi: 10.1186/scrt128
    [107] Cloake NC, Yan J, Aminian A, et al. (2018) PLP1 mutations in patients with multiple sclerosis: Identification of a new mutation and potential pathogenicity of the mutations. J Clin Med 7: 100. doi: 10.3390/jcm7050100
    [108] Hobson GM, Kamholz J (1993) PLP1-Related Disorders. GeneReviews®.
    [109] Garbern JY (2007) Pelizaeus-Merzbacher disease: Genetic and cellular pathogenesis. Cell Mol Life Sci 64: 50–65. doi: 10.1007/s00018-006-6182-8
    [110] Numasawa-Kuroiwa Y, Okada Y, Shibata S, et al. (2014) Involvement of ER stress in dysmyelination of pelizaeus-merzbacher disease with PLP1 missense mutations shown by iPSC-derived oligodendrocytes. Stem Cell Rep 2: 648–661. doi: 10.1016/j.stemcr.2014.03.007
    [111] Takahashi T, Suzuki T (2012) Role of sulfatide in normal and pathological cells and tissues. J Lipid Res 53: 1437–1450. doi: 10.1194/jlr.R026682
    [112] Patil SA, Maegawa GHB (2013) Developing therapeutic approaches for metachromatic leukodystrophy. Drug Des Devel Ther 7: 729–745.
    [113] Ramakrishnan H, Hedayati KK, Lüllmann-Rauch R, et al. (2007) Increasing sulfatide synthesis in myelin-forming cells of arylsulfatase A-deficient mice causes demyelination and neurological symptoms reminiscent of human metachromatic leukodystrophy. J Neurosci 27: 9482–9490. doi: 10.1523/JNEUROSCI.2287-07.2007
    [114] Deconinck N, Messaaoui A, Ziereisen F, et al. (2008) Metachromatic leukodystrophy without arylsulfatase A deficiency: A new case of saposin-B deficiency. Eur J Paediatr Neurol 12: 46–50. doi: 10.1016/j.ejpn.2007.05.004
    [115] Frati G, Luciani M, Meneghini V, et al. (2018) Human iPSC-based models highlight defective glial and neuronal differentiation from neural progenitor cells in metachromatic leukodystrophy. Cell Death Dis 9: 698. doi: 10.1038/s41419-018-0737-0
    [116] Meneghini V, Frati G, Sala D, et al. (2017) Generation of human induced pluripotent stem cell-derived bona fide neural stem cells for ex vivo gene therapy of metachromatic leukodystrophy. Stem Cells Transl Med 6: 352–368. doi: 10.5966/sctm.2015-0414
    [117] Messing A, LaPash Daniels CM, Hagemann TL (2010) Strategies for Treatment in Alexander Disease. Neurotherapeutics 7: 507–515. doi: 10.1016/j.nurt.2010.05.013
    [118] Prust M, Wang J, Morizono H, et al. (2011) GFAP mutations, age at onset, and clinical subtypes in Alexander disease. Neurology 77: 1287–1294. doi: 10.1212/WNL.0b013e3182309f72
    [119] Jones JR, Kong L, Hanna MG, et al. (2018) Mutations in GFAP Disrupt the Distribution and Function of Organelles in Human Astrocytes. Cell Rep 25: 947–958.e4. doi: 10.1016/j.celrep.2018.09.083
    [120] Kondo T, Funayama M, Miyake M, et al. (2016) Modeling Alexander disease with patient iPSCs reveals cellular and molecular pathology of astrocytes. Acta Neuropathol Commun 4: 69. doi: 10.1186/s40478-016-0337-0
    [121] Li L, Tian E, Chen X, et al. (2018) GFAP Mutations in Astrocytes Impair Oligodendrocyte Progenitor Proliferation and Myelination in an hiPSC Model of Alexander Disease. Cell Stem Cell 23: 239–251.e6. doi: 10.1016/j.stem.2018.07.009
    [122] Anheim M, Lagier-Tourenne C, Stevanin G, et al. (2009) SPG11 spastic paraplegia: A new cause of juvenile parkinsonism. J Neurol 256: 104–108. doi: 10.1007/s00415-009-0083-3
    [123] Örlén H, Melberg A, Raininko R, et al. (2009) SPG11 mutations cause Kjellin syndrome, a hereditary spastic paraplegia with thin corpus callosum and central retinal degeneration. Am J Med Genet B Neuropsychiatric Genet 150: 984–992.
    [124] Renvoisé B, Chang J, Singh R, et al. (2014) Lysosomal abnormalities in hereditary spastic paraplegia types SPG15 and SPG11. Ann Clin Trans Neurol 1: 379–389. doi: 10.1002/acn3.64
    [125] Murmu RP, Martin E, Rastetter A, et al. (2011) Cellular distribution and subcellular localization of spatacsin and spastizin, two proteins involved in hereditary spastic paraplegia. Mol Cell Neurosci 47: 191–202. doi: 10.1016/j.mcn.2011.04.004
    [126] Hirst J, Borner GH, Edgar J, et al. (2013) Interaction between AP-5 and the hereditary spastic paraplegia proteins SPG11 and SPG15. Mole Biol Cell 24: 2558–2569. doi: 10.1091/mbc.e13-03-0170
    [127] Pérez-Brangulí F, Mishra HK, Prots I, et al. (2014) Dysfunction of spatacsin leads to axonal pathology in SPG11-linked hereditary spastic paraplegia. Hum Mol Genet 23: 4859–4874. doi: 10.1093/hmg/ddu200
    [128] Mishra HK, Prots I, Havlicek S, et al. (2016) GSK3ß-dependent dysregulation of neurodevelopment in SPG11-patient induced pluripotent stem cell model. Ann Neurol 79: 826–840. doi: 10.1002/ana.24633
    [129] Varga RE, Khundadze M, Damme M, et al. (2015) In vivo evidence for lysosome depletion and impaired autophagic clearance in hereditary spastic paraplegia type SPG11. PLoS Genet 11: e1005454. doi: 10.1371/journal.pgen.1005454
    [130] LaMarca EA, Powell SK, Akbarian S, et al. (2018) Modeling neuropsychiatric and neurodegenerative diseases with induced pluripotent stem cells. Front Pediatr 6: 82. doi: 10.3389/fped.2018.00082
    [131] Tian F, Hao J, Hu L, et al. (2018) Modeling neurodegenerative diseases by human pluripotent stem cell-induced brain organoid. Transl Surg 3: 42. doi: 10.4103/ts.ts_4_18
    [132] Wang H (2018) Modeling neurological diseases with human brain organoids. Front Synaptic Neurosci 10: 15. doi: 10.3389/fnsyn.2018.00015
    [133] Gabriel E, Wason A, Ramani A, et al. (2016) CPAP promotes timely cilium disassembly to maintain neural progenitor pool. EMBO J 35: 803–819. doi: 10.15252/embj.201593679
    [134] Li Y, Muffat J, Omer A, et al. (2016) Induction of expansion and folding in human cerebral organoids. Cell Stem Cell 20: 385–396.e3.
    [135] Mariani J, Coppola G, Zhang P, et al. (2015) FOXG1-Dependent Dysregulation of GABA/Glutamate Neuron Differentiation in Autism Spectrum Disorders. Cell 162: 375–390. doi: 10.1016/j.cell.2015.06.034
    [136] Kim YG, Cha J, Chandrasegaran S (1996) Hybrid restriction enzymes: zinc finger fusions to Fok I cleavage domain. Proc Natl Acad Sci U S A 93: 1156–1160. doi: 10.1073/pnas.93.3.1156
    [137] Porteus MH, Baltimore D (2003) Chimeric nucleases stimulate gene targeting in human cells. Science 300: 763. doi: 10.1126/science.1078395
    [138] Wang Y, Zhang WY, Hu S, et al. (2012) Genome Editing of Human Embryonic Stem Cells and Induced Pluripotent Stem Cells With Zinc Finger Nucleases for Cellular Imaging. Circ Res 111: 1494–1503. doi: 10.1161/CIRCRESAHA.112.274969
    [139] Ramalingam S, London V, Kandavelou K, et al. (2013) Generation and genetic engineering of human induced pluripotent stem cells using designed zinc finger nucleases. Stem Cells Dev 22: 595–610. doi: 10.1089/scd.2012.0245
    [140] Wood AJ, Lo TW, Zeitler B, et al. (2011) Targeted Genome Editing Across Species Using ZFNs and TALENs. Science 333: 307–307. doi: 10.1126/science.1207773
    [141] Sanjana NE, Cong L, Zhou Y, et al. (2012) A transcription activator-like effector toolbox for genome engineering. Nat Protoc 7: 171–192. doi: 10.1038/nprot.2011.431
    [142] Cong L, Ran FA, Cox D, et al. (2013) Multiplex genome engineering using CRISPR/Cas systems. Science 339: 819–823. doi: 10.1126/science.1231143
    [143] Mali P, Yang L, Esvelt KM, et al. (2013) RNA-guided human genome engineering via Cas9. Science 339: 823–826. doi: 10.1126/science.1232033
    [144] Suzuki T, Asami M, Perry ACF (2015) Asymmetric parental genome engineering by Cas9 during mouse meiotic exit. Sci Rep 4: 7621. doi: 10.1038/srep07621
    [145] Smith C, Gore A, Yan W, et al. (2014) Whole-genome sequencing analysis reveals high specificity of CRISPR/Cas9 and TALEN-based genome editing in human iPSCs. Cell Stem Cell 15: 12–13. doi: 10.1016/j.stem.2014.06.011
    [146] Smith C, Abalde-Atristain L, He C, et al. (2015) Efficient and allele-specific genome editing of disease loci in human iPSCs. Mol Ther 23: 570–577. doi: 10.1038/mt.2014.226
    [147] He X, Li YX, Feng B (2018) New Turns for High Efficiency Knock-In of Large DNA in Human Pluripotent Stem Cells. Stem Cells Int 2018: 1–7.
    [148] Fu Y, Reyon D, Keith Joung J (2014) Targeted genome editing in human cells using CRISPR/Cas nucleases and truncated guide RNAs. Methods Enzymol 546: 21–45.
    [149] Tsai SQ, Wyvekens N, Khayter C, et al. (2014) Dimeric CRISPR RNA-guided FokI nucleases for highly specific genome editing. Nat Biotechnol 32: 569–576. doi: 10.1038/nbt.2908
    [150] Raitano S, Ordovàs L, De Muynck L, et al. (2015) Restoration of progranulin expression rescues cortical neuron generation in an induced pluripotent stem cell model of frontotemporal dementia. Stem Cell Rep 4: 16–24. doi: 10.1016/j.stemcr.2014.12.001
    [151] Ebert AD, Yu J, Rose FF, et al. (2009) Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature 457: 277–280. doi: 10.1038/nature07677
    [152] Simone C, Ramirez A, Bucchia M, et al. (2016) Is spinal muscular atrophy a disease of the motor neurons only: Pathogenesis and therapeutic implications? Cell Mol Life Sci 73: 1003–1020. doi: 10.1007/s00018-015-2106-9
    [153] Ramirez A, Crisafulli SG, Rizzuti M, et al. (2018) Investigation of New Morpholino Oligomers to Increase Survival Motor Neuron Protein Levels in Spinal Muscular Atrophy. Int J Mol Sci 19: E167. doi: 10.3390/ijms19010167
    [154] Klose RJ, Sarraf SA, Schmiedeberg L, et al. (2005) DNA Binding Selectivity of MeCP2 Due to a Requirement for A/T Sequences Adjacent to Methyl-CpG. Mol Cell 19: 667–678. doi: 10.1016/j.molcel.2005.07.021
    [155] Mellios N, Woodson J, Garcia RI, et al. (2014) β2-Adrenergic receptor agonist ameliorates phenotypes and corrects microRNA-mediated IGF1 deficits in a mouse model of Rett syndrome. Proc Natl Acad Sci U S A 111: 9947–9952. doi: 10.1073/pnas.1309426111
    [156] Chang Q, Khare G, Dani V, et al. (2006) The disease progression of Mecp2 mutant mice is affected by the level of BDNF expression. Neuron 49: 341–348. doi: 10.1016/j.neuron.2005.12.027
    [157] Wang H, Chan SA, Ogier M, et al. (2006) Dysregulation of brain-derived neurotrophic factor expression and neurosecretory function in Mecp2 null mice. J Neurosci 26: 10911–10915. doi: 10.1523/JNEUROSCI.1810-06.2006
    [158] Li W, Pozzo-Miller L (2014) BDNF deregulation in Rett syndrome. Neuropharmacology 76: 737–746. doi: 10.1016/j.neuropharm.2013.03.024
    [159] Williams EC, Zhong X, Mohamed A, et al. (2014) Mutant astrocytes differentiated from Rett syndrome patients-specific iPSCs have adverse effects on wild-type neurons. Hum Mol Genet 23: 2968–2980. doi: 10.1093/hmg/ddu008
    [160] Zhu JM, Zhao YY, Chen SD, et al. (2011) Functional recovery after transplantation of neural stem cells modified by brain-derived neurotrophic factor in rats with cerebral ischaemia. J Int Med Res 39: 488–498. doi: 10.1177/147323001103900216
    [161] Behrstock S, Ebert AD, Klein S, et al. (2008) Lesion-induced increase in survival and migration of human neural progenitor cells releasing GDNF. Cell Transplant 17: 753–762. doi: 10.3727/096368908786516819
    [162] Hjelm BE, Grunseich C, Gowing G, et al. (2016) Mifepristone-inducible transgene expression in neural progenitor cells in vitro and in vivo. Gene Ther 23: 424–437. doi: 10.1038/gt.2016.13
  • Reader Comments
  • © 2019 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(6161) PDF downloads(1674) Cited by(1)

Article outline

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog