IMR Press / JIN / Volume 19 / Issue 1 / DOI: 10.31083/j.jin.2020.01.1142
Open Access Mini-Review
Neural stem cell secretome and its role in the treatment of neurodegenerative disorders
Show Less
1 Beijing Yinfeng Dingcheng Biological Engineering Technology Ltd., Beijing, 100176, P. R. China
2 Shandong Centers for Disease Control and Prevention, Jinan, Shandong, 250014, P. R. China
*Correspondence: han201419@163.com (Qilong Cao)
J. Integr. Neurosci. 2020, 19(1), 179–185; https://doi.org/10.31083/j.jin.2020.01.1142
Submitted: 6 August 2019 | Accepted: 13 November 2019 | Published: 30 March 2020
Copyright: © 2020 Zhang et al. Published by IMR press.
This is an open access article under the CC BY-NC 4.0 license (https://creativecommons.org/licenses/by-nc/4.0/).
Abstract

Neurological diseases in the central nervous system are mostly characterized by the failure of endogenous repair to restore tissue damage and salvage lost function. Currently, studies have shown that neural stem cell transplantation provides a good therapeutic effect on neurological diseases. For this reason, neural stem cell transplantation has been explored as a cell replacement therapy. Although transplanted cells can replace cells lost during or post central nervous system injury, many studies have shown that this mechanism is insufficient as most of these newly formed cells fail to integrate and eventually die. Although it was initially thought that neural stem cell could only replace lost cells, recent experiments have shown that transplanted neural stem cell can also play bystander roles such as neuroprotection and immune regulation, promote tissue repair by preventing tissue damage, interfere with pathogenic processes, or by rescuing endogenous nerve cells. However, compelling evidence has raised concerns about this bystander effect, which can be caused by several biologically active molecules (collectively known as the secretome) produced by neural stem cells. These results also raise the possibility of the neural stem cell secretome as a potential candidate for neural stem cell transplantation therapies based on the bystander effect. A better understanding of the molecules and mechanisms of this effect is of critical importance for neural stem cell-based therapies. This review aims to discuss the function and application of neural stem cell secretome in the treatment of neurodegenerative disorders.

Keywords
Neuroplasticity
bystander effect
secretome
neurodegenerative disorders
neural stem cell
1. Introduction

Neural stem cells (NSC) are heterogeneous cells with mitotic activity, self-renewal, and multipotency, which drive neurogenesis and gliogenesis throughout the life of most mammals, including humans. When a neural injury occurs, NSCs have been shown to retain the capacity to migrate to damaged areas, promote functional and structural tissue repair and generate both neurons and glia with complex gene expression patterns and differences in space and time (Adams and Morshead, 2018; Göritz and Frisén, 2012). Recent technical advances allow NSCs to be obtained from three sources (Tang et al., 2017), including direct extraction from primary tissues (Belenguer et al., 2016), differentiation from pluripotent stem cells (Banda and Grabel, 2016) and transdifferentiation from somatic cells (Shahbazi et al., 2017).

Neural stem cells (NSC) are heterogeneous cells with mitotic activity, self-renewal, and multipotency, which drive neurogenesis and gliogenesis throughout the life of most mammals, including humans. When a neural injury occurs, NSCs have been shown to retain the capacity to migrate to damaged areas, promote functional and structural tissue repair and generate both neurons and glia with complex gene expression patterns and differences in space and time (Adams and Morshead, 2018; Göritz and Frisén, 2012). Recent technical advances allow NSCs to be obtained from three sources (Tang et al., 2017), including direct extraction from primary tissues (Belenguer et al., 2016), differentiation from pluripotent stem cells (Banda and Grabel, 2016) and transdifferentiation from somatic cells (Shahbazi et al., 2017).

The various characteristics of NSC transplantation offers great promise for the treatment of a wide variety of diseases, such as neurological disease (including spinal cord injury (Suzuki et al., 2017; Zhu et al., 2018), traumatic brain injury (Duan et al., 2016; Koutsoudaki et al., 2016), epilepsy (Lippert et al., 2017; Shetty, 2014), cerebral palsy (Tan et al., 2014; Zheng et al., 2012) and cerebrovascular diseases, including stroke (Lee et al., 2010). Ischemic and hemorrhagic stroke (Mozaffarian et al., 2016), is a leading worldwide cause of disability and death (Yu et al., 2018). A recent study has shown that transplanted NSCs could improve neurological deficits and reduced the severity of ischemic stroke by restoring endogenous striated spinous neurons, as well as by inhibiting inflammation and glial scar formation (Bacigaluppi et al., 2009). The results of other experiments have shown that rats show good functional performance in neurological tests 2-8 weeks after NSCs transplantation, which further demonstrates that exogenous NSCs could be used for functional recovery after hemorrhagic stroke (Lee et al., 2010). On this evidence, NSC transplantation is undoubtedly a promising therapy for the treatment of cardiovascular and neurological diseases.

2. The therapeutic mechanism of neural stem cell transplantation

Current methods for NSC transplantation in animal experiments and clinical applications include intravenous or intrathecal injections (Wang et al., 2017), intracerebral injection (Sullivan and Armstrong, 2017), and stereotactic (Li et al., 2013) and intraventricular injection (Griffin et al., 2015). As Chu et al. (2004) have shown, intravenously transplanted human NSCs can migrate to and differentiate within lesion sites to improve neural function, NSC transplantation has also been shown by the rotary table, limb position and turning ability tests to improve sensorimotor performance. Another study has shown that stereotactic transplanted NSCs can differentiate into neuronal (50%) and astroglial (15%) cells, which may then provide the replacement of cells where they are lost following injury (Kelly et al., 2004).

Although the therapeutic mechanisms of cell replacement for transplanted NSCs have led to a large number of studies, until now, they have increasing suggested that direct replacement of missing cells may not be the main mechanism of NSC transplantation (Bernstock et al., 2017; Marsh and Blurton-Jones, 2017). Several studies have shown this mechanism to be largely inadequate because most of the newly formed cells fail to integrate and eventually die (Arvidsson et al., 2002; Thored et al., 2010). Much research has shown that the beneficial role of NSCs may be attributed to other biological characteristics. For example, transplanted neural progenitor cells (NPCs) can play a bystander effect of neural protection and immune regulation in addition to the role of cell replacement, thus promoting tissue repair by prevention of tissue damage, interfering with pathogenic processes, or saving endogenous nerve cells (De Feo et al., 2012). Additionally, different studies have confirmed other bystander effects induced by NSCs, such as ameliorating damage to neurophysical structures, immunoregulation (Kim et al., 2018), neuroprotection (Cui et al., 2015; Pang et al., 2017), neurotrophication (Bernstock et al., 2017; Marsh and Blurton-Jones, 2017) and angiogenesis (Boese et al., 2018), as well as by having an endogenous impact in disease models (Jablonska et al., 2016).

Although NSC transplantation is undoubtedly a promising therapy for neurological disease, their limited persistence/survival in the harsh microenvironment of ischemic regions rich in reactive oxygen species means that the clinical transformation of this method remains elusive (Bernstock et al., 2017). However, the NSC secretome may not only help circumvent this difficult problem but also provide a consistent “on-demand” solution. There is, therefore, a promising therapeutic prospect held out by the NSC secretome in both replacement and auxiliary cell transplantation.

3. NSC secretomes

In addition to the abilities of self-renewal and differentiation into all neural cell lineages, NSCs secrete through the autocrine/paracrine pathway, a large number of bioactive compounds that play important therapeutic roles. Studies have shown that in pathological conditions, NSCs can regulate the microenvironment through a paracrine mechanism. This plays an important role in maintaining the characteristics of stem cells and directional differentiation. NSCs can also perform intercellular responses through paracrine signaling (Bollini et al., 2013; Drago et al., 2013). It is bioactive compounds, such as chemokines, cytokines, early inflammatory cytokines, growth factors, nutritional factors, stem cell regulatory factors, extracellular vesicle and other similar molecules that are generally referred as the NSC secretome (Fig. 1), which is considered important in regulating several critical biological processes, such as cell survival, proliferation and differentiation, immune regulation, anti-apoptosis and stimulation of adjacent cells in tissues ( Cossetti et al., 2012; Skalnikova et al., 2011). Up until the present, studies have shown that neurotrophic factors derived from NSCs play important roles in cell cycle regulation (Einstein and Ben-Hur, 2008), cell survival (Marsh and Blurton-Jones, 2017), development and adult differentiation (Hyman et al., 1991; Numakawa et al., 2017).

Figure 1.

Bystander effect generated by NSC secretome. NSC transplantation for the treatment of neurodegenerative disorders, including spinal cord injury, traumatic brain injury, epilepsy, cerebral palsy, and stroke, among others. The main mechanism for NSC transplantation may not only be by direct replacement of missing cells but also through the bystander effect, which includes neuroprotection, neuroplasticity, immunoregulation by NSC secretomes (paracrine factor, microvesicles, and exosomes).

Drago et al. (2013) have shown that significant repair of damaged brains can be achieved by injecting biological agents secreted by stem cells, rather than by directly inserting stem cells for cell replacement.

Further, brain-derived neurotrophic factor (BDNF) has been shown to have some success in ameliorating symptoms in several studies of preclinical models of Huntington's disease (Gharami et al., 2007; Giralt et al., 2009). Trials are underway to treat Alzheimer's disease with nerve growth factor (NGF) (Allen et al., 2013) which is administered either as injected proteins (Tian et al., 2012), released locally by implanted NGF-secreting cells (Tuszynski et al., 2005) or by introducing an adeno-associated virus-based gene delivery vector that encodes for human NGF (Bishop et al., 2008). Additionally, stem cell-secreted factors have been used in several clinical studies (Drago et al., 2013), and the clinical trial of autologous mesenchymal stem cell secreting factor for the treatment of amyotrophic lateral sclerosis has been completed (ClinicalTrials.gov Identifier: NCT01051882), as has intraventricular infusion (Nutt et al., 2003) or putaminal infusion (Lang et al., 2006) of glial cell-derived nutrient factor (GDNF) which has recently been shown to be an effective treatment for Parkinson's disease and is currently undergoing clinical trials (Allen et al., 2013).

In addition to endogenous NSCs, transplanted NSCs can also secrete a series of factors such as chemokines, cytokines, growth factors, and stem cell regulators, which exert immune regulation and nutritional support after reaching a lesion site (Kokaia et al., 2012). Following a study of an amyotrophic lateral sclerosis disease model which found that transplanted NSCs secrete neurotrophic factors that promote the survival and regeneration of motor neurons, Kim et al. (2018) found that the secretome of transplanted NSCs also plays a role in immunoregulation and neuroprotection (Kim et al., 2018; Xu et al., 2006).

4. The function of NSC secretome neuroprotection
4.1 Protect neurons

Despite the initial perception that NSC transplantation only served to replace lost cells, recent experimental studies have shown that transplanted NSCs also exert additional bystander effects such as the neuroprotection that promote tissue repair (Aharonowiz et al., 2008). To verify this characteristic of NSCs, Liang et al. (2014) removed the neural stem cell from a normal culture medium to acquire neural stem cell-conditioned medium to exclude the effect of cell substitution. Results showed that continuous administration of conditioned medium in rats with spinal cord injury increases bridging between the corticospinal tract and intermediate neurons reduces neuron apoptosis, and promotes the recovery of motor function. Other findings show that the conditioned medium of NSCs may provide a better neuroprotective effect than the normal medium. This was attributed to neurotrophic factors such as NGF, GDNF, and a vascular endothelial growth factor (VEGF) secreted by NSCs (Lee et al., 2017). Additionally, another studies have found that in primary and secondary neurodegenerative disorders such as Huntington’s disease (Ryu et al., 2004), spinal cord injury (Ziv et al., 2006) and stroke (Chu et al., 2004), the main mechanism of NSCs in prevention of neuronal programmed cell death is also mediated by NGF and BDNF, ciliary neurotrophic factor (CNTF) and GDNF, which are secreted by transplanted NSCs/NPCs.

4.2 Protect oligodendrocytes

It has been shown that CNTF secreted by NPCs can participate in the survival and differentiation of oligodendrocyte progenitor cells (OPCs) (Marriott et al., 2010). Further, in a demyelinating disease model, transplanted NSCs induces the proliferation of OPCs and promotes myelin regeneration by secreting platelet-derived growth factor (PDGF)-AA and basic fibroblast growth factor (FGF-2) (Einstein et al., 2009). The protective effects of these two factors on OPCs have been widely demonstrated: a growing number of research groups have demonstrated that FGF-2 (Chen et al., 2015; Raff et al., 1983) and PDGF (Bertold et al., 1999; Watzlawik et al., 2013) promote the survival and proliferation of OPCs in vitro and (Barres et al., 1992; Bertold et al., 1999).

4.3 Other effects

The neuroprotective effects of transplanted undifferentiated human fetal NSCs in ischemic stroke are attributed not only to the secretion of nutrient factors (i.e., VEGF) but also to the re-expression of developmental molecules, i.e., Slit-a family of secreted extracellular matrix proteins (Andres et al., 2011).

In addition to factors, the secretome also contains many extracellular vesicles, including exosomes and microvesicles. It is well known that exosomes are carriers of intercellular communication, transporting and transferring microRNA between cells (Camussi et al., 2010; Lotvall and Valadi, 2007). Currently, researchers have identified miRNA in exosomes of NSCs using second-generation sequencing technology and found 113 miRNA types, many of which are relevant for neural regeneration, neuroprotection and neural plasticity (Stevanato et al., 2016). Until now, various miRNA has been found to have repair and treatment effects on central nervous system (CNS) injuries, such as stroke, traumatic brain injury, and spinal cord injury (Faravelli and Corti, 2017; Sun et al., 2018). Some studies have shown that cortical neurogenesis is increased by targeting exosomal miR-124 after stroke (Yang et al., 2017).

Further, another study has shown downregulated miR‐134 by electroacupuncture enhances expression of LIM domain kinase and increases synaptic‐dendritic plasticity following ischemic stroke in a rat model (Schratt et al., 2006). Since their discovery and development, new therapeutic approaches to stroke are being based on NSC-derived exosomal miRNAs (Zhang et al., 2018). Furthermore, other researchers have determined proteins of the human NSCs exosome by tandem mass spectrometry and identified 103 proteins, including HSP70 and the transferrin receptor (Kang et al., 2008). Among them, HSP70 is considered a protective agent by its inhibition of oxidative stress that affects a variety of neurological diseases, including craniocerebral injury, chemical-toxic nerve injury, and Alzheimer's disease (Cossetti et al., 2014).

5. The function of NSC secretome: Neuroplasticity
5.1 Nerve regeneration

NSCs transplantation has been shown to increase the expression of VEGF, platelet reactive protein 1/2, and cysteine-rich acidic proteins in the brain of stroke patients. These are key factors in promoting dendritic growth, axonal plasticity, and axonal transport (Andres et al., 2011). In an experimental model of spinocerebellar ataxia type 1, transplanted NSCs have been shown to induce electrochemical coupling with damaged neurons through the formation of gap junctions, thereby preventing local cell damage (Jäderstad et al., 2010).

5.2 Angiogenesis

Since stroke may damage the brain vascular system, angiogenesis may help restore neural function. Relevant studies have shown that CTX0E03 cells, a human NSC line, express angiogenic factors in vitro, and may promote angiogenesis through paracrine factors and direct physical interactions. Additionally, in vivo results show a significant increase in microvessels in the rodent brain where cells were implanted. Accordingly, NSC-based therapy may provide significant benefits to stroke patients by upregulation of angiogenesis in ischemic lesions (Caroline et al., 2013; Stroemer et al., 2009). Moreover, a further study demonstrated for the first time that NSC-secreted VEGF is required to enhance the angiogenesis signaling pathway in vivo (Horie et al., 2015). The authors suggest that NSCs affect not only endothelial cells but also other key components of the vascular system in a VEGF-dependent manner, such as by enhancing the expression of β-dystroglycan, an extracellular matrix adhesion protein rich in perivascular astrocyte endings and expressed in peripheral and endothelial cells (Milner et al., 2008; Zaccaria et al., 2001).

6. The function of NSC secretome: Immunoregulation

One study has confirmed that NSCs transplantation exhibits an immunoregulatory role in spinal cord injury. This is mediated by decreased production of M1-type macrophages after injury and increased production of M2-type macrophages, which phagocytize disintegration products and secrete anti-inflammatory factors, thus inhibit the inflammatory response (Cusimano et al., 2012).

Additionally, another study shows that NPC transplantation not only inhibits T-cell proliferation (Einstein et al., 2003) but also promotes apoptosis of infiltrating T cells (Pluchino et al., 2005). This may be achieved by two mechanisms: one is the induction of expression of death receptor ligands, such as FasL, Trail, and Apo3L; the other is by promoting the production of soluble molecules associated with mitochondrial-mediated apoptosis, such as GDNF, interferon-γ, and leukemia inhibitory factor (De Feo et al., 2012).

Interestingly, although VEGF is generally thought to be pro-inflammatory, increasing literature has confirmed the anti-inflammatory properties of VEGF secreted by NSCs. Some studies have found that VEGF not only inhibits the development of antigen-presenting dendritic cells and T cells but also suppresses their activation (Gabrilovich et al., 1996; Ohm et al., 2003). VEGF may also influence macrophage proliferation in a dose-dependent manner since Manoonkitiwongsa et al. (2006) found that low-dose VEGF treatment of ischemic brain injury reduces macrophage number, while high-dose VEGF increases macrophage density. Of note, other experiments confirmed that VEGF is key for the immunosuppressive effects of NSCs and may be involved in altering the integrity of the blood-brain barrier (Horie et al., 2015).

7. Conclusion

In conclusion, NSC secretomes contain various factors and extracellular vesicles that play a role in neuroprotection, neuroplasticity, and immunoregulation of the nervous system. Although the paracrine mechanism of NSCs is presently not well understood, current research shows that the NSC secretome has considerable therapeutic significance and prospects. Compared with the direct use of NSCs, the secretome may have many unique advantages, such as easy access by ultrafiltration centrifugation or other similar methods, longer storage time, simpler transportation mode, and body lesions can be simulated to stimulate the production of more specific paracrine products. Overall, more understanding about the role of NSC secretomes and their clinical benefits in neurological disease could contribute to the discovery and development of new therapeutic approaches.

Abbreviations

ALS: amyotrophic lateral sclerosis; BDNF: brain-derived neurotrophic factor; CNS: central nervous system; CNTF: ciliary neurotrophic factor; CSPG: chondroitin sulfate proteoglycan; GDNF: glial cell-derived nutrient factor; HSP70: heat shock protein 70; NGF: nerve growth factor; NPCs: neural progenitor cells; NSCs: neural stem cell; NT-3: neurotrophin 3; VEGF: vascular endothelial growth factor.

Acknowledgment

This work is supported by the National Natural Science Foundation of China (Grant NO. : 81572495).

Conflict of Interest

The authors declare no conflicts of interest.

References
[1]
Adams, K. V. and Morshead, C. M. (2018) Neural stem cell heterogeneity in the mammalian forebrain. Progress in Neurobiology 170, 2-36.
[2]
Aharonowiz, M., Einstein, O., Fainstein, N., Lassmann, H., Reubinoff, B. and Ben-Hur, T. (2008) Neuroprotective effect of transplanted human embryonic stem cell-derived neural precursors in an animal model of multiple sclerosis. Plos One 3, e3145.
[3]
Allen, S. J., Watson, J. J., Shoemark, D. K., Barua, N. U. and Patel, N. K. (2013) GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacology & Therapeutics 138, 155-175.
[4]
Andres, R. H., Horie, N., Slikker, W., Keren-Gill, H., Zhan, K., Sun, G., Manley, N. C., Pereira, M. P., Sheikh, L. A., McMillan, E. L., Schaar, B. T., Svendsen, C. N., Bliss, T. M. and Steinberg, G. K. (2011) Human neural stem cells enhance structural plasticity and axonal transport in the ischaemic brain. Brain 134, 1777-1789.
[5]
Arvidsson, A., Collin, T., Kirik, D., Kokaia, Z. and Lindvall, O. (2002) Neuronal replacement from endogenous precursors in the adult brain after stroke. Nature Medicine 8, 963-970.
[6]
Bacigaluppi, M., Pluchino, S., Peruzzotti-Jametti, L., Kilic, E., Kilic, U., Salani, G., Brambilla, E., West, M. J., Comi, G., Martino, G. and Hermann, D. M. (2009) Delayed post-ischaemic neuroprotection following systemic neural stem cell transplantation involves multiple mechanisms. Brain 132, 2239-2251.
[7]
Banda, E. and Grabel, L. (2016) Directed differentiation of human embryonic stem cells into neural progenitors. Methods in Molecular Biology 1307, 289-298.
[8]
Barres, B. A., Hart, I. K., Coles, H. S., Burne, J. F., Voyvodic, J. T., Richardson, W. D. and Raff, M. C. (1992) Cell death and control of cell survival in the oligodendrocyte lineage. Cell 70, 31-46.
[9]
Belenguer, G., Domingo-Muelas, A., Ferrón, S. R., Morante-Redolat, J. M. and Fariñas, I. (2016) Isolation, culture and analysis of adult subependymal neural stem cells. Differentiation 91, 28-41.
[10]
Bernstock, J. D., Peruzzotti-Jametti, L., Ye, D., Gessler, F. A., Maric, D., Vicario, N., Lee, Y. J., Pluchino, S. and Hallenbeck, J. M. (2017) Neural stem cell transplantation in ischemic stroke: A role for preconditioning and cellular engineering. Journal of Cerebral Blood Flow & Metabolism 37, 2314-2319.
[11]
Bertold, C. H., Boström, H., Richardson, W. S., Calver, A. R., Karlsson, S. L., Willetts, K., Abramsson, A., Hall, D. A., Heath, C. J. and Betsholtz, C. (1999) Defective oligodendrocyte development and severe hypomyelination in PDGF-A knockout mice. Development 126, 457-467.
[12]
Bishop, K. M., Hofer, E. K., Mehta, A., Ramirez, A., Sun, L., Tuszynski, M. and Bartus, R. T. (2008) Therapeutic potential of CERE-110 (AAV2-NGF): Targeted, stable, and sustained NGF delivery and trophic activity on rodent basal forebrain cholinergic neurons. Experimental Neurology 211, 574-584.
[13]
Boese, A. C., Le, Q. E., Pham, D., Hamblin, M. H. and Lee, L. P. (2018) Neural stem cell therapy for subacute and chronic ischemic stroke. Stem Cell Research & Therapy 9, 154.
[14]
Bollini, S., Chiara, G., Roberta, T. and Ranieri, C. (2013) The regenerative role of the fetal and adult stem cell secretome. Journal of Clinical Medicine 2, 302-327.
[15]
Camussi, G., Deregibus, M. C., Bruno, S., Cantaluppi, V. and Biancone, L. (2010) Exosomes/microvesicles as a mechanism of cell-to-cell communication. Kidney International 78, 838-848.
[16]
Caroline, H., Stevanato, L., Stroemer, R. P., Tang, E., Richardson, S. and Sinden, J. D. (2013) In vivo and in vitro characterization of the angiogenic effect of CTX0E03 human neural stem cells. Cell Transplantation 22, 1541-1552.
[17]
Chen, Y. J., Zhang, J. X., Shen, L., Qi, Q., Cheng, X. X., Zhong, Z. R., Jiang, Z. Q., Wang, R., , H. Z. and Hu, J. G. (2015) Schwann cells induce proliferation and migration of oligodendrocyte precursor cells through secretion of PDGF-AA and FGF-2. Journal of Molecular Neuroscience 56, 999-1008.
[18]
Chu, K., Kim, M., Park, K., Jeong, S., Park, H. K., Jung, K., Lee, S. T., Kang, L., Lee, K., Park, D. K., Kim, S. U. and Ro, J. (2004) Human neural stem cells improve sensorimotor deficits in the adult rat brain with experimental focal ischemia. Brain Research 1016, 145-153.
[19]
Cossetti, C., Alfaro-Cervello, C., Donegà, M., Tyzack, G. and Pluchino, S. (2012) New perspectives of tissue remodelling with neural stem and progenitor cell-based therapies. Cell & Tissue Research 349, 321-329.
[20]
Cossetti, C., Iraci, N., Mercer, T. R., Leonardi, T., Alpi, E., Drago, D., Alfaro-Cervello, C., Saini, H. K., Davis, M. P., Schaeffer, J., Vega, B., Stefanini, M., Zhao, C., Muller, W., Garcia-Verdugo, J. M., Mathivanan, S., Bachi, A., Enright, A. J., Mattick, J. S. and Pluchino, S. (2014) Extracellular vesicles from neural stem cells transfer IFN-γ via Ifngr1 to activate Stat1 signaling in target cells. Molecular Cell 56, 193-204.
[21]
Cui, G. H., Shao, S. J., Yang, J. J., Liu, J. R. and Guo, H. D. (2015) Designer self-assemble peptides maximize the therapeutic benefits of neural stem cell transplantation for alzheimer’s disease via enhancing neuron differentiation and paracrine action. Molecular Neurobiology 53, 1108-1123.
[22]
Cusimano, M., Biziato, D., Brambilla, E., Donegà, M., Alfaro-Cervello, C., Snider, S., Salani, G., Pucci, F., Comi, G., Garcia-Verdugo, J. M., De Palma, M., Martino, G. and Pluchino, S. (2012) Transplanted neural stem/precursor cells instruct phagocytes and reduce secondary tissue damage in the injured spinal cord. Brain 135, 447-460.
[23]
De Feo, D., Merlini, A., Laterza, C. and Martino, G. (2012) Neural stem cell transplantation in central nervous system disorders: from cell replacement to neuroprotection. Current Opinion in Neurology 25, 322-333.
[24]
Drago, D., Cossetti, C., Iraci, N., Gaude, E., Musco, G., Bachi, A. and Pluchino, S. (2013) The stem cell secretome and its role in brain repair. Biochimie 95, 2271-2285.
[25]
Duan, H., Li, X., Wang, C., Hao, P., Song, W., Li, M., Zhao, W., Gao, Y. and Yang, Z. (2016) Functional hyaluronate collagen scaffolds induce NSCs differentiation into functional neurons in repairing the traumatic brain injury. Acta Biomaterialia 45, 182-195.
[26]
Einstein, O. and Ben-Hur, T. (2008) The changing face of neural stem cell therapy in neurologic diseases. Archives of Neurology 65, 452-456.
[27]
Einstein, O., Friedman-Levi, Y., Grigoriadis, N. and Tamir, B. H. (2009) Transplanted neural precursors enhance host brain-derived myelin regeneration. Journal of Neuroscience 29, 15694-15702.
[28]
Einstein, O., Karussis, D., Grigoriadis, N., Mizrachi-Kol, R., Reinhartz, E., Abramsky, O. and Ben-Hur, T. (2003) Intraventricular transplantation of neural precursor cell spheres attenuates acute experimental allergic encephalomyelitis. Molecular & Cellular Neuroscience 24, 1074-1082.
[29]
Faravelli, I. and Corti, S. (2017) MicroRna-directed neuronal reprogramming as a therapeutic strategy for neurological diseases. Molecular Neurobiology 55, 4428-4436.
[30]
Gabrilovich, D. I., Chen, H. L., Girgis, K. R., Cunningham, H. T., Meny, G. M., Nadaf, S., Kavanaugh, D. and Carbone, D. P. (1996) Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nature Medicine 2, 1096-1103.
[31]
Gharami, K., Xie, Y., An, J. J., Tonegawa, S. and Xu, B. (2007) Brain-derived neurotrophic factor over-expression in the forebrain ameliorates Huntington's disease phenotypes in mice. Journal of Neurochemistry 105, 369-379.
[32]
Giralt, A., Rodrigo, T., Martín, E. D., Gonzalez, J. R., Milà, M., Ceña, V., Dierssen, M., Canals, J. M. and Alberch, J. (2009) Brain-derived neurotrophic factor modulates the severity of cognitive alterations induced by mutant huntingtin: involvement of phospholipaseCgamma activity and glutamate receptor expression. Neuroscience 158, 1234-1250.
[33]
Göritz, C. and Frisén, J. (2012) Neural stem cells and neurogenesis in the adult. Cell Stem Cell 10, 657-659.
[34]
Griffin, T. A., Anderson, H. C. and Wolfe, J. H. (2015) Ex vivo gene therapy using patient ipsc-derived nscs reverses pathology in the brain of a homologous mouse model. Stem Cell Reports 4, 835-846.
[35]
Horie, N., Pereira, M. P., Niizuma, K., Sun, G., Keren-Gill, H., Encarnacion, A., Shamloo, M., Hamilton, S. A., Jiang, K., Huhn, S., Palmer, T. D., Bliss, T. M. and Steinberg, G. K. (2015) Transplanted stem cell-secreted vascular endothelial growth factor effects poststroke recovery, inflammation, and vascular repair. Stem Cells 29, 274-285.
[36]
Hyman, C., Hofer, M., Barde, Y. A., Juhasz, M., Yancopoulos, G. D., Squinto, S. P. and Lindsay, R. M. (1991) BDNF is a neurotrophic factor for dopaminergic neurons of the substantia nigra. Nature 350, 230-232.
[37]
Jablonska, A., Drela, K., Wojcik-Stanaszek, L., Janowski, M., Zalewska, T. and Lukomska, B. (2016) Short-lived human umbilical cord-blood-derived neural stem cells influence the endogenous secretome and increase the number of endogenous neural progenitors in a rat model of lacunar stroke. Molecular Neurobiology 53, 6413-6425.
[38]
Jäderstad, J., Jäderstad, L. M., Li, J., Chintawar, S., Salto, C., Pandolfo, M., Ourednik, V., Teng, Y. D., Sidman, R. L., Arenas, E., Snyder, E. Y. and Herlenius, E. (2010) Communication via gap junctions underlies early functional and beneficial interactions between grafted neural stem cells and the host. Proceedings of the National Academy of Sciences of the United States of America 107, 5184-5189.
[39]
Kang, D., Oh, S., Ahn, S. M., Lee, B. H. and Moon, M. H. (2008) Proteomic analysis of exosomes from human neural stem cells by flow field-flow fractionation and nanoflow liquid chromatography-tandem mass spectrometry. Journal of Proteome Research 7, 3475-3480.
[40]
Kelly, S., Bliss, T. M., Shah, A. K., Sun, G. H., Ma, M., Foo, W. C., Masel, J., Yenari, M. A., Weissman, I. L., Uchida, N., Palmer, T. and Steinberg, G. K. (2004) Transplanted human fetal neural stem cells survive, migrate, and differentiate in ischemic rat cerebral cortex. Proceedings of the National Academy of Sciences of the United States of America 101, 11839-11844.
[41]
Kim, H. W., Lee, H. S., Kang, J. M., Bae, S. H., Kim, C., Lee, S. H., Schwarz, J., Kim, G. J., Kim, J. S., Cha, D. H., Kim, J., Chang, S. W., Lee, T. H. and Moon, J. (2018) Dual effects of human placenta-derived neural cells on neuroprotection and the inhibition of neuroinflammation in a rodent model of parkinson's disease. Cell Transplantation 27, 814-830.
[42]
Kokaia, Z., Martino, G., Schwartz, M. and Lindvall, O. (2012) Cross-talk between neural stem cells and immune cells: the key to better brain repair? Nature Neuroscience 15, 1078-1087.
[43]
Koutsoudaki, P. N., Papastefanaki, F., Stamatakis, A., Kouroupi, G., Xingi, E., Stylianopoulou, F. and Matsas, R. (2016) Neural stem/progenitor cells differentiate into oligodendrocytes, reduce inflammation, and ameliorate learning deficits after transplantation in a mouse model of traumatic brain injury. Glia 64, 763-779.
[44]
Lang, A. E., Gill, S., Patel, N. K., Lozano, A., Nutt, J. G., Penn, R., Brooks, D. J., Hotton, G., Moro, E., Heywood, P., Brodsky, M. A., Burchiel, K., Kelly, P., Dalvi, A., Scott, B., Stacy, M., Turner, D., Wooten, V. G., Elias, W. J., Laws, E. R., Dhawan, V., Stoessl, A. J., Matcham, J., Coffey, R. J. and Traub, M. (2006) Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Annals of Neurology 59, 459-466.
[45]
Lee, H. J., Kim, K. S., Kim, E. J., Choi, H. B., Lee, K. H., Park, I. H., Ko, Y., Jeong, S. W. and Kim, S. U. (2010) Brain transplantation of immortalized human neural stem cells promotes functional recovery in mouse intracerebral hemorrhage stroke model. Stem Cells 25, 1204-1212.
[46]
Lee, N. M., Chae, S. A. and Lee, H. J. (2017) Effects of neural stem cell media on hypoxic injury in rat hippocampal slice cultures. Brain Research 1677, 20-25.
[47]
Li, M., Liu, X., Yue, H., Xiong, W., Gu, J. and Xu, M. (2013) Transplantation of N-acetyl aspartyl-glutamate synthetase-activated neural stem cells after experimental traumatic brain injury significantly improves neurological recovery. Cellular Physiology & Biochemistry 32, 1776-1789.
[48]
Liang, P., Liu, J., Xiong, J., Liu, Q., Zhao, J., Liang, H., Zhao, L. and Tang, H. (2014) Neural stem cell conditioned medium protects neurons and promotes propriospinal neurons relay neural circuit reconnection after spinal cord injury. Cell Transplantation 23, S45-S56.
[49]
Lippert, T., Gelineau, L., Napoli, E. and Borlongan, C. V. (2017) Harnessing neural stem cells for treating psychiatric symptoms associated to fetal alcohol spectrum disorder and epilepsy. Progress in Neuro-psychopharmacology & Biological Psychiatry 80, 10-22.
[50]
Lotvall, J. and Valadi, H. (2007) Cell to cell signalling via exosomes through esRNA. Cell Adhesion & Migration 1, 156-158.
[51]
Manoonkitiwongsa, P. S., Schultz, R. L., Whitter, E. F. and Lyden, P. D. (2006) Contraindications of VEGF-based therapeutic angiogenesis: Effects on macrophage density and histology of normal and ischemic brains. Vascular Pharmacology 44, 316-325.
[52]
Marriott, M. P., Emery, B., Cate, H. S., Binder, M. D., Kemper, D., Wu, Q., Kolbe, S., Gordon, I. R., Wang, H., Egan, G., Murray, S., Butzkueven, H. and Kilpatrick, T. J. (2010) Leukemia inhibitory factor signaling modulates both central nervous system demyelination and myelin repair. Glia 56, 686-698.
[53]
Marsh, S. E. and Blurton-Jones, M. (2017) Neural stem cell therapy for neurodegenerative disorders: The role of neurotrophic support. Neurochemistry International 106, 94-100.
[54]
Mozaffarian, D., Benjamin, E. J., Go, A. S., Arnett, D. K., Blaha, M. J., Cushman, M., Das, S. R., de Ferranti, S., Després, J. P., Fullerton, H. J., Howard, V. J., Huffman, M. D., Isasi, C. R., Jiménez, M. C., Judd, S. E., Kissela, B. M., Lichtman, J. H., Lisabeth, L. D., Liu, S., Mackey, R. H., Magid, D. J., McGuire, D. K., MohlerIII, E. R., Moy, C. S., Muntner, P., Mussolino, M. E., Nasir, K., Neumar, R. W., Nichol, G., Palaniappan, L., Pandey, D. K., Reeves, M. J., Rodriguez, C. J., Rosamond, W., Sorlie, P. D., Stein, J., Towfighi, A., Turan, T. N., Virani, S. S., Woo, D., Yeh, R. W., Turner, M. B. and on behalf of the American Heart Association Statistics Committee and Stroke Statistics Subcommittee. (2016) Heart disease and stroke statistics-2016 update: A report from the american heart association. Circulation 133, e38-e360.
[55]
Milner, R., Hung, S., Wang, X., Spatz, M. and del Zoppo, G. J. (2008) The rapid decrease in astrocyte-associated dystroglycan expression by focal cerebral ischemia is protease-dependent. Journal of Cerebral Blood Flow & Metabolism 28, 812-823.
[56]
Numakawa, T., Odaka, H. and Adachi, N. (2017) Actions of brain-derived neurotrophic factor and glucocorticoid stress in neurogenesis. International Journal of Molecular Sciences 18, 2312.
[57]
Nutt, J. G., Burchiel, K. J., Comella, C. L., Jankovic, J., Lang, A. E., Laws, E. R., Lozano, A. M., Penn, R. D., Simpson, R. K., Stacy, M., Wooten, G. F. and ICV GDNF Study Group. (2003) Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology 60, 69-73.
[58]
Ohm, J. E., Gabrilovich, D. I., Sempowski, G. D., Kisseleva, E., Parman, K. S., Nadaf, S. and Carbone, D. P. (2003) VEGF inhibits T-cell development and may contribute to tumor-induced immune suppression. Blood 101, 4878-4886.
[59]
Pang, A. L., Xiong, L. L., Xia, Q. J., Liu, F., Wang, Y. C., Liu, F., Zhang, P., Meng, B. L., Tan, S. and Wang, T. H. (2017) Neural stem cell transplantation is associated with inhibition of apoptosis, bcl-xl upregulation, and recovery of neurological function in a rat model of traumatic brain injury. Cell Transplantation 26, 1262-1275.
[60]
Pluchino, S., Zanotti, L., Rossi, B., Brambilla, E., Ottoboni, L., Salani, G., Martinello, M., Cattalini, A., Bergami, A., Furlan, R., Comi, G., Constantin, G. and Martino, G. (2005) Neurosphere-derived multipotent precursors promote neuroprotection by an immunomodulatory mechanism. Nature 436, 266-271.
[61]
Raff, M. C., Miller, R. H. and Noble, M. (1983) A glial progenitor cell that develops in vitro into an astrocyte or an oligodendrocyte depending on culture medium. Nature 303, 390-396.
[62]
Ryu, J. K., Kim, J., Cho, S. J., Hatori, K., Nagai, A., Choi, H. B., Lee, M. C., McLarnon, J. G. and Kim, S. U. (2004) Proactive transplantation of human neural stem cells prevents degeneration of striatal neurons in a rat model of Huntington disease. Neurobiology of Disease 16, 68-77.
[63]
Schratt, G. M., Tuebing, F., Nigh, E. A., Kane, C. G., Sabatini, M. E., Kiebler, M. and Greenberg, M. E. (2006) A brain-specific microRNA regulates dendritic spine development. Nature 439, 283-289.
[64]
Shahbazi, E., Mirakhori, F., Ezzatizadeh, V. and Baharvand, H. (2017) Reprogramming of somatic cells to induced neural stem cells. Methods 133, 21-28.
[65]
Shetty, A. K. (2014) Hippocampal injury-induced cognitive and mood dysfunction, altered neurogenesis, and epilepsy: Can early neural stem cell grafting intervention provide protection? Epilepsy & Behavior 38, 117-124.
[66]
Skalnikova, H., Motlik, J., Gadher, S. J. and Kovarova, H. (2011) Mapping of the secretome of primary isolates of mammalian cells, stem cells and derived cell lines. Proteomics 11, 691-708.
[67]
Stevanato, L., Thanabalasundaram, L., Vysokov, N. and Sinden, J. D. (2016) Investigation of content, stoichiometry and transfer of miRNA from human neural stem cell line derived exosomes. Plos One 11, e0146353.
[68]
Stroemer, P., Patel, S., Hope, A., Oliveira, C., Pollock, K. and Sinden, J. (2009) The neural stem cell line CTX0E03 promotes behavioral recovery and endogenous neurogenesis after experimental stroke in a dose-dependent fashion. Neurorehabilitation & Neural Repair 23, 895-909.
[69]
Sullivan, G. M. and Armstrong, R. C. (2017) Transplanted adult neural stem cells express sonic hedgehog in vivo and suppress white matter neuroinflammation after experimental traumatic brain injury. Stem Cells International 2017, 9342534.
[70]
Sun, P., Liu, D. Z., Jickling, G. C., Sharp, F. R. and Yin, K. J. (2018) MicroRNA-based therapeutics in central nervous system injuries. Journal of Cerebral Blood Flow & Metabolism 38, 1125-1148.
[71]
Suzuki, H., Ahuja, C. S., Salewski, R. P., Li, L., Satkunendrarajah, K., Nagoshi, N., Shibata, S. and Fehlings, M. G. (2017) Neural stem cell mediated recovery is enhanced by Chondroitinase ABC pretreatment in chronic cervical spinal cord injury. Plos One 12, e0182339.
[72]
Tan, J., Zheng, X., Zhang, S., Yang, Y., Wang, X., Yu, X. and Zhong, L. (2014) Response of the sensorimotor cortex of cerebral palsy rats receiving transplantation of vascular endothelial growth factor 165-transfected neural stem cells. Neural Regeneration Research 9, 1763-1769.
[73]
Tang, Y., Yu, P. and Cheng, L. (2017) Current progress in the derivation and therapeutic application of neural stem cells. Cell Death & Disease 8, e3108.
[74]
Thored, P., Arvidsson, A., Cacci, E., Ahlenius, H., Kallur, T., Darsalia, V., Ekdahl, C. T., Kokaia, Z. and Lindvall, O. (2010) Persistent production of neurons from adult brain stem cells during recovery after stroke. Stem Cells 24, 739-747.
[75]
Tian, L., Guo, R., Yue, X., Lv, Q., Ye, X., Wang, Z., Chen, Z., Wu, B., Xu, G. and Liu, X. (2012) Intranasal administration of nerve growth factor ameliorate β-amyloid deposition after traumatic brain injury in rats. Brain Research 1440, 47-55.
[76]
Tuszynski, M. H., Thal, L., Pay, M., Salmon, D. P., Hs, U., Bakay, R., Patel, P., Blesch, A., Vahlsing, H. L., Ho, G., Tong, G., Potkin, S. G., Fallon, J., Hansen, L., Mufson, E. J., Kordower, J. H., Gall, C. and Conner, J. (2005) A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nature Medicine 11, 551-555.
[77]
Wang, Z., Luo, Y., Chen, L. and Liang, W. (2017) Safety of neural stem cell transplantation in patients with severe traumatic brain injury. Experimental & Therapeutic Medicine 13, 3613-3618.
[78]
Watzlawik, J. O., Warrington, A. E. and Rodriguez, M. (2013) PDGF is required for remyelination-promoting IgM stimulation of oligodendrocyte progenitor cell proliferation. Plos One 8, e55149.
[79]
Xu, L., Yan, J., Chen, D., Welsh, A. M., Hazel, T., Johe, K., Hatfield, G. and Koliatsos, V. E. (2006) Human neural stem cell grafts ameliorate motor neuron disease in SOD-1 transgenic rats. Transplantation 82, 865-875.
[80]
Yang, J., Zhang, X., Chen, X., Wang, L. and Yang, G. (2017) Exosome mediated delivery of miR-124 promotes neurogenesis after ischemia. Molecular Therapy Nucleic Acids 7, 278-287.
[81]
Yu, X., Wang, X., Zeng, S. and Tuo, X. (2018) Protective effects of primary neural stem cell treatment in ischemic stroke models. Experimental and Therapeutic Medicine 16, 2219-2228.
[82]
Zaccaria, M. L., Di Tommaso, F., Brancaccio, A., Paggi, P. and Petrucci, T. C. (2001) Dystroglycan distribution in adult mouse brain: a light and electron microscopy study. Neuroscience 104, 311-324.
[83]
Zhang, G., Chen, L., Guo, X., Wang, H., Chen, W., Wu, G., Gu, B., Miao, W., Kong, J., Jin, X., Yi, G., You, Y., Su, X. and Gu, N. (2018) Comparative analysis of microRNA expression profiles of exosomes derived from normal and hypoxic preconditioning human neural stem cells by next generation sequencing. Journal of Biomedical Nanotechnology 14, 1075-1089.
[84]
Zheng, X. R., Zhang, S. S., Yin, F., Tang, J. L., Yang, Y. J., Wang, X. and Zhong, L. (2012) Neuroprotection of VEGF-expression neural stem cells in neonatal cerebral palsy rats. Behavioural Brain Research 230, 108-115.
[85]
Zhu, Y., Uezono, N., Yasui, T. and Nakashima, K. (2018) Neural stem cell therapy aiming at better functional recovery after spinal cord injury. Developmental Dynamics 247, 75-84.
[86]
Ziv, Y., Avidan, H., Pluchino, S., Martino, G. and Schwartz, M. (2006) Synergy between immune cells and adult neural stem/progenitor cells promotes functional recovery from spinal cord injury. Proceedings of the National Academy of Sciences of the United States of America 103, 13174-13179.
Share
Back to top