Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Apr 1.
Published in final edited form as: Arch Immunol Ther Exp (Warsz). 2015 Nov 26;64(2):89–99. doi: 10.1007/s00005-015-0376-4

Neurotrophic Factors and Their Potential Applications in Tissue Regeneration

Nan Xiao 1,, Quynh-Thu Le 2
PMCID: PMC4805470  NIHMSID: NIHMS764715  PMID: 26611762

Abstract

Neurotrophic factors are growth factors that can nourish neurons and promote neuron survival and regeneration. They have been studied as potential drug candidates for treating neurodegenerative diseases. Since their identification, there are more and more evidences to indicate that neurotrophic factors are also expressed in non-neuronal tissues and regulate the survival, anti-inflammation, proliferation and differentiation in these tissues. This mini review summarizes the characteristics of the neurotrophic factors and their potential clinical applications in the regeneration of neuronal and non-neuronal tissues.

Keywords: Neurotrophic factor, Tissue regeneration

Introduction

Neurotrophic factors are growth factors that can promote the survival and regeneration of the neurons. They are sometimes referred as neurotrophins (NTs). The nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), other NTs all belong to the group. The glial cell derived neurotrophic factors (GDNF) family and neuropoietic cytokines, such as ciliary neurotrophic factor (CNTF) and leukemia inhibitory factor, are also considered members of neurotrophic factor family (Kerschensteiner et al. 2003; Saarma 2000; Stolp 2013).

The neurotrophic factors have long been extensively investigated for their roles in supporting the survival, proliferation and maturation of certain neurons. They have been shown to improve neural regeneration in neurodegerative diseases, such as Alzheimer's (Heese et al. 2006–2007), Parkinson's (de Munter et al. 2014) and Huntington's disease (Rosser and Svendsen 2014). Recent researches have indicated that neurotrophic factors can be found in the tissue-specific adult stem cell niche and promote tissue regeneration outside of the nervous system. These works suggest that neurotrophic factors can serve as potential therapeutic candidates in adult tissue regeneration.

Nerve Growth Factor

The first neurotrophic factor identified is the NGF. It was originally found to enhance the growth of sensory and sympathetic neurons in the chicken embryo (Levi-Montalcini and Hamburger 1951). NGF is enriched in the brain, with the highest level in the hippocampus (Shelton and Reichardt 1986). NGF elevation is related to the nervous system development, and it is found to reduce the degeneration of the cholinergic neurons (Hefti and Will 1987; Korsching et al. 1986).

NGF knockout mice are born alive, but are smaller than the wild type littermates. Their lifespan is less than 4 weeks postnatal. There is a marked reduction in the number of lumbar dorsal root ganglia as well as cholinergic neurons in the knockout mice, which exhibit severe impairment in spatial learning and motor coordination (Ruberti et al. 2000). The NGF receptor p75 and tyrosine kinase A receptor (TrkA) are critical in mediating the NGF effect. Reduction of either receptors leads to severe loss of sympathetic neurons and cholinergic neurons in mice, which is similar to reducing NGF expression (Lee et al. 1992; Smeyne et al. 1994).

Transplanting immortalized NGF secreting neural progenitors into the rat brain significantly enhance the spatial memory, as verified by the Morris water maze test 7 weeks after the transplantation (Martinez-Serrano et al. 1996). Recombinant NGF can reduce the death of sympathetic ganglionic neurons and cholinergic neurons in mice, as well as in humans. Recombinant human NGF has been introduced into the brain of patients with Alzheimer's disease via either viral mediated infection or implantable devices on clinical trials. There has not been any sign of significant toxicity and patients showed improved cognition, reduced death of cholinergic neurons, and less brain shrinkage (Aloe et al. 2012; Eriksdotter-Jonhagen et al. 2012; Ferreira et al. 2015; Mandel 2010; Petty et al. 1994; Sofroniew et al. 2001; Tuszynski et al. 2005). NGF delivery or the p75 receptor overexpression has been shown to improve survival and neurite growth of basal ganglia cells, and reduce the bradykinesia in patients (Olson et al. 1991) or animal (Pezzoli et al. 1988) with Parkinson's disease. It can also stimulate cerebral perfusion and the neurogenesis in hypoxic–ischemic brain injury in infants (Chiaretti et al. 2008). The molecular mechanism downstream of NGF may be related to inhibition of apoptosis (Nguyen et al. 2009) by down regulating the Bcl-2 pathway (Lu et al. 2013), and promoting the survival (Ji et al. 2014), proliferation (Moscatelli et al. 2009) and differentiation of the neural stem cells by upregulating the AKT and MAPK pathway (Yuan et al. 2013).

Besides the nervous system, NGF has been noted to be highly expressed in the hematopoietic stem cells (Durand et al. 2007). NGF increases the colony formation unit of the granulocytes and monocytes in a dose-dependent manner both in cell culture (Matsuda et al. 1988) and in injured mice model (Huang and Zhu 2008; Huang et al. 2008). NGF over expression in the bone marrow stem cells (BMSCs) has a stronger rescue effect on rat models with vascular dementia by increasing BMSCs proliferation (Wang et al. 2014a).

NGF and its receptor TrkA are also highly expressed in the rat neonatal cardiac myocytes (Caporali et al. 2008) as well as in the adult human myocardium (Meloni et al. 2010). Blocking NGF signal pathway increases the apoptosis of the cardiomyocyte, while NGF gene transfer facilitates cardiomyocyte survival and regeneration in mice by inhibiting apoptosis (Caporali et al. 2008; Lavasani et al. 2006; Mahmoud et al. 2015; Meloni et al. 2010).

NGF and its receptor TrkA, co-receptor p75 are found highly expressed in the rat pancreas during both embryonic and adult stage (Miralles et al. 1998; Oberg-Welsh and Welsh 1996). Lesion of pancreas significantly increases the level of NGF in both islet and the exocrine cells (Larrieta et al. 2006; Teitelman et al. 1998). This might be related to the endogenous mechanism to repair the damage. NGF treatment significantly improves the viability of the isolated mice pancreatic islets (Hata et al. 2015; Miao et al. 2006). Interestingly, NGF treatment also increases insulin secretion in cultured islet (Rosenbaum et al. 2001), as well as in transplanted islet (Miao et al. 2005). Injection of 6 μg NGF can restore the glucose tolerance in male Balb/c mice to the level that is comparable to that of islet transplanted mice (Miao et al. 2005), or the non-diabetic control mice (Hussey et al. 2010).

In human salivary glands duct, NGF and its receptors are also found expressed at high levels. The signals overlap with stem cell marker CD49f and Thy-1 (Sato et al. 2007), indicating that NGF may potentially involved regulating in salivary gland stem cell activities.

The evidences show that NGF pathway is important for the development of sympathetic and cholinergic neurons. It also improves survival and recovery of neurons post damage, most likely through inhibiting apoptosis. High expression of NGF can be found in stem cells in several non-neuronal tissues, where it also promotes the tissue regeneration. Besides inhibiting apoptosis, NGF also enhances proliferation and functional recovery of the non-neuronal tissues, the mechanism of which still needs further investigation.

Brain Derived Neurotrophic Factor

Brain derived neurotrophic factor (BDNF) was first purified from the pig brain as a growth factor that can increase the survival of cultured embryonic chicken spinal sensory neurons (Barde et al. 1982). Later, BDNF was found to be a pro-survival factor for many different neurons, including sensory neurons in the dorsal root ganglia (Acheson et al. 1995), hippocampal neurons, and cortical neurons (Huang and Reichardt 2001). BDNF has primarily an excitatory effect at the synapse by enhancing the excitatory postsynaptic potential (Kang and Schuman 1995; Kang et al. 1997; Patterson et al. 1996) and reducing the inhibitory postsynaptic potential (Marty et al. 1996). Because of its high expression in the hippocampus (Hall et al. 2000) and its role in long-term potential, BDNF is thought to play an important role in learning and memory. The signal is mainly through the TrkB pathway (Howells et al. 2000; Kang and Schuman 1995; Kang et al. 1997; Minichiello 2009; Patterson et al. 1996; Yamada and Nabeshima 2003). By activating the receptor TrkB (Drake et al. 1999), BDNF can inhibit the phosphorylation of the GABA receptor (Jovanovic et al. 2004), thereby reducing the conductance of these receptors (Rivera et al. 2002). The MAPK pathway has also been shown to be downstream of the TrkB activation, and mediates neuronal survival and differentiation (Criscuolo et al. 2015; Liu et al. 2013). BDNF also activates PI3 K pathway and upregulate the phosphorylation of the transcription factor CREB (cAMP response element-binding protein), which inhibits apoptosis and prevents neurodegeneration (Chen et al. 2015; Jain et al. 2013).

Most of BDNF knockout mice die within a couple of days. Some survive for a few weeks after birth. There is a reduction of sensory neurons, but not motor neurons (Conover et al. 1995; Erickson et al. 1996; Ernfors et al. 1994a; Jones et al. 1994). The long-term potential is impaired in BDNF knockout mice (Frerking et al. 1998; Korte et al. 1995; Wardle and Poo 2003), which can be rescued by adding recombinant BDNF (Korte et al. 1996; Patterson et al. 1996). Heterozygous BDNF mice are generally smaller than normal mice, and characterized by defects in movement, poor coordination, and obesity (Kernie et al. 2000; Lyons et al. 1999). Their spacial learning capability is reduced (Linnarsson et al. 1997). Reduction of BDNF expression has been associated with several neurodegenerative diseases including Parkinson's (Howells et al. 2000), Alzheimer's (Ferrer et al. 1999; Phillips et al. 1991) and more recently Huntington's disease (Zuccato et al. 2001, 2003). A drastic drop of BDNF expression in the brain has become a feature of the Hunt-ington's disease (Zuccato et al. 2008). The underlying mechanism may be related to lower transcription of the protein in the Huntington's patients (Zuccato et al. 2001). These evidences indicate that BDNF is critical in neuron development and prevents the sensory neuron from degeneration.

Co-injection of BDNF and CNTF subcutaneously improves regeneration of motor neurons (Mitsumoto et al. 1994). Virus mediated delivery of BDNF into the hippocampal neurons in normal rats (Jeon et al. 2015) or the striatum of quinolinic acid-induced Huntington rats (Kells et al. 2004) significantly improves neuronal regeneration. One time injection of 50 μg BDNF could successfully improve the morphology and rescue the electrophysiological properties of injured optic nerve in rabbits. BDNF was also shown to enhance regeneration after injury of the cervical spinal cord (Gransee et al. 2015), cavernous sinus nerve of the penis (Kim et al. 2012), sciatic nerve (Dadon-Nachum et al. 2012), optic nerve (Zhang et al. 2015), and olfactory epithelium (Frontera et al. 2015; Uranagase et al. 2012) in animal models.

Because the blood–brain barrier filters many substances from the plasma, including the neurotrophic factors, substances that regulate BDNF level are being tested in patients with neurodegenerative diseases. Citalopram (Celexa), which is a selective serotonin reuptake inhibitor used as an antidepressant, can efficiently increase BDNF level in the plasma (Goekint et al. 2011; Haghighi et al. 2013; Ladea and Bran 2013) and has been shown to enhance neuronal regeneration in a murine ischemic stroke model (Espinera et al. 2013). Ampakines, recently used as a compound to enhance learning and memory, also increases BDNF level (Lauterborn et al. 2009). Another substance that can increase BDNF level and potentially be used to treat Huntington's disease is Cystamine, which is a transglutaminase inhibitor (Borrell-Pages et al. 2006).

Besides the neuronal tissues, BDNF can be secreted by mesenchymal stem cells, such as BMSCs. There are many reports on the ability of BDNF to induce differentiation of BMSCs to neuron like cells, as verified by electrophysiological properties and neuronal specific markers (Han et al. 2015; Long et al. 2005; Sanchez-Ramos et al. 2000; Zhao et al. 2004). Transplantation of adipose tissue derived marrow stem cells significantly increases BDNF level in the brain, and is capable of reducing neuron damage in animal models (Berg et al. 2015; Han et al. 2014; Schwerk et al. 2015). Moreover, transplantation of marrow stem cells that overexpress BDNF or GDNF significantly improves the limb placement behavior in focal cerebral ischemia rats (Kurozumi et al. 2005). BDNF also enhances the proliferation and vascularization of the hematopoietic stem cells (Shmelkov et al. 2005).

There are not many reports of BDNF's function outside of the nervous system and the mesenchymal stem cells. However, when the BDNF signaling pathway is over activated, it promotes the growth of malignant gliomas (Lawn et al. 2015), breast cancer (Yin et al. 2015) and lung cancer (Sinkevicius et al. 2014), indicating BDNF can enhance the cell proliferation in certain types of cancer cells. Whether the effect is through cancer stem cell would require more evidences.

Other Neurotrophins

Neurotrophin (NT)-3 was the third neurotrophic factors identified based on the sequence identities to NGF and BDNF (Hohn et al. 1990; Maisonpierre et al. 1990). Another neurotrophic factor being investigated is NT-4 (Ip et al. 1992), also known as NT-5 in mice (Berkemeier et al. 1991). Similar to NGF and BDNF, NTs are required for the survival of sensory neurons isolated from the rat dorsal root ganglia, and promote the proliferation of the sensory neurons (Memberg and Hall 1995). NTs also promote chicken motor neuron survival (Becker et al. 1998). Similar to NGF and BDNF, NTs bind to the neurotrophin receptor p75 at low affinity. The binding between NGF, BDNF, NTs and the receptor tyrosine kinase are stronger and more specific. As mentioned above, NGF specifically binds to TrkA while BDNF preferentially binds to TrkB. NT-3 preferentially binds to TrkC, but can also activate the TrkA and TrkB, while NT-4/5 preferentially binds to TrkB (Berkemeier et al. 1991; Klein et al. 1992; Reichardt 2006). The PI3K/ AKT, MEK/ERK are reported to be downstream of the neurotrophin activation (Skaper 2012). During cerebral cortex development, NT-3 increases BrdU incorporation and the differentiation of phenotype specific neurons in the laminar formation through the MAPK pathway (Ohtsuka et al. 2013).

Similar to other neurotrophic factors, NT-3 is important in embryonic neuron development. NT-3 knockout mice die within a couple of weeks after birth. Peripheral sensory and sympathetic neurons are diminished (Ernfors et al. 1994b; Fox et al. 2013; Gacek and Khetarpal 1998), and motor neuron apoptosis increases (Usui et al. 2012; Woolley et al. 2005). NT-3 mutation leads to reduced number of muscle sensory neuron (Tessarollo et al. 1994) and fewer myenteric and submucosal neuron plexus in the enteric nervous system (Chalazonitis 2004). NT-4 knockout mice are viable but showed reduction in sensory neurons and long term memory (Liu et al. 1995; Smith et al. 2003; Xie et al. 2000).

Neurotrophins are required to maintain the neural stem cell niche. In the mouse sub-ependymal region, NT-3 can be secreted by the ependymal endothelia. It can slow down the cell proliferation through activation of the TrkC pathway, which induces the synthesis of the nitric oxide and promotes quiescence of the neural stem cells (Delgado et al. 2014). Virus-mediated overexpression or sustained delivery through conduit of NT-3 improves neuron stem cell survival, proliferation and differentiation in vitro (Lu et al. 2011; Tang et al. 2014; Zhu et al. 2012), as well as in the injured spinal cord of animal (Elliott Donaghue et al. 2015). NT-3 has a distinct effect in the cochlear nerve terminals and the inner hair cells by promoting synaptic regeneration post noise damage (Wan et al. 2014).

NT-3 is showed to facilitate BMSC survival and neuronal differentiation. Bone marrow cells co-cultured on poly lactic-acid-co-glycolic acid with NT-3 (Zhang et al. 2012), or with pharmacologically active microcarriers releasing NT-3 (Daviaud et al. 2015), have a significant increase of survival and neuronal differentiation. Moreover, transplantation of NT-3 overexpressed fibroblast into injured spinal cord improves the motor neuron response to electric stimuli (Arvanian et al. 2003). Overexpression of NT-3 also increases the survival and differentiation of BMSCs into neuron like cells (Dong et al. 2014; Gong et al. 2015; Yang et al. 2014). NT-3 transfected BMSCs or fibroblasts show stronger motor neuron axon regeneration, synaptic regeneration and remyelination after spinal cord injury in rodents (Arvanian et al. 2003; Liu et al. 2015; Thomas et al. 2014; Wang et al. 2014c). NT-3 and its receptor TrkC are found in the ovarian follicles and play a role in the follicle transition (Nilsson et al. 2009), indicating that NT-3 promotes germ cell differentiation.

NT-4/5 is less well studied compared to other neurotrophic factors. It has overlapping effect as BDNF in taste sensory neuron development (Huang and Krimm 2014). It also has synergistic effect with GDNF in promoting neuron survival in cultured embryonic rat brain slice (Meyer et al. 2001). NT-4/5 is reported to promote the oligodendrocyte precursors proliferation in culture (Scarisbrick et al. 2000). It also increases embryonic neural stem cell differentiation through inhibiting STAT3 phosphorylation (Shen et al. 2010). Overexpression of NT-4/5 protects the cochlear hair cell from by kanamycin toxicity and improves auditory function in guinea pig (Zheng et al. 2013). NT-4/5 and BDNF are found enriched in the umbilical cord blood, and may play a role in the hematopoietic stem cell proliferation (Fan et al. 2005).

GDNF Family

GDNF belongs to the GDNF family of ligands (GFL), which also includes neuturin (NRTN), artemin (ARTN) and persephin (PSPN). GFL binds to the GDNF family receptors (GFR). GDNF preferentially binds GFRα1, NRTN preferentially binds GFRα2, ARTN binds GFRα3, and PSPN binds GFRα4 (Airaksinen and Saarma 2002; Sariola and Saarma 2003).

GDNF is the most well-studied member of the family. GDNF was first purified as a potent neurotrophic factor that can enhance the survival of the dopaminergic neurons in the midbrain (Lin et al. 1993). It is reported to improve dopaminergic and enteric neuron survival, proliferation and migration (Airaksinen and Saarma 2002; Granholm et al. 2000; Sariola and Saarma 2003). GDNF preferentially binds to the GFRα1, which then activates the receptor tyrosine kinase RET or the neural cell adhesion molecule (Zhou et al. 2003). The PI3K/AKT, MEK/ERK, SRC/c-Jun kinase, FYN/focal adhesion kinase pathways have all been reported to be downstream of the GDNF signal (Charoy et al. 2012; Euteneuer et al. 2013; McAlhany et al. 2000; Oatley et al. 2007; Paratcha et al. 2003; Tang et al. 2002; Villegas et al. 2006).

GDNF knockout mice die soon after birth. There is renal agenesis due to undeveloped ureteric bud, early sequester of kidney development and complete absence of the enteric neurons (Costantini 2010; Pichel et al. 1996; Sanchez et al. 1996). The RET signaling pathway and the ETS transcription factors ETV4 and ETV5 have been demonstrated to be directly involved in the embryonic kidney development downstream of GDNF stimulation (Jain et al. 2006; Lu et al. 2009; Tang et al. 2002).

Recombinant human GDNF promotes the survival of the dopaminergic neurons in the midbrain, and has been tested for the treatment of Parkinson's disease in clinical trials (Lin et al. 1993). Amgen conducted a phase I clinical trial of monthly intraputamental injection of 15 μg/day recombinant human GDNF (Liatermin) in 34 Parkinson's patients. Based on the Unified Parkinson Disease Rating Scale and motor tests, it is inconclusive that GDNF application shows a clinical benefit in patients within 6 months of treatment (Lang et al. 2006). Another phase I trial was performed in the United Kingdom. GDNF (14.4–28.8 μg/day) was delivered to five patients by a pump imbedded in the abdomen and continuously infused into the putamen through a catheter for 6 months. There was fewer side effects compared to the direct injection route, and there was a substantial improvement in symptoms and Dopa influx up to 1 year of treatment in some patients (Gill et al. 2003; Love et al. 2005). Another Phase I trial conducted at the University of Kentucky also showed significant improvement of bilateral motor balance and gait in ten patients, who received unilateral intraputaminal infusion of up to 30 μg/day GDNF for 8 weeks through a catheter (Slevin et al. 2005). Several preclinical studies performed on rodents and primates indicated that delivery of sufficient GDNF to the lesion improved neuronal regeneration and reduces the symptoms (Bartus et al. 2011; Gasmi et al. 2007; Richardson et al. 2011). In addition, over expression of GNDF through virus (Kells et al. 2004) or stable cell line (Pineda et al. 2007) has been shown to improve the neuronal survival in the animal models of Huntington's disease.

Outside of the nervous system and renal development, GDNF maintains the sperm stem cell pool by promoting spermatogonial self-renewal. It also regulates spermatogenesis and sperm differentiation (Chen et al. 2005; Meng et al. 2000). Similar to the ureteric bud generation pathway, RET signal is essential in mediating spermatogenesis (Jijiwa et al. 2008; Oatley et al. 2007). GDNF also protects the salivary gland from radiation induced damage by promoting the salivary gland stem cell regeneration and proliferation (Xiao et al. 2014). Human mesenchymal stem cells also release GDNF, which in turn facilitates the motor neuron regeneration in rats (Krakora et al. 2013).

GDNF family member NRTN induces dopaminergic neuron regeneration (Liu et al. 2009; Vourc'h et al. 2005). NRTN is also reported to support enteric neuron survival and proliferation through RET pathway (Heuckeroth et al. 1998). It is also involved in mouse embryonic salivary gland development and regeneration after radiation damage (Hai et al. 2014; Knox et al. 2013).

Another GDNF family member PSPN has very similar function as GDNF, such as supporting motor neuron survival (Milbrandt et al. 1998). It is known to increase the survival of both dopaminergic neurons (Akerud et al. 2002; Roussa et al. 2008) and mesenchymal stem cells (Yin et al. 2014).

GDNF family members and their signaling pathways have been associated with cancer cell growth, invasion, metastasis and resistance to therapy (Poteriaev and Saarma 2001). Specifically they have been linked to the growth of neuroblastoma (Komminoth et al. 1996), breast cancer (Banerjee et al. 2012; Ding et al. 2014), small cell lung cancer (Rudin et al. 2014), thyroid cancer (Hong et al. 2008; Wells and Santoro 2009), pancreatic cancer (Donahue and Hines 2009) and testicular cancer (Sariola and Meng 2003). GDNF promotes perineural invasion and metastasis of brain tumor (Ilhan-Mutlu et al. 2013), head and neck squamous cell carcinoma (Roh et al. 2015), glioma (Shabtay-Orbach et al. 2014), pancreatic cancer (He et al. 2014; Wang et al. 2014b) and colon cancer (Huang et al. 2014). GDNF family members also increase resistance to the chemotherapy in the prostate (Huber et al. 2015) and breast cancer (Ding et al. 2014; Morandi et al. 2013), while reduction of GDNF level decreased metastasis of mammary gland cancer and related pain in the bone of rat (Meng et al. 2015). These research works suggest that GDNF level is also important in regulating the cancer cell proliferation in multiple non-neuronal tissues.

Conclusion

Here, we reviewed different neurotrophic factors and their potential clinical applications in tissue regeneration. The neurotrophic factors not only nourish neurons during development, they are also critical in regulating survival, proliferation and differentiation of neuronal and non-neuronal cells. Some are also reported to be involved in abnormal cell behavior that leads to neoplasms. Further investigation on the role of these neurotrophic factors and the mechanism of actions will help to exploit their function for future functional restoration of organs in patients.

References

  1. Acheson A, Conover JC, Fandl JP, et al. A BDNF autocrine loop in adult sensory neurons prevents cell death. Nature. 1995;374:450–453. doi: 10.1038/374450a0. [DOI] [PubMed] [Google Scholar]
  2. Airaksinen MS, Saarma M. The GDNF family: signalling, biological functions and therapeutic value. Nat Rev Neurosci. 2002;3:383–394. doi: 10.1038/nrn812. [DOI] [PubMed] [Google Scholar]
  3. Akerud P, Holm PC, Castelo-Branco G, et al. Persephin-overexpressing neural stem cells regulate the function of nigral dopaminergic neurons and prevent their degeneration in a model of Parkinson's disease. Mol Cell Neurosci. 2002;21:205–222. doi: 10.1006/mcne.2002.1171. [DOI] [PubMed] [Google Scholar]
  4. Aloe L, Rocco ML, Bianchi P, et al. Nerve growth factor: from the early discoveries to the potential clinical use. J Transl Med. 2012;10:239. doi: 10.1186/1479-5876-10-239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Arvanian VL, Horner PJ, Gage FH, et al. Chronic neurotrophin-3 strengthens synaptic connections to motoneurons in the neonatal rat. J Neurosci. 2003;23:8706–8712. doi: 10.1523/JNEUROSCI.23-25-08706.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Banerjee A, Qian P, Wu ZS, et al. Artemin stimulates radio- and chemo-resistance by promoting TWIST1-BCL-2-dependent cancer stem cell-like behavior in mammary carcinoma cells. J Biol Chem. 2012;287:42502–42515. doi: 10.1074/jbc.M112.365163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Barde YA, Edgar D, Thoenen H. Purification of a new neurotrophic factor from mammalian brain. EMBO J. 1982;1:549–553. doi: 10.1002/j.1460-2075.1982.tb01207.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bartus RT, Brown L, Wilson A, et al. Properly scaled and targeted AAV2-NRTN (neurturin) to the substantia nigra is safe, effective and causes no weight loss: support for nigral targeting in Parkinson's disease. Neurobiol Dis. 2011;44:38–52. doi: 10.1016/j.nbd.2011.05.026. [DOI] [PubMed] [Google Scholar]
  9. Becker E, Soler RM, Yuste VJ, et al. Development of survival responsiveness to brain-derived neurotrophic factor, neurotrophin 3 and neurotrophin 4/5, but not to nerve growth factor, in cultured motoneurons from chick embryo spinal cord. J Neurosci. 1998;18:7903–7911. doi: 10.1523/JNEUROSCI.18-19-07903.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Berg J, Roch M, Altschüler J, et al. Human adipose-derived mesenchymal stem cells improve motor functions and are neuroprotective in the 6-hydroxydopamine-rat model for Parkinson's disease when cultured in monolayer cultures but suppress hippocampal neurogenesis and hippocampal memory function when cultured in spheroids. Stem Cell Rev. 2015;11:133–419. doi: 10.1007/s12015-014-9551-y. [DOI] [PubMed] [Google Scholar]
  11. Berkemeier LR, Winslow JW, Kaplan DR, et al. Neurotrophin-5: a novel neurotrophic factor that activates trk and trkB. Neuron. 1991;7:857–866. doi: 10.1016/0896-6273(91)90287-a. [DOI] [PubMed] [Google Scholar]
  12. Borrell-Pages M, Canals JM, Cordelieres FP, et al. Cystamine and cysteamine increase brain levels of BDNF in Huntington disease via HSJ1b and transglutaminase. J Clin Invest. 2006;116:1410–1424. doi: 10.1172/JCI27607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Caporali A, Sala-Newby GB, Meloni M, et al. Identification of the prosurvival activity of nerve growth factor on cardiac myocytes. Cell Death Differ. 2008;15:299–311. doi: 10.1038/sj.cdd.4402263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Chalazonitis A. Neurotrophin-3 in the development of the enteric nervous system. Prog Brain Res. 2004;146:243–263. doi: 10.1016/S0079-6123(03)46016-0. [DOI] [PubMed] [Google Scholar]
  15. Charoy C, Nawabi H, Reynaud F, et al. gdnf activates midline repulsion by Semaphorin3B via NCAM during commissural axon guidance. Neuron. 2012;75:1051–1066. doi: 10.1016/j.neuron.2012.08.021. [DOI] [PubMed] [Google Scholar]
  16. Chen C, Ouyang W, Grigura V, et al. ERM is required for transcriptional control of the spermatogonial stem cell niche. Nature. 2005;436:1030–1034. doi: 10.1038/nature03894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Chen LX, Ma SM, Zhang P, et al. Neuroprotective effects of oligodendrocyte progenitor cell transplantation in premature rat brain following hypoxic-ischemic injury. PLoS One. 2015;10:e0115997. doi: 10.1371/journal.pone.0115997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chiaretti A, Antonelli A, Genovese O, et al. Intraventricular nerve growth factor infusion improves cerebral blood flow and stimulates doublecortin expression in two infants with hypoxic-ischemic brain injury. Neurol Res. 2008;30:223–228. doi: 10.1179/016164107X247948. [DOI] [PubMed] [Google Scholar]
  19. Conover JC, Erickson JT, Katz DM, et al. Neuronal deficits, not involving motor neurons, in mice lacking BDNF and/or NT4. Nature. 1995;375:235–238. doi: 10.1038/375235a0. [DOI] [PubMed] [Google Scholar]
  20. Costantini F. GDNF/Ret signaling and renal branching morphogenesis: From mesenchymal signals to epithelial cell behaviors. Organogenesis. 2010;6:252–262. doi: 10.4161/org.6.4.12680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Criscuolo C, Fabiani C, Bonadonna C, et al. BDNF prevents amyloid-dependent impairment of LTP in the entorhinal cortex by attenuating p38 MAPK phosphorylation. Neurobiol Aging. 2015;36:1303–1309. doi: 10.1016/j.neurobiolaging.2014.11.016. [DOI] [PubMed] [Google Scholar]
  22. Dadon-Nachum M, Ben-Zur T, Srugo I, et al. Therapeutic effect of myogenic cells modified to express neurotrophic factors in a rat model of sciatic nerve injury. J Stem Cells Regen Med. 2012;8:21–27. doi: 10.46582/jsrm.0801004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Daviaud N, Garbayo E, Sindji L, et al. Survival, differentiation, and neuroprotective mechanisms of human stem cells complexed with neurotrophin-3-releasing pharmacologically active microcarriers in an ex vivo model of Parkinson's disease. Stem Cells Transl Med. 2015;4:670–684. doi: 10.5966/sctm.2014-0139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. de Munter JP, Melamed E, Wolters E. Stem cell grafting in parkinsonism–why, how and when. Parkinsonism Relat Disord. 2014;20(Suppl 1):S150–S153. doi: 10.1016/S1353-8020(13)70036-1. [DOI] [PubMed] [Google Scholar]
  25. Delgado AC, Ferron SR, Vicente D, et al. Endothelial NT-3 delivered by vasculature and CSF promotes quiescence of subependymal neural stem cells through nitric oxide induction. Neuron. 2014;83:572–585. doi: 10.1016/j.neuron.2014.06.015. [DOI] [PubMed] [Google Scholar]
  26. Ding K, Banerjee A, Tan S, et al. Artemin, a member of the glial cell line-derived neurotrophic factor family of ligands, is HER2-regulated and mediates acquired trastuzumab resistance by promoting cancer stem cell-like behavior in mammary carcinoma cells. J Biol Chem. 2014;289:16057–16071. doi: 10.1074/jbc.M113.529552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Donahue TR, Hines OJ. CXCR2 and RET single nucleotide polymorphisms in pancreatic cancer. World J Surg. 2009;33:710–715. doi: 10.1007/s00268-008-9826-z. [DOI] [PubMed] [Google Scholar]
  28. Dong Y, Yang L, Yang L, et al. Transplantation of neurotrophin-3-transfected bone marrow mesenchymal stem cells for the repair of spinal cord injury. Neural Regen Res. 2014;9:1520–1524. doi: 10.4103/1673-5374.139478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Drake CT, Milner TA, Patterson SL. Ultrastructural localization of full-length trkB immunoreactivity in rat hippocampus suggests multiple roles in modulating activity-dependent synaptic plasticity. J Neurosci. 1999;19:8009–8026. doi: 10.1523/JNEUROSCI.19-18-08009.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Durand C, Robin C, Bollerot K, et al. Embryonic stromal clones reveal developmental regulators of definitive hematopoietic stem cells. Proc Natl Acad Sci USA. 2007;104:20838–20843. doi: 10.1073/pnas.0706923105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Elliott Donaghue I, Tator CH, et al. Sustained delivery of bioactive neurotrophin-3 to the injured spinal cord. Biomater Sci. 2015;3:65–72. doi: 10.1039/c4bm00311j. [DOI] [PubMed] [Google Scholar]
  32. Erickson JT, Conover JC, Borday V, et al. Mice lacking brain-derived neurotrophic factor exhibit visceral sensory neuron losses distinct from mice lacking NT4 and display a severe developmental deficit in control of breathing. J Neurosci. 1996;16:5361–5371. doi: 10.1523/JNEUROSCI.16-17-05361.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Eriksdotter-Jonhagen M, Linderoth B, Lind G, et al. Encapsulated cell biodelivery of nerve growth factor to the Basal forebrain in patients with Alzheimer's disease. Dement Geriatr Cogn Disord. 2012;33:18–28. doi: 10.1159/000336051. [DOI] [PubMed] [Google Scholar]
  34. Ernfors P, Lee KF, Jaenisch R. Mice lacking brain-derived neurotrophic factor develop with sensory deficits. Nature. 1994a;368:147–150. doi: 10.1038/368147a0. [DOI] [PubMed] [Google Scholar]
  35. Ernfors P, Lee KF, Kucera J, et al. Lack of neurotrophin-3 leads to deficiencies in the peripheral nervous system and loss of limb proprioceptive afferents. Cell. 1994b;77:503–512. doi: 10.1016/0092-8674(94)90213-5. [DOI] [PubMed] [Google Scholar]
  36. Espinera AR, Ogle ME, Gu X, et al. Citalopram enhances neurovascular regeneration and sensorimotor functional recovery after ischemic stroke in mice. Neuroscience. 2013;247:1–11. doi: 10.1016/j.neuroscience.2013.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Euteneuer S, Yang KH, Chavez E, et al. Glial cell line-derived neurotrophic factor (GDNF) induces neuritogenesis in the cochlear spiral ganglion via neural cell adhesion molecule (NCAM) Mol Cell Neurosci. 2013;54:30–43. doi: 10.1016/j.mcn.2012.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Fan CG, Zhang QJ, Tang FW, et al. Human umbilical cord blood cells express neurotrophic factors. Neurosci Lett. 2005;380:322–325. doi: 10.1016/j.neulet.2005.01.070. [DOI] [PubMed] [Google Scholar]
  39. Ferreira D, Westman E, Eyjolfsdottir H, et al. Brain changes in Alzheimer's disease patients with implanted encapsulated cells releasing nerve growth factor. J Alzheimer's Dis. 2015;43:1059–1072. doi: 10.3233/JAD-141068. [DOI] [PubMed] [Google Scholar]
  40. Ferrer I, Marin C, Rey MJ, et al. BDNF and full-length and truncated TrkB expression in Alzheimer disease. Implications in therapeutic strategies. J Neuropathol Exp Neurol. 1999;58:729–739. doi: 10.1097/00005072-199907000-00007. [DOI] [PubMed] [Google Scholar]
  41. Fox EA, Biddinger JE, Baquet ZC, et al. Loss of neurotrophin-3 from smooth muscle disrupts vagal gastrointestinal afferent signaling and satiation. Am J Physiol Regul Integr Comp Physiol. 2013;305:R1307–R1322. doi: 10.1152/ajpregu.00337.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Frerking M, Malenka RC, Nicoll RA. Brain-derived neurotrophic factor (BDNF) modulates inhibitory, but not excitatory, transmission in the CA1 region of the hippocampus. J Neurophysiol. 1998;80:3383–3386. doi: 10.1152/jn.1998.80.6.3383. [DOI] [PubMed] [Google Scholar]
  43. Frontera JL, Cervino AS, Jungblut LD, et al. Brain-derived neurotrophic factor (BDNF) expression in normal and regenerating olfactory epithelium of Xenopus laevis. Ann Anat. 2015;198:41–48. doi: 10.1016/j.aanat.2014.10.010. [DOI] [PubMed] [Google Scholar]
  44. Gacek RR, Khetarpal U. Neurotrophin 3, not brain-derived neurotrophic factor or neurotrophin 4, knockout mice have delay in vestibular compensation after unilateral labyrinthectomy. Laryngoscope. 1998;108:671–678. doi: 10.1097/00005537-199805000-00009. [DOI] [PubMed] [Google Scholar]
  45. Gasmi M, Herzog CD, Brandon EP, et al. Striatal delivery of neurturin by CERE-120, an AAV2 vector for the treatment of dopaminergic neuron degeneration in Parkinson's disease. Mol Ther. 2007;15:62–68. doi: 10.1038/sj.mt.6300010. [DOI] [PubMed] [Google Scholar]
  46. Gill SS, Patel NK, Hotton GR, et al. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat Med. 2003;9:589–595. doi: 10.1038/nm850. [DOI] [PubMed] [Google Scholar]
  47. Goekint M, Roelands B, Heyman E, et al. Influence of citalopram and environmental temperature on exercise-induced changes in BDNF. Neurosci Lett. 2011;494:150–154. doi: 10.1016/j.neulet.2011.03.001. [DOI] [PubMed] [Google Scholar]
  48. Gong Y, Wang H, Xia H. Stable transfection into rat bone marrow mesenchymal stem cells by lentivirus-mediated NT-3. Mol Med Rep. 2015;11:367–733. doi: 10.3892/mmr.2014.2727. [DOI] [PubMed] [Google Scholar]
  49. Granholm AC, Reyland M, Albeck D, et al. Glial cell line-derived neurotrophic factor is essential for postnatal survival of midbrain dopamine neurons. J Neurosci. 2000;20:3182–3190. doi: 10.1523/JNEUROSCI.20-09-03182.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Gransee HM, Zhan WZ, Sieck GC, et al. Localized delivery of brain-derived neurotrophic factor-expressing mesenchymal stem cells enhances functional recovery following cervical spinal cord injury. J Neurotrauma. 2015;32:185–193. doi: 10.1089/neu.2014.3464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Haghighi M, Salehi I, Erfani P, et al. Additional ECT increases BDNF-levels in patients suffering from major depressive disorders compared to patients treated with citalopram only. J Psychiatr Res. 2013;47:908–915. doi: 10.1016/j.jpsychires.2013.03.006. [DOI] [PubMed] [Google Scholar]
  52. Hai B, Qin L, Yang Z, et al. Transient activation of hedgehog pathway rescued irradiation-induced hyposalivation by preserving salivary stem/progenitor cells and parasympathetic innervation. Clin Cancer Res. 2014;20:140–150. doi: 10.1158/1078-0432.CCR-13-1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Hall J, Thomas KL, Everitt BJ. Rapid and selective induction of BDNF expression in the hippocampus during contextual learning. Nat Neurosci. 2000;3:533–535. doi: 10.1038/75698. [DOI] [PubMed] [Google Scholar]
  54. Han C, Zhang L, Song L, et al. Human adipose-derived mesenchymal stem cells: a better cell source for nervous system regeneration. Chin Med J. 2014;127:329–337. [PubMed] [Google Scholar]
  55. Han ZM, Huang HM, Wang FF. Brain-derived neurotrophic factor gene-modified bone marrow mesenchymal stem cells. Exp Ther Med. 2015;9:519–522. doi: 10.3892/etm.2014.2113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Hata T, Sakata N, Yoshimatsu G, et al. Nerve growth factor improves survival and function of transplanted islets via TrkA-mediated beta cell proliferation and revascularization. Transplantation. 2015;99:1132–1143. doi: 10.1097/TP.0000000000000655. [DOI] [PubMed] [Google Scholar]
  57. He S, Chen CH, Chernichenko N, et al. GFRalpha1 released by nerves enhances cancer cell perineural invasion through GDNF-RET signaling. Proc Natl Acad Sci USA. 2014;111:E2008–E2017. doi: 10.1073/pnas.1402944111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Heese K, Low JW, Inoue N. Nerve growth factor, neural stem cells and Alzheimer's disease. Neuro-Signals. 2006–2007;15:1–12. doi: 10.1159/000094383. [DOI] [PubMed] [Google Scholar]
  59. Hefti F, Will B. Nerve growth factor is a neurotrophic factor for forebrain cholinergic neurons; implications for Alzheimer's disease. J Neural Transm Suppl. 1987;24:309–315. [PubMed] [Google Scholar]
  60. Heuckeroth RO, Lampe PA, Johnson EM, et al. Neurturin and GDNF promote proliferation and survival of enteric neuron and glial progenitors in vitro. Dev Biol. 1998;200:116–129. doi: 10.1006/dbio.1998.8955. [DOI] [PubMed] [Google Scholar]
  61. Hohn A, Leibrock J, Bailey K, et al. Identification and characterization of a novel member of the nerve growth factor/ brain-derived neurotrophic factor family. Nature. 1990;344:339–341. doi: 10.1038/344339a0. [DOI] [PubMed] [Google Scholar]
  62. Hong D, Ye L, Gagel R, et al. Medullary thyroid cancer: targeting the RET kinase pathway with sorafenib/tipifarnib. Mol Cancer Ther. 2008;7:1001–1006. doi: 10.1158/1535-7163.MCT-07-2422. [DOI] [PubMed] [Google Scholar]
  63. Howells DW, Porritt MJ, Wong JY, et al. Reduced BDNF mRNA expression in the Parkinson's disease substantia nigra. Exp Neurol. 2000;166:127–135. doi: 10.1006/exnr.2000.7483. [DOI] [PubMed] [Google Scholar]
  64. Huang T, Krimm RF. BDNF and NT4 play interchangeable roles in gustatory development. Dev Biol. 2014;386:308–320. doi: 10.1016/j.ydbio.2013.12.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Huang EJ, Reichardt LF. Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci. 2001;24:677–736. doi: 10.1146/annurev.neuro.24.1.677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Huang XQ, Zhu BD. Effect of nerve growth factor on erythropoiesis in mice and its underlying mechanism. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2008;16:1365–1371. [PubMed] [Google Scholar]
  67. Huang XQ, Zhu BD, Jiang-Yang-ze-ren The effect of NGF on hematopoiesis of granulocyte-macrophage in normal and radiation, chemistry injury mice. Sichuan Da Xue Xue Bao Yi Xue Ban. 2008;39:757–762. [PubMed] [Google Scholar]
  68. Huang SM, Chen TS, Chiu CM, et al. GDNF increases cell motility in human colon cancer through VEGF-VEGFR1 interaction. Endocr Relat Cancer. 2014;21:73–84. doi: 10.1530/ERC-13-0351. [DOI] [PubMed] [Google Scholar]
  69. Huber RM, Lucas JM, Gomez-Sarosi LA, et al. DNA damage induces GDNF secretion in the tumor microenvironment with paracrine effects promoting prostate cancer treatment resistance. Oncotarget. 2015;6:2134–2147. doi: 10.18632/oncotarget.3040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Hussey AJ, Winardi M, Wilson J, et al. Pancreatic islet transplantation using vascularised chambers containing nerve growth factor ameliorates hyperglycaemia in diabetic mice. Cells Tissues Organs. 2010;191:382–393. doi: 10.1159/000276595. [DOI] [PubMed] [Google Scholar]
  71. Ilhan-Mutlu A, Wagner L, Widhalm G, et al. Exploratory investigation of eight circulating plasma markers in brain tumor patients. Neurosurg Rev. 2013;36:45–55. doi: 10.1007/s10143-012-0401-6. discussion 55–56. [DOI] [PubMed] [Google Scholar]
  72. Ip NY, Ibanez CF, Nye SH, et al. Mammalian neurotrophin-4: structure, chromosomal localization, tissue distribution, and receptor specificity. Proc Natl Acad Sci USA. 1992;89:3060–3064. doi: 10.1073/pnas.89.7.3060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Jain S, Encinas M, Johnson EM, Jr, et al. Critical and distinct roles for key RET tyrosine docking sites in renal development. Genes Dev. 2006;20:321–333. doi: 10.1101/gad.1387206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Jain V, Baitharu I, Prasad D, et al. Enriched environment prevents hypobaric hypoxia induced memory impairment and neurodegeneration: role of BDNF/PI3K/GSK3beta pathway coupled with CREB activation. PLoS One. 2013;8:e62235. doi: 10.1371/journal.pone.0062235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Jeon MT, Nam JH, Shin W, et al. In vivo AAV1 transduction with hRheb(S16H) protects hippocampal neurons by BDNF production. Mol Ther. 2015;23:445–455. doi: 10.1038/mt.2014.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Ji R, Meng L, Jiang X, et al. TAM receptors support neural stem cell survival, proliferation and neuronal differentiation. PLoS One. 2014;9:e115140. doi: 10.1371/journal.pone.0115140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Jijiwa M, Kawai K, Fukihara J, et al. GDNF-mediated signaling via RET tyrosine 1062 is essential for maintenance of spermatogonial stem cells. Genes Cells. 2008;13:365–374. doi: 10.1111/j.1365-2443.2008.01171.x. [DOI] [PubMed] [Google Scholar]
  78. Jones KR, Farinas I, Backus C, et al. Targeted disruption of the BDNF gene perturbs brain and sensory neuron development but not motor neuron development. Cell. 1994;76:989–999. doi: 10.1016/0092-8674(94)90377-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Jovanovic JN, Thomas P, Kittler JT, et al. Brain-derived neurotrophic factor modulates fast synaptic inhibition by regulating GABA(A) receptor phosphorylation, activity, and cell-surface stability. J Neurosci. 2004;24:522–530. doi: 10.1523/JNEUROSCI.3606-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Kang H, Schuman EM. Long-lasting neurotrophin-induced enhancement of synaptic transmission in the adult hippocampus. Science. 1995;267:1658–1662. doi: 10.1126/science.7886457. [DOI] [PubMed] [Google Scholar]
  81. Kang H, Welcher AA, Shelton D, et al. Neurotrophins and time: different roles for TrkB signaling in hippocampal long-term potentiation. Neuron. 1997;19:653–664. doi: 10.1016/s0896-6273(00)80378-5. [DOI] [PubMed] [Google Scholar]
  82. Kells AP, Fong DM, Dragunow M, et al. AAV-mediated gene delivery of BDNF or GDNF is neuroprotective in a model of Huntington disease. Mol Ther. 2004;9:682–688. doi: 10.1016/j.ymthe.2004.02.016. [DOI] [PubMed] [Google Scholar]
  83. Kernie SG, Liebl DJ, Parada LF. BDNF regulates eating behavior and locomotor activity in mice. EMBO J. 2000;19:1290–1300. doi: 10.1093/emboj/19.6.1290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Kerschensteiner M, Stadelmann C, Dechant G, et al. Neurotrophic cross-talk between the nervous and immune systems: implications for neurological diseases. Ann Neurol. 2003;53:292–304. doi: 10.1002/ana.10446. [DOI] [PubMed] [Google Scholar]
  85. Kim SJ, Choi SW, Hur KJ, et al. Synergistic effect of mesenchymal stem cells infected with recombinant adenovirus expressing human BDNF on erectile function in a rat model of cavernous nerve injury. Korean J Urol. 2012;53:726–732. doi: 10.4111/kju.2012.53.10.726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Klein R, Lamballe F, Bryant S, et al. The trkB tyrosine protein kinase is a receptor for neurotrophin-4. Neuron. 1992;8:947–956. doi: 10.1016/0896-6273(92)90209-v. [DOI] [PubMed] [Google Scholar]
  87. Knox SM, Lombaert IM, Haddox CL, et al. Parasympathetic stimulation improves epithelial organ regeneration. Nat Commun. 2013;4:1494. doi: 10.1038/ncomms2493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Komminoth P, Roth J, Muletta-Feurer S, et al. RET proto-oncogene point mutations in sporadic neuroendocrine tumors. J Clin Endocrinol Metab. 1996;81:2041–2046. doi: 10.1210/jcem.81.6.8964826. [DOI] [PubMed] [Google Scholar]
  89. Korsching S, Heumann R, Thoenen H, et al. Cholinergic denervationof the rat hippocampus byfimbrial transection leads to a transient accumulation of nerve growth factor (NGF) without change in mRNANGF content. Neurosci Lett. 1986;66:175–180. doi: 10.1016/0304-3940(86)90186-2. [DOI] [PubMed] [Google Scholar]
  90. Korte M, Carroll P, Wolf E, et al. Hippocampal long-term potentiation is impaired in mice lacking brain-derived neurotrophic factor. Proc Natl Acad Sci USA. 1995;92:8856–8860. doi: 10.1073/pnas.92.19.8856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Korte M, Griesbeck O, Gravel C, et al. Virus-mediated gene transfer into hippocampal CA1 region restores long-term potentiation in brain-derived neurotrophic factor mutant mice. Proc Natl Acad Sci USA. 1996;93:12547–12552. doi: 10.1073/pnas.93.22.12547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Krakora D, Mulcrone P, Meyer M, et al. Synergistic effects of GDNF and VEGF on lifespan and disease progression in a familial ALS rat model. Mol Ther. 2013;21:1602–1610. doi: 10.1038/mt.2013.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Kurozumi K, Nakamura K, Tamiya T, et al. Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion model. Mol Ther. 2005;11:96–104. doi: 10.1016/j.ymthe.2004.09.020. [DOI] [PubMed] [Google Scholar]
  94. Ladea M, Bran M. Brain derived neurotrophic factor (BDNF) levels in depressed women treated with open-label escitalopram. Psychiatr Danub. 2013;25:128–132. [PubMed] [Google Scholar]
  95. Lang AE, Gill S, Patel NK, et al. Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann Neurol. 2006;59:459–466. doi: 10.1002/ana.20737. [DOI] [PubMed] [Google Scholar]
  96. Larrieta ME, Vital P, Mendoza-Rodriguez A, et al. Nerve growth factor increases in pancreatic beta cells after streptozotocin-induced damage in rats. Exp Biol Med. 2006;231:396–402. doi: 10.1177/153537020623100405. [DOI] [PubMed] [Google Scholar]
  97. Lauterborn JC, Pineda E, Chen LY, et al. Ampakines cause sustained increases in brain-derived neurotrophic factor signaling at excitatory synapses without changes in AMPA receptor subunit expression. Neuroscience. 2009;159:283–925. doi: 10.1016/j.neuroscience.2008.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Lavasani M, Lu A, Peng H, et al. Nerve growth factor improves the muscle regeneration capacity of muscle stem cells in dystrophic muscle. Hum Gene Ther. 2006;17:180–192. doi: 10.1089/hum.2006.17.180. [DOI] [PubMed] [Google Scholar]
  99. Lawn S, Krishna N, Pisklakova A, et al. Neurotrophin signaling via TrkB and TrkC receptors promotes the growth of brain tumor-initiating cells. J Biol Chem. 2015;290:3814–3824. doi: 10.1074/jbc.M114.599373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Lee KF, Li E, Huber L, et al. Targeted mutation of the gene encoding the low affinity NGF receptor p75 leads to deficits in the peripheral sensory nervous system. Cell. 1992;69:737–749. doi: 10.1016/0092-8674(92)90286-l. [DOI] [PubMed] [Google Scholar]
  101. Levi-Montalcini R, Hamburger V. Selective growth stimulating effects of mouse sarcoma on the sensory and sympathetic nervous system of the chick embryo. J Exp Zool. 1951;116:321–361. doi: 10.1002/jez.1401160206. [DOI] [PubMed] [Google Scholar]
  102. Lin LF, Doherty DH, Lile JD, et al. GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science. 1993;260:1130–1132. doi: 10.1126/science.8493557. [DOI] [PubMed] [Google Scholar]
  103. Linnarsson S, Bjorklund A, Ernfors P. Learning deficit in BDNF mutant mice. Eur J Neurosci. 1997;9:2581–2587. doi: 10.1111/j.1460-9568.1997.tb01687.x. [DOI] [PubMed] [Google Scholar]
  104. Liu X, Ernfors P, Wu H, et al. Sensory but not motor neuron deficits in mice lacking NT4 and BDNF. Nature. 1995;375:238–241. doi: 10.1038/375238a0. [DOI] [PubMed] [Google Scholar]
  105. Liu WG, Wang XJ, Lu GQ, et al. Retraction: dopaminergic regeneration by neurturin-overexpressing c17.2 neural stem cells in a rat model of Parkinson's disease. Mol Neurodegener. 2009;4:45. doi: 10.1186/1750-1326-4-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Liu Y, Tao L, Fu X, et al. BDNF protects retinal neurons from hyperglycemia through the TrkB/ERK/MAPK pathway. Mol Med Rep. 2013;7:1773–1778. doi: 10.3892/mmr.2013.1433. [DOI] [PubMed] [Google Scholar]
  107. Liu Z, He B, Zhang RY, et al. Electroacupuncture promotes the differentiation of transplanted bone marrow mesenchymal stem cells pre-induced with neurotrophin-3 and retinoic acid into oligodendrocyte-like cells in demyelinated spinal cord of rats. Cell Transpl. 2015;24:1265–1281. doi: 10.3727/096368914X682099. [DOI] [PubMed] [Google Scholar]
  108. Long X, Olszewski M, Huang W, et al. Neural cell differentiation in vitro from adult human bone marrow mesenchymal stem cells. Stem Cells Dev. 2005;14:65–69. doi: 10.1089/scd.2005.14.65. [DOI] [PubMed] [Google Scholar]
  109. Love S, Plaha P, Patel NK, et al. Glial cell line-derived neurotrophic factor induces neuronal sprouting in human brain. Nat Med. 2005;11:703–704. doi: 10.1038/nm0705-703. [DOI] [PubMed] [Google Scholar]
  110. Lu BC, Cebrian C, Chi X, et al. Etv4 and Etv5 are required downstream of GDNF and Ret for kidney branching morphogenesis. Nat Genet. 2009;41:1295–1302. doi: 10.1038/ng.476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Lu HX, Hao ZM, Jiao Q, et al. Neurotrophin-3 gene transduction of mouse neural stem cells promotes proliferation and neuronal differentiation in organotypic hippocampal slice cultures. Med Sci Monit. 2011;17:BR305–BR311. doi: 10.12659/MSM.882039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Lu J, Frerich JM, Turtzo LC, et al. Histone deacetylase inhibitors are neuroprotective and preserve NGF-mediated cell survival following traumatic brain injury. Proc Natl Acad Sci USA. 2013;110:10747–10752. doi: 10.1073/pnas.1308950110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Lyons WE, Mamounas LA, Ricaurte GA, et al. Brain-derived neurotrophic factor-deficient mice develop aggressiveness and hyperphagia in conjunction with brain serotonergic abnormalities. Proc Natl Acad Sci USA. 1999;96:15239–15244. doi: 10.1073/pnas.96.26.15239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Mahmoud AI, O'Meara CC, Gemberling M, et al. Nerves regulate cardiomyocyte proliferation and heart regeneration. Dev Cell. 2015;34:387–399. doi: 10.1016/j.devcel.2015.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Maisonpierre PC, Belluscio L, Squinto S, et al. Neurotrophin-3: a neurotrophic factor related to NGF and BDNF. Science. 1990;247(4949 Pt 1):1446–1451. doi: 10.1126/science.247.4949.1446. [DOI] [PubMed] [Google Scholar]
  116. Mandel RJ. CERE-110, an adeno-associated virus-based gene delivery vector expressing human nerve growth factor for the treatment of Alzheimer's disease. Curr Opin Mol Ther. 2010;12:240–247. [PubMed] [Google Scholar]
  117. Martinez-Serrano A, Fischer W, Soderstrom S, et al. Long-term functional recovery from age-induced spatial memory impairments by nerve growth factor gene transfer to the rat basal forebrain. Proc Natl Acad Sci USA. 1996;93:6355–6360. doi: 10.1073/pnas.93.13.6355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Marty S, Berninger B, Carroll P, et al. GABAergic stimulation regulates the phenotype of hippocampal interneurons through the regulation of brain-derived neurotrophic factor. Neuron. 1996;16:565–570. doi: 10.1016/s0896-6273(00)80075-6. [DOI] [PubMed] [Google Scholar]
  119. Matsuda H, Coughlin MD, Bienenstock J, et al. Nerve growth factor promotes human hemopoietic colony growth and differentiation. Proc Natl Acad Sci USA. 1988;85:6508–6512. doi: 10.1073/pnas.85.17.6508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. McAlhany RE, Jr, West JR, Miranda RC. Glial-derived neurotrophic factor (GDNF) prevents ethanol-induced apoptosis and JUN kinase phosphorylation. Brain Res Dev Brain Res. 2000;119:209–216. doi: 10.1016/s0165-3806(99)00171-6. [DOI] [PubMed] [Google Scholar]
  121. Meloni M, Caporali A, Graiani G, et al. Nerve growth factor promotes cardiac repair following myocardial infarction. Circ Res. 2010;106:1275–1284. doi: 10.1161/CIRCRESAHA.109.210088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Memberg SP, Hall AK. Proliferation, differentiation, and survival of rat sensory neuron precursors in vitro require specific trophic factors. Mol Cell Neurosci. 1995;6:323–335. doi: 10.1006/mcne.1995.1025. [DOI] [PubMed] [Google Scholar]
  123. Meng X, Lindahl M, Hyvonen ME, et al. Regulation of cell fate decision of undifferentiated spermatogonia by GDNF. Science. 2000;287:1489–1493. doi: 10.1126/science.287.5457.1489. [DOI] [PubMed] [Google Scholar]
  124. Meng FF, Xu Y, Dan QQ, et al. Intrathecal injection of lentivirus-mediated GDNF interference RNA relieves bone cancer induced pain in rats. Cancer Sci. 2015;106:430–437. doi: 10.1111/cas.12609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Meyer M, Matarredona ER, Seiler RW, et al. Additive effect of glial cell line-derived neurotrophic factor and neurotrophin-4/5 on rat fetal nigral explant cultures. Neuroscience. 2001;108:273–284. doi: 10.1016/s0306-4522(01)00418-3. [DOI] [PubMed] [Google Scholar]
  126. Miao G, Mace J, Kirby M, et al. Beneficial effects of nerve growth factor on islet transplantation. Transpl Proc. 2005;37:3490–3492. doi: 10.1016/j.transproceed.2005.09.057. [DOI] [PubMed] [Google Scholar]
  127. Miao G, Mace J, Kirby M, et al. In vitro and in vivo improvement of islet survival following treatment with nerve growth factor. Transplantation. 2006;81:519–524. doi: 10.1097/01.tp.0000200320.16723.b3. [DOI] [PubMed] [Google Scholar]
  128. Milbrandt J, de Sauvage FJ, Fahrner TJ, et al. Persephin, a novel neurotrophic factor related to GDNF and neurturin. Neuron. 1998;20:245–253. doi: 10.1016/s0896-6273(00)80453-5. [DOI] [PubMed] [Google Scholar]
  129. Minichiello L. TrkB signalling pathways in LTP and learning. Nat Rev Neurosci. 2009;10:850–860. doi: 10.1038/nrn2738. [DOI] [PubMed] [Google Scholar]
  130. Miralles F, Philippe P, Czernichow P, et al. Expression of nerve growth factor and its high-affinity receptor Trk-A inthe rat pancreas during embryonic and fetal life. J Endocrinol. 1998;156:431–439. doi: 10.1677/joe.0.1560431. [DOI] [PubMed] [Google Scholar]
  131. Mitsumoto H, Ikeda K, Klinkosz B, et al. Arrest of motor neuron disease in wobbler mice cotreated with CNTF and BDNF. Science. 1994;265:1107–1110. doi: 10.1126/science.8066451. [DOI] [PubMed] [Google Scholar]
  132. Morandi A, Martin LA, Gao Q, et al. GDNF-RET signaling in ER-positive breast cancers is a key determinant of response and resistance to aromatase inhibitors. Cancer Res. 2013;73:3783–3795. doi: 10.1158/0008-5472.CAN-12-4265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Moscatelli I, Pierantozzi E, Camaioni A, et al. p75 neurotrophin receptor is involved in proliferation of undifferentiated mouse embryonic stem cells. Exp Cell Res. 2009;315:3220–3232. doi: 10.1016/j.yexcr.2009.08.014. [DOI] [PubMed] [Google Scholar]
  134. Nguyen N, Lee SB, Lee YS, et al. Neuroprotection by NGF and BDNF against neurotoxin-exerted apoptotic death in neural stem cells are mediated through Trk receptors, activating PI3-kinase and MAPK pathways. Neurochem Res. 2009;34:942–951. doi: 10.1007/s11064-008-9848-9. [DOI] [PubMed] [Google Scholar]
  135. Nilsson E, Dole G, Skinner MK. Neurotrophin NT3 promotes ovarian primordial to primary follicle transition. Reproduction. 2009;138:697–707. doi: 10.1530/REP-09-0179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Oatley JM, Avarbock MR, Brinster RL. Glial cell line-derived neurotrophic factor regulation of genes essential for self-renewal of mouse spermatogonial stem cells is dependent on Src family kinase signaling. J Biol Chem. 2007;282:25842–25851. doi: 10.1074/jbc.M703474200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Oberg-Welsh C, Welsh M. Effects of certain growth factors on in vitro maturation of rat fetal islet-like structures. Pancreas. 1996;12:334–339. doi: 10.1097/00006676-199605000-00002. [DOI] [PubMed] [Google Scholar]
  138. Ohtsuka M, Soumiya H, Hanai M, et al. Neurotrophin-3 influences the number and the laminar fate of cortical progenitors in the developing cerebral cortex of mice through the MEK/ ERK1/2 signaling pathway. Biomed Res. 2013;34:231–239. doi: 10.2220/biomedres.34.231. [DOI] [PubMed] [Google Scholar]
  139. Olson L, Backlund EO, Ebendal T, et al. Intraputaminal infusion of nerve growth factor to support adrenal medullary autografts in Parkinson's disease. One-year follow-up of first clinical trial. Arch Neurol. 1991;48:373–381. doi: 10.1001/archneur.1991.00530160037011. [DOI] [PubMed] [Google Scholar]
  140. Paratcha G, Ledda F, Ibanez CF. The neural cell adhesion molecule NCAM is an alternative signaling receptor for GDNF family ligands. Cell. 2003;113:867–879. doi: 10.1016/s0092-8674(03)00435-5. [DOI] [PubMed] [Google Scholar]
  141. Patterson SL, Abel T, Deuel TA, et al. Recombinant BDNF rescues deficits in basal synaptic transmission and hippocampal LTP in BDNF knockout mice. Neuron. 1996;16:1137–1145. doi: 10.1016/s0896-6273(00)80140-3. [DOI] [PubMed] [Google Scholar]
  142. Petty BG, Cornblath DR, Adornato BT, et al. The effect of systemically administered recombinant human nerve growth factor in healthy human subjects. Ann Neurol. 1994;36:244–246. doi: 10.1002/ana.410360221. [DOI] [PubMed] [Google Scholar]
  143. Pezzoli G, Fahn S, Dwork A, et al. Non-chromaffin tissue plus nerve growth factor reduces experimental parkinsonism in aged rats. Brain Res. 1988;459:398–403. doi: 10.1016/0006-8993(88)90659-2. [DOI] [PubMed] [Google Scholar]
  144. Phillips HS, Hains JM, Armanini M, et al. BDNF mRNA is decreased in the hippocampus of individuals with Alzheimer's disease. Neuron. 1991;7:695–702. doi: 10.1016/0896-6273(91)90273-3. [DOI] [PubMed] [Google Scholar]
  145. Pichel JG, Shen L, Sheng HZ, et al. Defects in enteric innervation and kidney development in mice lacking GDNF. Nature. 1996;382:73–76. doi: 10.1038/382073a0. [DOI] [PubMed] [Google Scholar]
  146. Pineda JR, Rubio N, Akerud P, et al. Neuroprotection by GDNF-secreting stem cells in a Huntington's disease model: optical neuroimage tracking of brain-grafted cells. Gene Ther. 2007;14:118–128. doi: 10.1038/sj.gt.3302847. [DOI] [PubMed] [Google Scholar]
  147. Poteriaev DA, Saarma M. The GDNF family: from neurotrophic factors to oncogenesis. Mol Biol. 2001;35:309–320. [PubMed] [Google Scholar]
  148. Reichardt LF. Neurotrophin-regulated signalling pathways. Philos Trans R Soc Lond B Biol Sci. 2006;361:1545–1564. doi: 10.1098/rstb.2006.1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Richardson RM, Kells AP, Rosenbluth KH, et al. Interventional MRI-guided putaminal delivery of AAV2-GDNF for a planned clinical trial in Parkinson's disease. Mol Ther. 2011;19:1048–1057. doi: 10.1038/mt.2011.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Rivera C, Li H, Thomas-Crusells J, et al. BDNF-induced TrkB activation down-regulates the K+-Cl− cotransporter KCC2 and impairs neuronal Cl− extrusion. J Cell Biol. 2002;159:747–752. doi: 10.1083/jcb.200209011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Roh J, Muelleman T, Tawfik O, et al. Perineural growth in head and neck squamous cell carcinoma:a review. Oral Oncol. 2015;51:16–23. doi: 10.1016/j.oraloncology.2014.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Rosenbaum T, Sanchez-Soto MC, Hiriart M. Nerve growth factor increases insulin secretion and barium current in pancreatic beta-cells. Diabetes. 2001;50:1755–1762. doi: 10.2337/diabetes.50.8.1755. [DOI] [PubMed] [Google Scholar]
  153. Rosser A, Svendsen CN. Stem cells for cell replacement therapy: a therapeutic strategy for HD? Mov Disord. 2014;29:1446–1454. doi: 10.1002/mds.26026. [DOI] [PubMed] [Google Scholar]
  154. Roussa E, Oehlke O, Rahhal B, et al. Transforming growth factor beta cooperates with persephin for dopaminergic phenotype induction. Stem Cells. 2008;26:1683–1694. doi: 10.1634/stemcells.2007-0805. [DOI] [PubMed] [Google Scholar]
  155. Ruberti F, Capsoni S, Comparini A, et al. Phenotypic knockout of nerve growth factor in adult transgenic mice reveals severe deficits in basal forebrain cholinergic neurons, cell death in the spleen, and skeletal muscle dystrophy. J Neurosci. 2000;20:2589–2601. doi: 10.1523/JNEUROSCI.20-07-02589.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Rudin CM, Drilon A, Poirier JT. RET mutations in neuroendocrine tumors: including small-cell lung cancer. J Thoracic Oncol. 2014;9:1240–1242. doi: 10.1097/JTO.0000000000000301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Saarma M. GDNF—a stranger in the TGF-beta superfamily? Eur J Biochem. 2000;267:696968–696971. doi: 10.1046/j.1432-1327.2000.01826.x. [DOI] [PubMed] [Google Scholar]
  158. Sanchez MP, Silos-Santiago I, Frisen J, et al. Renal agenesis and the absence of enteric neurons in mice lacking GDNF. Nature. 1996;382:70–73. doi: 10.1038/382070a0. [DOI] [PubMed] [Google Scholar]
  159. Sanchez-Ramos J, Song S, Cardozo-Pelaez F, et al. Adult bone marrow stromal cells differentiate into neural cells in vitro. Exp Neurol. 2000;164:247–256. doi: 10.1006/exnr.2000.7389. [DOI] [PubMed] [Google Scholar]
  160. Sariola H, Meng X. GDNF-induced seminomatous tumours in mouse–an experimental model for human seminomas? APMIS. 2003;111:192–196. doi: 10.1034/j.1600-0463.2003.11101231.x. discussion 196. [DOI] [PubMed] [Google Scholar]
  161. Sariola H, Saarma M. Novel functions and signalling pathways for GDNF. J Cell Sci. 2003;116(Pt 19):3855–3862. doi: 10.1242/jcs.00786. [DOI] [PubMed] [Google Scholar]
  162. Sato A, Okumura K, Matsumoto S, et al. Isolation, tissue localization, and cellular characterization of progenitors derived from adult human salivary glands. Cloning Stem Cells. 2007;9:191–205. doi: 10.1089/clo.2006.0054. [DOI] [PubMed] [Google Scholar]
  163. Scarisbrick IA, Asakura K, Rodriguez M. Neurotrophin-4/5 promotes proliferation of oligodendrocyte-type-2 astrocytes (O-2A) Brain Res Dev Brain Res. 2000;123:87–90. doi: 10.1016/s0165-3806(00)00077-8. [DOI] [PubMed] [Google Scholar]
  164. Schwerk A, Altschuler J, Roch M, et al. Human adipose-derived mesenchymal stromal cells increase endogenous neurogenesis in the rat subventricular zone acutely after 6-hydroxydopamine lesioning. Cytotherapy. 2015;17:199–214. doi: 10.1016/j.jcyt.2014.09.005. [DOI] [PubMed] [Google Scholar]
  165. Shabtay-Orbach A, Amit M, Binenbaum Y, et al. Paracrine regulation of glioma cells invasion by astrocytes is mediated by glial-derived neurotrophic factor. Int J Cancer. 2014 doi: 10.1002/ijc.29380. [DOI] [PubMed] [Google Scholar]
  166. Shelton DL, Reichardt LF. Studies on the expression of the beta nerve growth factor (NGF) gene in the central nervous system: level and regional distribution of NGF mRNA suggest that NGF functions as a trophic factor for several distinct populations of neurons. Proc Natl Acad Sci USA. 1986;83:2714–2718. doi: 10.1073/pnas.83.8.2714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Shen Y, Inoue N, Heese K. Neurotrophin-4 (ntf4) mediates neurogenesis in mouse embryonic neural stem cells through the inhibition of the signal transducer and activator of transcription-3 (stat3) and the modulation of the activity of protein kinase B. Cell Mol Neurobiol. 2010;30:909–916. doi: 10.1007/s10571-010-9520-1. [DOI] [PubMed] [Google Scholar]
  168. Shmelkov SV, Meeus S, Moussazadeh N, et al. Cytokine preconditioning promotes codifferentiation of human fetal liver CD133+ stem cells into angiomyogenic tissue. Circulation. 2005;111:1175–1183. doi: 10.1161/01.CIR.0000157155.44008.0F. [DOI] [PubMed] [Google Scholar]
  169. Sinkevicius KW, Kriegel C, Bellaria K, et al. Neurotrophin receptor TrkB promotes lung adenocarcinoma metastasis. Proc Natl Acad Sci USA. 2014;111:10299–10304. doi: 10.1073/pnas.1404399111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Skaper SD. The neurotrophin family of neurotrophic factors: an overview. Methods Mol Biol. 2012;846:1–12. doi: 10.1007/978-1-61779-536-7_1. [DOI] [PubMed] [Google Scholar]
  171. Slevin JT, Gerhardt GA, Smith CD, et al. Improvement of bilateral motor functions in patients with Parkinson disease through the unilateral intraputaminal infusion of glial cell line-derived neurotrophic factor. J Neurosurg. 2005;102:216–222. doi: 10.3171/jns.2005.102.2.0216. [DOI] [PubMed] [Google Scholar]
  172. Smeyne RJ, Klein R, Schnapp A, et al. Severe sensory and sympathetic neuropathies in mice carrying a disrupted Trk/NGF receptor gene. Nature. 1994;368:246–249. doi: 10.1038/368246a0. [DOI] [PubMed] [Google Scholar]
  173. Smith DJ, Leil TA, Liu X. Neurotrophin-4 is required for tolerance to morphine in the mouse. Neurosci Lett. 2003;340:103–106. doi: 10.1016/s0304-3940(03)00084-3. [DOI] [PubMed] [Google Scholar]
  174. Sofroniew MV, Howe CL, Mobley WC. Nerve growth factor signaling, neuroprotection, and neural repair. Annu Rev Neurosci. 2001;24:1217–1281. doi: 10.1146/annurev.neuro.24.1.1217. [DOI] [PubMed] [Google Scholar]
  175. Stolp HB. Neuropoietic cytokines in normal brain development and neurodevelopmental disorders. Mol Cell Neurosci. 2013;53:63–68. doi: 10.1016/j.mcn.2012.08.009. [DOI] [PubMed] [Google Scholar]
  176. Tang MJ, Cai Y, Tsai SJ, et al. Ureteric bud outgrowth in response to RET activation is mediated by phosphatidylinositol 3-kinase. Dev Biol. 2002;243:128–136. doi: 10.1006/dbio.2001.0557. [DOI] [PubMed] [Google Scholar]
  177. Tang S, Liao X, Shi B, et al. The effects of controlled release of neurotrophin-3 from PCLA scaffolds on the survival and neuronal differentiation of transplanted neural stem cells in a rat spinal cord injury model. PLoS One. 2014;9:e107517. doi: 10.1371/journal.pone.0107517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Teitelman G, Guz Y, Ivkovic S, et al. Islet injury induces neurotrophin expression in pancreatic cells and reactive gliosis of peri-islet Schwann cells. J Neurobiol. 1998;34:304–318. doi: 10.1002/(sici)1097-4695(199803)34:4<304::aid-neu2>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
  179. Tessarollo L, Vogel KS, Palko ME, et al. Targeted mutation in the neurotrophin-3 gene results in loss of muscle sensory neurons. Proc Natl Acad Sci USA. 1994;91:11844–11848. doi: 10.1073/pnas.91.25.11844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Thomas AM, Seidlits SK, Goodman AG, et al. Sonic hedgehog and neurotrophin-3 increase oligodendrocyte numbers and myelination after spinal cord injury. Integr Biol. 2014;6:694–705. doi: 10.1039/c4ib00009a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Tuszynski MH, Thal L, Pay M, et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med. 2005;11:551–555. doi: 10.1038/nm1239. [DOI] [PubMed] [Google Scholar]
  182. Uranagase A, Katsunuma S, Doi K, et al. BDNF expression in olfactory bulb and epithelium during regeneration of olfactory epithelium. Neurosci Lett. 2012;516:45–49. doi: 10.1016/j.neulet.2012.03.051. [DOI] [PubMed] [Google Scholar]
  183. Usui N, Watanabe K, Ono K, et al. Role of motoneuron-derived neurotrophin 3 in survival and axonal projection of sensory neurons during neural circuit formation. Development. 2012;139:1125–1132. doi: 10.1242/dev.069997. [DOI] [PubMed] [Google Scholar]
  184. Villegas SN, Njaine B, Linden R, et al. Glial-derived neurotrophic factor (GDNF) prevents ethanol (EtOH) induced B92 glial cell death by both PI3 K/AKT and MEK/ERK signaling pathways. Brain Res Bull. 2006;71:116–126. doi: 10.1016/j.brainresbull.2006.08.014. [DOI] [PubMed] [Google Scholar]
  185. Vourc'h P, Lacar B, Mignon L, et al. Effect of neurturin on multipotent cells isolated from the adult skeletal muscle. Biochem Biophys Res Commun. 2005;332:215–223. doi: 10.1016/j.bbrc.2005.04.104. [DOI] [PubMed] [Google Scholar]
  186. Wan G, Gomez-Casati ME, Gigliello AR, et al. Neurotrophin-3 regulates ribbon synapse density in the cochlea and induces synapse regeneration after acoustic trauma. Elife. 2014 doi: 10.7554/eLife.03564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Wang F, Chang G, Geng X. NGF and TERT co-transfected BMSCs improve the restoration of cognitive impairment in vascular dementia rats. PLoS One. 2014a;9:e98774. doi: 10.1371/journal.pone.0098774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Wang K, Demir IE, D'Haese JG, et al. The neurotrophic factor neurturin contributes toward an aggressive cancer cell phenotype, neuropathic pain and neuronal plasticity in pancreatic cancer. Carcinogenesis. 2014b;35:103–113. doi: 10.1093/carcin/bgt312. [DOI] [PubMed] [Google Scholar]
  189. Wang LJ, Zhang RP, Li JD. Transplantation of neurotrophin-3-expressing bone mesenchymal stem cells improves recovery in a rat model of spinal cord injury. Acta Neurochirur. 2014c;156:1409–1418. doi: 10.1007/s00701-014-2089-6. [DOI] [PubMed] [Google Scholar]
  190. Wardle RA, Poo MM. Brain-derived neurotrophic factor modulation of GABAergic synapses by postsynaptic regulation of chloride transport. J Neurosci. 2003;23:8722–8732. doi: 10.1523/JNEUROSCI.23-25-08722.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Wells SA, Jr, Santoro M. Targeting the RET pathway in thyroid cancer. Clin Cancer Res. 2009;15:7119–7123. doi: 10.1158/1078-0432.CCR-08-2742. [DOI] [PubMed] [Google Scholar]
  192. Woolley AG, Sheard PW, Duxson MJ. Neurotrophin-3 null mutant mice display a postnatal motor neuropathy. Eur J Neurosci. 2005;21:2100–2110. doi: 10.1111/j.1460-9568.2005.04052.x. [DOI] [PubMed] [Google Scholar]
  193. Xiao N, Lin Y, Cao H, et al. Neurotrophic factor GDNF promotes survival of salivary stem cells. J Clin Invest. 2014;124:3364–3377. doi: 10.1172/JCI74096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Xie CW, Sayah D, Chen QS, et al. Deficient long-term memory and long-lasting long-term potentiation in mice with a targeted deletion of neurotrophin-4 gene. Proc Natl Acad Sci USA. 2000;97:8116–8121. doi: 10.1073/pnas.140204597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Yamada K, Nabeshima T. Brain-derived neurotrophic factor/ TrkB signaling in memory processes. J Pharmacol Sci. 2003;91:267–270. doi: 10.1254/jphs.91.267. [DOI] [PubMed] [Google Scholar]
  196. Yang J, Yan Y, Xia Y, et al. Neurotrophin 3 transduction augments remyelinating and immunomodulatory capacity of neural stem cells. Mol Ther. 2014;22:440–450. doi: 10.1038/mt.2013.241. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  197. Yin X, Xu H, Jiang Y, et al. The effect of lentivirus-mediated PSPN genetic engineering bone marrow mesenchymal stem cells on Parkinson's disease rat model. PLoS One. 2014;9:e105118. doi: 10.1371/journal.pone.0105118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Yin B, Ma ZY, Zhou Z, et al. The TrkB+ cancer stem cells contribute to post-chemotherapy recurrence of triple-negative breast cancers in an orthotopic mouse model. Oncogene. 2015;34:761–770. doi: 10.1038/onc.2014.8. [DOI] [PubMed] [Google Scholar]
  199. Yuan J, Huang G, Xiao Z, et al. Overexpression of beta-NGF promotes differentiation of bone marrow mesenchymal stem cells into neurons through regulation of AKT and MAPK pathway. Mol Cell Biochem. 2013;383:201–211. doi: 10.1007/s11010-013-1768-6. [DOI] [PubMed] [Google Scholar]
  200. Zhang YQ, He LM, Xing B, et al. Neurotrophin-3 gene-modified Schwann cells promote TrkC gene-modified mesenchymal stem cells to differentiate into neuron-like cells in poly(lactic-acid-co-glycolic acid) multiple-channel conduit. Cells Tissues Organs. 2012;195:313–322. doi: 10.1159/000327724. [DOI] [PubMed] [Google Scholar]
  201. Zhang ZJ, Li YJ, Liu XG, et al. Human umbilical cord blood stem cells and brain-derived neurotrophic factor for optic nerve injury: a biomechanical evaluation. Neural Regen Res. 2015;10:1134–1138. doi: 10.4103/1673-5374.160110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Zhao LX, Zhang J, Cao F, et al. Modification of the brain-derived neurotrophic factor gene: a portal to transform mesenchymal stem cells into advantageous engineering cells for neuroregeneration and neuroprotection. Exp Neurol. 2004;190:396–406. doi: 10.1016/j.expneurol.2004.06.025. [DOI] [PubMed] [Google Scholar]
  203. Zheng G, Zhu Z, Zhu K, et al. Therapeutic effect of adeno-associated virus (AAV)-mediated ADNF-9 expression on cochlea of kanamycin-deafened guinea pigs. Acta Otolaryngol. 2013;133:1022–1029. doi: 10.3109/00016489.2013.799777. [DOI] [PubMed] [Google Scholar]
  204. Zhou FQ, Zhong J, Snider WD. Extracellular crosstalk: when GDNF meets N-CAM. Cell. 2003;113:814–815. doi: 10.1016/s0092-8674(03)00467-7. [DOI] [PubMed] [Google Scholar]
  205. Zhu G, Sun C, Liu W. Effects of neurotrophin-3 on the differentiation of neural stem cells into neurons and oligodendrocytes. Neural Regen Res. 2012;7:1483–1487. doi: 10.3969/j.issn.1673-5374.2012.19.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Zuccato C, Ciammola A, Rigamonti D, et al. Loss of huntingtin-mediated BDNF gene transcription in Huntington's disease. Science. 2001;293:493–498. doi: 10.1126/science.1059581. [DOI] [PubMed] [Google Scholar]
  207. Zuccato C, Tartari M, Crotti A, et al. Huntingtin interacts with REST/NRSF to modulate the transcription of NRSE-controlled neuronal genes. Nat Genet. 2003;35:76–83. doi: 10.1038/ng1219. [DOI] [PubMed] [Google Scholar]
  208. Zuccato C, Marullo M, Conforti P, et al. Systematic assessment of BDNF and its receptor levels in human cortices affected by Huntington's disease. Brain Pathol. 2008;18:225–238. doi: 10.1111/j.1750-3639.2007.00111.x. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES