Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jan 10.
Published in final edited form as: Expert Opin Biol Ther. 2016 May 21;16(8):1025–1033. doi: 10.1080/14712598.2016.1183641

Engineered BDNF producing cells as a potential treatment for neurologic disease

Peter Deng a,b, Johnathon D Anderson a, Abigail S Yu b, Geralyn Annett a, Kyle D Fink a, Jan A Nolta a
PMCID: PMC5762114  NIHMSID: NIHMS932050  PMID: 27159050

Abstract

Introduction

Brain-derived neurotrophic factor (BDNF) has been implicated in wide range of neurological diseases and injury. This neurotrophic factor is vital for neuronal health, survival, and synaptic connectivity. Many therapies focus on the restoration or enhancement of BDNF following injury or disease progression.

Areas covered

The present review will focus on the mechanisms in which BDNF exerts its beneficial functioning, current BDNF therapies, issues and potential solutions for delivery of neurotrophic factors to the central nervous system, and other disease indications that may benefit from overexpression or restoration of BDNF.

Expert opinion

Due to the role of BDNF in neuronal development, maturation, and health, BDNF is implicated in numerous neurological diseases making it a prime therapeutic agent. Numerous studies have shown the therapeutic potential of BDNF in a number of neurodegenerative disease models and in acute CNS injury, however clinical translation has fallen short due to issues in delivering this molecule. The use of MSC as a delivery platform for BDNF holds great promise for clinical advancement of neurotrophic factor restoration. The ease with which MSC can be engineered opens the door to the possibility of using this cell-based delivery system to advance a BDNF therapy to the clinic.

Keywords: BDNF, Mesenchymal Stem Cell, Gene Therapy, Huntington’s disease, Neurodegeneration

1. Brain-derived neurotrophic factor introduction

There is an increasing body of evidence for the importance of neurotrophins (NTF) and their role in developmental and mature neuronal cell health. The first NTF, nerve growth factor (NGF), was discovered in the 1950s by a group of embryologists,[1] and over the past 50 years additional NTFs such as brain-derived neurotrophic factor (BDNF) in pig brain in 1982,[2] neurotrophin 3 (NT3),[3] and neurotrophin 4 (NT4) [4] have been discovered. In the mature brain, NTFs have two primary functions. First, NTFs offer trophic support that enhances neurogenesis and neuroprotection. Second, NTFs are potent modulators of synaptic plasticity, a defining role in learning throughout life. BDNF is of potential interest due to the widespread prevalence of TrkB, BDNF’s preferential receptor in the central nervous system (CNS), and its role in potentiating neuronal cell fate through various canonical signaling cascades [5] that influence survival, neuronal outgrowth,[6,7] and plasticity.[8] Loss of BDNF has been implicated in a number of neurodegenerative diseases,[9] while restoration or addition of exogenous BDNF has shown a physiologic therapeutic effect [1012] (Figure 1).

Figure 1.

Figure 1

BDNF function acts through various canonical signaling pathways once bound to its preferred receptor TrkB to influence survival, neuronal outgrowth, synaptic plasticity, learning, neurogenesis and neuroprotection. Loss of BDNF has been indicated in a number of neurodegenerative diseases, while restoration or addition of exogenous BDNF has shown a physiologic therapeutic effect.

This review will briefly describe the gene expression and signal transduction pathways of NTFs, the role of BDNF in the developing and mature nervous system, and its indication in a number of neurodegenerative diseases. The review will also summarize the preclinical and clinical research on BDNF, challenges with in vivo delivery of BDNF, and application to new disease indications.

1.1. BDNF mechanism

NTFs are a class of growth factors with vital roles in the development of mature neuronal systems. Mature neurotrophin action is mediated by high-affinity tropomyosin receptor kinase (Trk) family of receptors which are signaled by dimerization of two receptor molecules. The Trk receptor family consists of a TrkA, TrkB, and TrkC subtype, and activation is characterized by intracellular autophosphorylation and ensuing secondary signaling cascades following receptor dimerization. NTFs demonstrate preferential binding to specific Trk receptors. NGF preferentially binds to TrkA, BDNF and NT4 to TrkB, and NT3 to TrkC.[13] Interestingly, p75NTR has equally low affinity for mature NTFs but high-affinity toward pro-neutrophins. BDNF is initially synthesized as a precursor protein known as preproBDNF that is cleaved to proBDNF that is further cleaved into mature BDNF by tissue plasminogen activator. When bound to TrkB, mature BDNF induces receptor dimerization, and autophosphorylates tyrosine residues that initiates secondary cascades that regulate neurogenesis, synaptic plasticity, and apoptosis.[14] This is achieved by three major signaling pathways: the phosphatidyl inositol 3-kinase (PI3K)–serine–threonine kinase (AKT), mitogen-activated protein kinase (MAPK) extracellular-related kinase (ERK) pathway, and the phospholipase C y (PLCy)-Cam Kinase (CaMK) pathway. Interestingly, studies have shown that both proBDNF and mature BDNF have varying functions by different intracellular pathways. The binding of proBDNF to p75NTR has been shown to promote long-term depression in rodent hippocampal neurons, in contrast with long-term potentiation by mature BDNF binding to TrkB.[15]

BDNF is translated and released in an activity-dependent manner. The Bdnf gene consists of 11 exons in humans and a single exon coding for the proBDNF protein. Bdnf transcription is controlled by nine different promoters that are active in a developmental, tissue specific, and activity-dependent manner.[16] Transcription of Bdnf is initiated at each of the 5′ noncoding exons that are spliced into the common proBDNF protein-coding 3′ exons. Alternatively, there are variable ATG sequences upstream of the exons I, VII, and VIII in humans, resulting in production of proBDNF with variable amino acid lengths at the N-terminal end of the protein.[16] Bdnf has two alternative polyadenylation sites, resulting in two distinct species of mRNA with either a long or short 3′ UTR. While BDNF mRNA and protein are highly expressed in the hippocampus, cerebral cortex, amygdala, and cerebellum in the mammalian brain,[17] these BDNF transcripts are distributed in a structurally distinct manner as well, with short 3′ UTR transcripts present in the somata of hippocampal neurons, while the long 3′ UTR transcripts are found in the dendrites.[18] Studies in mice with a truncated long 3′ UTR demonstrated deficits in synaptic pruning, enlargement of dendritic spines, and selective impairment of long-term potential of dendrites but not the soma of hippocampal neurons.[19] BDNF is expressed to a lower extent in non-neural tissues such as heart, kidney, lung, and testis.

The multiple promoters present in Bdnf results in context-dependent responses to intracellular processes and extracellular disturbances. Promoter IV is of particular interest in that its expression is dependent on multiple transcriptional regulators: both Ca2+ response elements and Ca2+ sensitive transcription factors such as cyclic AMP-responsive element binding protein (CREB),[20] and methylated CpG binding protein.[21] Membrane depolarization and activation of glutamate receptors such as N-methyl-D-asparate (NMDA) receptors enhance BDNF expression.[22] Ca2+ dependent neural activity induced by NMDA receptors stimulates transcriptional expression of BDNF due to multiple Ca2 +-response elements in exons I and IV of Bdnf.[23] Interestingly, CREB is tightly controlled by multiple signaling pathways triggered by Ca2+ such as the rapidly accelerated fibrosarcoma/MAPK/ERK and the CaMK; important pathways for sensory stimuli and learning. Together, this suggests that BDNF expression is strongly regulated by CREB; however, CREB activation can be due to a myriad of Ca2+ induced pathways.

The proBDNF precursor is a 32-kDa protein generated in the endoplasmic reticulum and then cleaved to create 13-kDa mature BDNF protein via furin protease in the trans-Golgi network (TGN).[24] The pro-region of BDNF binds to the lipid-raft associated sorting receptor carboxypeptidase E (CPE) in the TGN and subsequently BDNF is shuttled into secretory vesicles, relaying the importance of the pro-region as a sorting motif.[25] A mutation (val66met) in the pro-region results in poorer sorting of BDNF into vesicles [26] and results in hippocampal volume loss [27], and altered experience dependent plasticity in motor neurons in human Alzheimer’s disease (AD) patients.[28] BDNF-containing vesicles are transported to secretory sites by motor protein complexes and can be released from the cell soma axon and dendrites in a depolarization-dependent manner in vitro [29] in response to Ca2+ influx as mentioned earlier. Interestingly, mutant Huntingtin, the causative gene in Huntington’s disease (HD), appears to attenuate vesicular transport of BDNF along microtubules in neurons [30] and wild-type Huntingtin acts as a transcription factor to enhance cortical BDNF.[31] Repression element silencing factor-1 (REST), a master regulator of neuronal expression, has been shown to have a strong association with Huntingtin and its normal function disrupted in presence of mutant Huntingtin. REST is normally found within the cytoplasm; however, mutant Huntingtin causes translocation of REST into the nucleus and represses several targets including BDNF.[32,33] This phenomenon is reversed upon silencing of REST.[34]

The proposed mechanism of BDNF action as a neuronal growth factor and synaptic regulator has been explored in several studies. Axonal outgrowth in hippocampal neurons is mediated by stabilization of cAMP/PKA activity following BDNF-induced local elevation of TrkB, driving axon initiation in undifferentiated neurites.[35] Both in vitro and in vivo studies have shown that BDNF promotes axon elongation and branching of sensory neurons [36] as well as increased dendritic outgrowth in BDNF-overexpressing neurons in rat cortical slice cultures.[37] Increased expression of BDNF in transgenic mice demonstrated preganglionic and axonal hypertrophy as well as increased synaptic innervation of sympathetic neurons.[11] Adult mutant mice with absent TrkB signaling during development showed less presynaptic terminals, reduced excitatory number, and altered structural morphology of synapses in the hippocampus.[38] Furthermore, aberrant branching and pruning of thalamic axon terminals was observed in conditional TrkB knockout mice in the thalamus.[39]

Decreased synaptic proteins and vesicle release have been shown in animal models with reduced TrkB signaling [40] whereas BDNF exposure results in increased number of docked synaptic vesicles in cultured hippocampal neurons. [41] BDNF potentiates neuronal circuitry by modulating synaptic efficiency. Exogenous BDNF increases glutamate transmission in rodent brain slices of the hippocampus [42] and visual cortex [43] and favors increased insertion of both NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors in the post-synapse of excitatory neurons,[44] important functions for learning. BDNF is a strong modulator for axonal development and synaptic connectivity in the mammalian brain.

The neurogenic potential of BDNF has been well known due to the intricate relationship between its preferential receptor, TrkB, to cell survival and proliferation pathways. However, BDNF has been implicated in a number of disease phenotypes such as ischemic stroke and epileptic seizure.[45] BDNF and TrkB expression in brain structures that are vulnerable to temporal lobe epilepsy (amygdala, etc.) are elevated following epileptic insult and acute infusion of BDNF into the murine brain appears to induce seizure.[46,47] opposing studies have shown that chronic infusion of BDNF reduces potential of reaching an epileptic state [48] and BDNF conditional knockout mice did not appear to demonstrate significant altered epileptic state.[49] Together, this suggests that while BDNF may have a role in epilepsy, it is not entirely well understood.

1.2. BDNF therapies

Deficits in BDNF have been an observed phenomenon in a number of diseases. Post-mortem studies in AD patients have shown significantly decreased BDNF expression in the hippocampus, temporal, and frontal cortex.[50] Spinocerebellar ataxia type 6 showed reduced BDNF mRNA expression in human cerebellum and sequestration of BDNF to abnormal loci in the neuron.[52] Reduced BDNF mRNA is observed in dopaminergic neurons in Parkinson’s disease (PD) in comparison to normal counterparts and reduced neuronal density was correlated to total BDNF mRNA expression,[51] suggesting that loss of BDNF compromised the well-being of neighboring neurons.

BDNF impairment has been related to a number of neuro-cognitive disorders (reviewed in [56]), particularly in anxiety [57,58] and may contribute to the cognitive and emotional disturbances observed in HD. It is interesting to note that Huntingtin appears to be a regulator of BDNF transport and presence of the mutant protein results in aberrant transport and expression in hippocampal and cortical neurons, respectively.[30] Association of REST with mutant Huntingtin results in reduction of BDNF expression.[32,33] The BDNF protein is necessary for medium spiny neurons (MSNs) survival;[54] however, striatal neurons only produce very low levels of neurotrophic factors and are dependent on BDNF to be produced in the cerebral cortex and transported via corticostriatal tracts to MSNs.[59] While it has been reported in experimental models of neurodegenerative diseases that deep-brain stimulation, specifically in the subthalamic nucleus, can increase BDNF protein and mRNA levels in the striatum, functional recovery was only observed in the presence of the stimulation,[60] thereby limiting the long-term benefits without chronic exposure to deep-brain stimulation.

Interestingly, it was found that patients with symptomatic HD and lower levels of BDNF serum concentrations had significantly worse motor and cognitive performances than individuals with normal BDNF levels.[54,55] It is known that BDNF is very important for survival and differentiation of striatal neurons and lower BDNF may be a reason why these neurons deteriorate in HD.[55] Objective biochemical measures can allow one to monitor the progression of HD, and peripheral BDNF levels may become a potential marker to measure the state of the disease and/or the effectiveness of a given treatment.[55]

There have been considerable efforts in developing BDNF as a therapeutic, given the large amount of evidence for its role as a neuroprotective and neurogenic element and associated deficiencies in a number of diseases. Treatment with exogenous BDNF demonstrated sparing of forebrain cholinergic neurons [61] and reduction in Aβ peptide [10] in rat brains. Early BDNF treatment in transgenic AD mice ameliorated cell loss and increased synaptophysin in the entorhinal cortex, while also demonstrating enhanced hippocampal fear conditioning.[62] This same effect was observed in nonhuman primates.[63] Enhanced striatal innervation and reduced cell loss were observed in a Parkinsonian nonhuman primate model following BDNF infusion.[64] Arrest of hippocampal neurogenesis results in impaired contextual fear conditioning following directed irradiation of the hippocampus,[65,66] suggestive that BDNF treatment results in enhanced neurogenesis and synaptic connection.

Marked sparing of motor neuron damage from acute spinal lesions is observed after administration of BDNF.[6769] Environmental enrichment through increased exercise rescued low BDNF levels in R6 HD mice, reducing weight loss and spared motor symptoms.[70] Interestingly, BDNF has been implicated in some forms for neuropathic pain. Diabetic mice showed synaptic dysfunction and both reduced BDNF mRNA and protein.[71] Gamma-Aminobutyric acid (GABA) is down regulated in the dorsal horn of spinal lesioned mice. This effect is ameliorated by BDNF-induced GABA release following intrathecal injections of NTF, resulting in reduced mechanical hypersensitivity and thermal hyperalgesia in treated animals. [72] Allodynia and hyperalgesia induced by chronic constriction injury of the sciatic nerve in rats were reversed 1-week following adeno-associated virus (AAV)-BDNF injections, suggesting a potential therapy for chronic pain.[73] More recently, cerebellar injections of a herpesviral vector encoding BDNF prevented apoptosis of cerebellar granule cells and ataxic phenotype in an induced Fredreich’s ataxia mouse model.[74]

Despite encouraging preclinical data using BDNF, the ability to deliver NTFs into the brain remains a challenge. NTFs are large, polarized proteins that do not readily cross the blood–brain barrier (BBB), necessitating delivery of NTFs directly into the CNS. BDNF administration in animal models of amyloid lateral sclerosis (ALS) demonstrated robust cell survival in spinal motor neurons and corticospinal neurons.[75] Ensuing clinical trials demonstrated BDNF presence in the cerebral spinal fluid following intrathecal administration of recombinant BDNF. While generally well-tolerated, high dose BDNF groups experienced adverse elements such as sleep disturbance and abnormal behavioral effects that necessitated dose reduction.[76] The large-scale, Phase III BDNF Study Group Trial enrolled 1135 patients and did not demonstrate statistical significance in a survival effect following subcutaneous recombinant BDNF administration.[77] Later, clinical trials were performed in smaller numbers and demonstrated the same lack of statistical significance following intrathecal BDNF infusion. Retrospective analysis suggests NTF administration either subcutaneously or intrathecally are unable to reach degenerating neurons [78,79] or cross the BBB [8082] effectively. It is important to note that the associated adverse events observed during the BDNF Study Group were due to dose and not an innate toxicity from BDNF. A similar phenomenon was seen in mice with AAV-BDNF overexpression at high levels.[83]

2. Delivery of BDNF in the CNS

With the knowledge that BDNF is neuroprotective, researchers have attempted to increase BDNF production from endogenous cells within the striatum via adenoviral injections [84,85] or through AAV vector injections into the striatum [86] prior to quinolinic acid lesions. Both groups observed an increase in the numbers of cells expressing Dopamine- and cAMP-regulated phosphoprotein of 32 kDa (Darpp32; specific to MSNs) relative to lesioned control animals. Interestingly, in the latter group, an AAV was also utilized to increase glial-derived neurotrophic factor release.

New approaches in identifying an optimal delivery method for BDNF have been explored in recent years. Transplantation of neural stem cells in an alpha-synuclein mouse improved performance in cognitive and motor domains following restoration of BDNF expression,[87] mimicking effects seen in AAV-mediated BDNF.[88] Nanoparticle-mediated therapy elevated BDNF in the hippocampus mitigated levels of Aβ and tau proteins while augmenting neuronal cell survival in AD transgenic rats.[89] Overexpression of BDNF using mesenchymal stem cells (MSCs) in HD mouse models have shown amelioration of behavioral deficits in HD mouse models,[90] increased striatal volume and neurogenesis.[12]

Effective delivery of BDNF for neurological disorders remains a major challenge due to its very short half-life, which severely limits the effectiveness of the recombinant protein. Several studies have examined various exogenous delivery methods that may be utilized to translate BDNF-based therapeutics to the clinic. Benraiss et al. used AAV vectors to express BDNF in striatal neurons and demonstrated that AAV delivery of BDNF-induced neurogenesis and promoted a longer lifespan in a murine model of HD. [91] Interestingly, this benefit was potentiated by a factor secreted by mesenchymal stem/stromal cells, noggin.[9294] Despite this success, the use of AAV in the clinic has proven difficult due to host immunogenicity to the virus and limited biodistribution.[95] In comparison, MSCs demonstrate paracrine effects such as local immune modulation and release of beneficial factors such as human growth factor, fibroblast growth factor-2, insulin growth factor 1 and vascular endothelial growth factor (VEGF), BDNF, Dickkopf-related protein 1, b-NGF that favor growth. [96] Furthermore, MSCs home to sites of inflammation, apoptosis, and hypoxia, [97] making it an attractive targeting vehicle for sites of injury.

Our group at the Institute for Regenerative Cures at the University of California, Davis have tested the safety and efficacy of genetically engineered human bone marrow MSCs in transgenic HD mouse models and published the results of our investigative new drug-enabling studies in Molecular Therapy. [12] These cells were prepared in a clinically compliant manner, mimicking cells proposed for a Phase I clinical trial. The YAC128 and R6/2 transgenic HD mouse models were used for behavioral and histological studies following transplantation to measure efficacy of MSC/BDNF as a putative therapy for HD.

Human MSC genetically engineered to produce BDNF reduced the total striatal atrophy observed in HD mice by approximately 50%, as compared to untreated transgenic mice. It was observed that engineered MSC also induced significant neurogenesis, a potential mechanism of recovery following BDNF administration. Engineered MSC also provided significant behavioral recovery in transgenic mouse model of HD. Following transplantation, treated mice displayed significantly less anxiety-like behaviors than untreated transgenic mice. These results, along with the abundance of peer-reviewed articles (see [98]) provide compelling evidence for the proposed use of genetically engineered MSC as a candidate therapy for changing the trajectory of disease progression in patients diagnosed with early-stage HD.

MSCs were chosen as the delivery platform for BDNF since they are known to secrete a variety of neurotrophic and other factors that reduce inflammation, reduce programmed cell death, enhance connections between neurons, and reduce cell toxicity.[98] MSCs have been shown to be readily engineered using viral vectors to robustly deliver growth factors. [99,100] Using gene-modified MSCs as a delivery strategy addresses certain safety concerns involved with the direct use of viral vectors, as MSCs do not permanently engraft into host tissues. In addition, MSCs do not require immunosuppression following allogeneic transplantation, and have a strong, demonstrable safety profile in clinical trials.[96,101] Most importantly, MSCs have been shown to be well-tolerated in phase I/II clinical trials without adverse events in a variety of diseases with no tissue matching. Currently, Atherys, SanBio, and Brain-Storm Therapeutics have all concluded Phase II clinical trials with MSCs with no reported adverse effects in either acute or chronic neurodegenerative insults such as ischemic stroke or amyotrophic lateral sclerosis. Importantly, Brain-Storm Cell Therapeutics concluded a phase I/IIa clinical trial in patients with amyotrophic lateral sclerosis using autologous MSCs induced to express neurotrophic factor (NurOwn) with mild and transient adverse effects reported. Strikingly, treated ALS patients demonstrated slowed disease progression following the conclusion of the phase IIa trial with improvements in breathing and reduced motor decline compared to pretreatment.[102]

MSC/BDNF combine the beneficial effects of BDNF administration to the striata along with the benefits of MSC secreted factor supplementation (e.g. noggin). Unlike direct BDNF delivery via viral vector injection or recombinant protein administration into the brain, MSCs migrate into areas of tissue damage and have been shown to have numerous tissue healing effects (reviewed in [98]). Studies have shown that MSCs do not permanently engraft into host tissues; however, the duration and strategic localization of BDNF production should be adequate to produce a beneficial effect that will outlast the survival of MSCs.[12,103]

2.1. Alternative disease indications for MSC-delivered BDNF

The adult nervous system has limited repair capabilities after injury due to intrinsic neuronal properties which prevent effective reinnervation. However, remaining uninjured axons can develop a few collaterals that reinnervate denervated neurons and this response is increased by NTFs such as BDNF. In addition, severed neurons may be rescued from degenerative atrophy and apoptosis upon treatment with BDNF.

Moreover, increased interest in BDNF as a co-therapeutic for acute spinal injury has been expressed due to its role in synaptic modulation and axonal regeneration. Enhanced BDNF is observed in subpopulations of motor nuclei in spinalized rats that underwent exercise, suggesting BDNF as marker for increased motor innervation.[104] Reduced BDNF is observed in both spared and resected dorsal root ganglia following partial ganglionectomy, but BDNF increased significantly several days following operation in spared ganglia, suggesting a role for BDNF in neuroplasticity following acute injury.[105] Increased BDNF application in spinal lesions in mice demonstrated increased sprouting of injured neurons.[106]

While multiple studies have demonstrated that BDNF can promote axonal regeneration and rewiring of injured nerve fibers, outcomes vary in their therapeutic effect. BDNF treatment may benefit from complementary signaling proteins such as those produced by MSC. After spinal cord injury, pro-apoptotic factors are typically upregulated; however, BDNF may induce pro-survival signaling cascades and rescue neurons that take part in restructuring neuronal circuitry.

Reinnervation of muscles is also critical to re-establishing the functional activity of the muscle–skeletal apparatus after peripheral nerve injuries. Several studies have determined that BDNF is critical to the repair of damaged peripheral nerves, and is upregulated via epigenetic mechanisms for several days post-injury.[107109] Studies have demonstrated that BDNF induced axonal regeneration and axonal sprouting from the proximal end of cut nerves into denervated nerve stumps. [110,111] Taken together, numerous studies have demonstrated that BDNF-related therapies may prove beneficial for spinal cord and peripheral nerve injuries (Figure 2).

Figure 2.

Figure 2

Therapeutic intervention using supplementation or replacement of BDNF has been implicated in a number of neurodegenerative disorders, spinal cord injuries, and nerve reinnervation. The upper left panel displays human MSC genetically engineered to produce BDNF in green (HuNu) following transplantation in the mouse brain with new axonal growth cones shown in red (Neurophilin).

3. Expert opinion

BDNF is a crucial NTF for development and mature neuronal health. It is highly implicated in numerous neurological diseases such as AD, PD, HD and other neurodegenerative disorders, making it a prime therapeutic agent. Countless studies have shown the therapeutic potential of BDNF in promoting axonal regrowth, preserving synaptic strength, sparing neuronal loss in a number of neurodegenerative disease models, and facilitating reinnervation of neurons in acute CNS injury. Despite numerous preclinical studies providing evidence on the therapeutic potential of BDNF, there has been difficulty in translating this work to the clinic. The methods to deliver BDNF to the CNS in a safe and physiologically relevant manner have historically been lacking. The use of recombinant protein is fraught with issues of protein degradation, immune response, and an inability to cross the BBB in appreciable quantities. The use of viral vectors also runs the risk of immune response as well as insertional mutagenesis and the potential of overloading the endogenous neurons with excess BDNF, a concern in cases of epilepsy. As such, researchers must adapt existing technologies and develop new methods to surmount the current challenges in delivering NTFs clinically.

The use of MSC as a delivery platform for NTF, specifically BDNF, holds great promise to advance the clinical use of NTF replacement or supplementation. The ease with which MSC can be engineered opens the door to the possibility of using this cell-based delivery system to advance poly-therapeutics such as trophic support in addition to potent gene editing molecules. Our research team at the Institute for Regenerative Cures at the University of California, Davis has specifically focused on translating promising preclinical data using MSC as a delivery platform for trophic factors including BDNF and VEGF for HD and critical limb ischemia, respectively. We have validated the preclinical efficacy and safety of MSC engineered to produce BDNF in transgenic mouse models of HD. The results from our animal studies indicate that transplantation of genetically engineered MSC are safe and provide significant behavioral and neuropathological benefits in two transgenic mouse models of HD.

We are currently conducting duration, biodistribution, and large animal (porcine) studies as recommended by the FDA for Investigational New Drug approval to potentially start a Phase I safety and tolerability study transplanting genetically engineered MSC in early-stage HD patients (HD-CELL). In anticipation of HD-CELL our group is conducting a Phase I observational trial (PRE-CELL NCT01937923) to longitudinally assess disease progression prior to potentially enrolling patients in a therapeutic trial. The parallel design of the preclinical and clinical observations not only pre-identifies a patient population that would be eligible to receive our MSC/BDNF stem cell product, but allows each subject to essentially serve as their own control when addressing disease progression. We feel that our strong preclinical data, along with the robust evidence in the literature that MSC-based delivery of BDNF or other NTF holds great promise to treat not only HD, but other neurological diseases or damage to the CNS.

Article highlights.

  • Brain-derived neurotrophic factor is a critical neurotrophin regulating cell survival, neuronal outgrowth, and plasticity.

  • Brain-derived neurotrophic factor regulates neurogenesis, synaptic plasticity, and apoptosis via PI3K-AKT, MAPK, ERK, and y-CaM kinase pathway.

  • Dysregulation of brain-derived neurotrophic factor has been implicated in many disorders including Alzheimer’s, Parkinson’s, and Huntington’s disease.

  • Delivery of brain-derived neurotrophic factor has been the major hurdle to its use in a clinical setting.

  • Engineered mesenchymal stem cells may circumvent many of the issues surrounding brain-derived neurotrophic factor delivery.

This box summarizes key points contained in the article.

Acknowledgments

Support for this project was provided by a NIH NIGMS Predoctoral Fellowship T32GM099608 (Deng), NSF GRFP 2011116000, NIH T32-GM008799, NSF GROW 201111600, T32-HL086350 NIH (Anderson), NRSA Postdoctoral Fellowship F32NS090722 (Fink), a NIH Director’s transformative award 1R01GM099688 (Nolta), The Stewart’s and Dake Family Gift (Nolta/Fink), California Institute for Regenerative Medicine (CIRM) DR2-05415 (Wheelock/Nolta), and philanthropic donors from the HD community, including the Roberson family and Team KJ.

Footnotes

Declaration of interests

This paper was supported by funds from the NIH, California Institute for Regenerative Medicine, and unrestricted philanthropic gifts. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

References

Papers of special note have been highlighted as:

• of interest

•• of considerable interest

  • 1.Cohen S, Levi-Montalcini R, Hamburger V. A nerve growth-stimulating factor isolated from SARCOM as 37 and 180. Proc Natl Acad Sci U S A. 1954 Oct;40(10):1014–1018. doi: 10.1073/pnas.40.10.1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Barde YA, Edgar D, Thoenen H. Purification of a new neurotrophic factor from mammalian brain. EMBO J. 1982;1(5):549–553. doi: 10.1002/j.1460-2075.1982.tb01207.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Maisonpierre PC, Belluscio L, Squinto S, et al. Neurotrophin-3: a neurotrophic factor related to NGF and BDNF. Science. 1990 Mar 23;247(4949 Pt 1):1446–1451. doi: 10.1126/science.247.4949.1446. [DOI] [PubMed] [Google Scholar]
  • 4.Berkemeier LR, Winslow JW, Kaplan DR, et al. Neurotrophin-5: a novel neurotrophic factor that activates trk and trkB. Neuron. 1991 Nov;7(5):857–866. doi: 10.1016/0896-6273(91)90287-A. [DOI] [PubMed] [Google Scholar]
  • 5.Kaplan DR, Miller FD. Neurotrophin signal transduction in the nervous system. Curr Opin Neurobiol. 2000 Jun;10(3):381–391. doi: 10.1016/S0959-4388(00)00092-1. [DOI] [PubMed] [Google Scholar]
  • 6.Corbit KC, Foster DA, Rosner MR. Protein kinase Cdelta mediates neurogenic but not mitogenic activation of mitogen-activated protein kinase in neuronal cells. Mol Cell Biol. 1999 Jun;19(6):4209–4218. doi: 10.1128/MCB.19.6.4209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dimitropoulou A, Bixby JL. Regulation of retinal neurite growth by alterations in MAPK/ERK kinase (MEK) activity. Brain Res. 2000 Mar 6;858(1):205–214. doi: 10.1016/s0006-8993(00)01946-6. [DOI] [PubMed] [Google Scholar]
  • 8.Bramham CR, Messaoudi E. BDNF function in adult synaptic plasticity: the synaptic consolidation hypothesis. Prog Neurobiol. 2005 Jun;76(2):99–125. doi: 10.1016/j.pneurobio.2005.06.003. [DOI] [PubMed] [Google Scholar]
  • 9.Lu B, Nagappan G, Guan X, et al. BDNF-based synaptic repair as a disease-modifying strategy for neurodegenerative diseases. Nat Rev Neurosci. 2013 Jun;14(6):401–416. doi: 10.1038/nrn3505. [DOI] [PubMed] [Google Scholar]
  • 10.Arancibia S, Silhol M, Moulière F, et al. Protective effect of BDNF against beta-amyloid induced neurotoxicity in vitro and in vivo in rats. Neurobiol Dis. 2008 Sep;31(3):316–326. doi: 10.1016/j.nbd.2008.05.012. [DOI] [PubMed] [Google Scholar]
  • 11.Causing CG, Gloster A, Aloyz R, et al. Synaptic innervation density is regulated by neuron-derived BDNF. Neuron. 1997 Feb;18(2):257–267. doi: 10.1016/S0896-6273(00)80266-4. [DOI] [PubMed] [Google Scholar]
  • 12••.Pollock K, Dahlenburg H, Nelson H, et al. Human mesenchymal stem cells genetically engineered to overexpress brain-derived neurotrophic factor improve outcomes in Huntington’s disease mouse models. Mol Ther J Am Soc Gene Ther. 2016 Jan 14; doi: 10.1038/mt.2016.12. This reference describes the results of a large preclinical efficacy study using MSC engineered to overexpress BDNF in two transgenic mouse models of Huntington’s disease. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Adachi N, Numakawa T, Richards M, et al. New insight in expression, transport, and secretion of brain-derived neurotrophic factor: implications in brain-related diseases. World J Biol Chem. 2014 Nov 26;5(4):409–428. doi: 10.4331/wjbc.v5.i4.409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Huang EJ, Reichardt LF. Trk receptors: roles in neuronal signal transduction. Annu Rev Biochem. 2003;72:609–642. doi: 10.1146/annurev.biochem.72.121801.161629. [DOI] [PubMed] [Google Scholar]
  • 15.Woo NH, Teng HK, Siao C-J, et al. Activation of p75NTR by proBDNF facilitates hippocampal long-term depression. Nat Neurosci. 2005 Aug;8(8):1069–1077. doi: 10.1038/nn1510. [DOI] [PubMed] [Google Scholar]
  • 16.Pruunsild P, Kazantseva A, Aid T, et al. Dissecting the human BDNF locus: bidirectional transcription, complex splicing, and multiple promoters. Genomics. 2007 Sep;90(3):397–406. doi: 10.1016/j.ygeno.2007.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yan Q, Rosenfeld RD, Matheson CR, et al. Expression of brain-derived neurotrophic factor protein in the adult rat central nervous system. Neuroscience. 1997 May;78(2):431–448. doi: 10.1016/S0306-4522(96)00613-6. [DOI] [PubMed] [Google Scholar]
  • 18.Timmusk T, Palm K, Metsis M, et al. Multiple promoters direct tissue-specific expression of the rat BDNF gene. Neuron. 1993 Mar;10(3):475–489. doi: 10.1016/0896-6273(93)90335-O. [DOI] [PubMed] [Google Scholar]
  • 19.An JJ, Gharami K, Liao G-Y, et al. Distinct role of long 3′ UTR BDNF mRNA in spine morphology and synaptic plasticity in hippocampal neurons. Cell. 2008 Jul 11;134(1):175–187. doi: 10.1016/j.cell.2008.05.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hong EJ, McCord AE, Greenberg ME. A biological function for the neuronal activity-dependent component of Bdnf transcription in the development of cortical inhibition. Neuron. 2008 Nov 26;60( 4):610–624. doi: 10.1016/j.neuron.2008.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chen WG, West AE, Tao X, et al. Upstream stimulatory factors are mediators of Ca2+-responsive transcription in neurons. J Neurosci Off J Soc Neurosci. 2003 Apr 1;23(7):2572–2581. doi: 10.1523/JNEUROSCI.23-07-02572.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zafra F, Lindholm D, Castrén E, et al. Regulation of brain-derived neurotrophic factor and nerve growth factor mRNA in primary cultures of hippocampal neurons and astrocytes. J Neurosci Off J Soc Neurosci. 1992 Dec;12(12):4793–4799. doi: 10.1523/JNEUROSCI.12-12-04793.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zheng F, Zhou X, Moon C, et al. Regulation of brain-derived neurotrophic factor expression in neurons. Int J Physiol Pathophysiol Pharmacol. 2012;4(4):188–200. [PMC free article] [PubMed] [Google Scholar]
  • 24.Seidah NG, Benjannet S, Pareek S, et al. Cellular processing of the nerve growth factor precursor by the mammalian pro-protein convertases. Biochem J. 1996 Mar 15;314(Pt 3):951–960. doi: 10.1042/bj3140951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lee R, Kermani P, Teng KK, et al. Regulation of cell survival by secreted proneurotrophins. Science. 2001 Nov 30;294(5548):1945–1948. doi: 10.1126/science.1065057. [DOI] [PubMed] [Google Scholar]
  • 26.Egan MF, Kojima M, Callicott JH, et al. The BDNF val66met polymorphism affects activity-dependent secretion of BDNF and human memory and hippocampal function. Cell. 2003 Jan 24;112( 2):257–269. doi: 10.1016/s0092-8674(03)00035-7. [DOI] [PubMed] [Google Scholar]
  • 27.Pezawas L, Verchinski BA, Mattay VS, et al. The brain-derived neurotrophic factor val66met polymorphism and variation in human cortical morphology. J Neurosci Off J Soc Neurosci. 2004 Nov 10;24( 45):10099–10102. doi: 10.1523/JNEUROSCI.2680-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Kleim JA, Chan S, Pringle E, et al. BDNF val66met polymorphism is associated with modified experience-dependent plasticity in human motor cortex. Nat Neurosci. 2006 Jun;9(6):735–737. doi: 10.1038/nn1699. [DOI] [PubMed] [Google Scholar]
  • 29.Lessmann V, Brigadski T. Mechanisms, locations, and kinetics of synaptic BDNF secretion: an update. Neurosci Res. 2009 Sep;65( 1):11–22. doi: 10.1016/j.neures.2009.06.004. [DOI] [PubMed] [Google Scholar]
  • 30.Gauthier LR, Charrin BC, Borrell-Pagès M, et al. Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell. 2004 Jul 9;118(1):127–138. doi: 10.1016/j.cell.2004.06.018. [DOI] [PubMed] [Google Scholar]
  • 31•.Cattaneo E, Zuccato C, Tartari M. Normal huntingtin function: an alternative approach to Huntington’s disease. Nat Rev Neurosci. 2005 Dec;6(12):919–930. doi: 10.1038/nrn1806. This article describes the relationship between expression of BDNF and the role of the mutant Huntingtin gene is disrupting normal BDNF functioning. [DOI] [PubMed] [Google Scholar]
  • 32.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 Sep;35(1):76–83. doi: 10.1038/ng1250. [DOI] [PubMed] [Google Scholar]
  • 33.Zuccato C, Belyaev N, Conforti P, et al. Widespread disruption of repressor element-1 silencing transcription factor/neuron-restrictive silencer factor occupancy at its target genes in Huntington’s disease. J Neurosci Off J Soc Neurosci. 2007 Jun 27;27(26):6972–6983. doi: 10.1523/JNEUROSCI.4278-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Soldati C, Bithell A, Johnston C, et al. Dysregulation of REST-regulated coding and non-coding RNAs in a cellular model of Huntington’s disease. J Neurochem. 2013 Feb;124(3):418–430. doi: 10.1111/jnc.12090. [DOI] [PubMed] [Google Scholar]
  • 35.Cheng P-L, Song A-H, Wong Y-H, et al. Self-amplifying autocrine actions of BDNF in axon development. Proc Natl Acad Sci U S A. 2011 Nov 8;108(45):18430–18435. doi: 10.1073/pnas.1115907108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Cohen-Cory S, Fraser SE. Effects of brain-derived neurotrophic factor on optic axon branching and remodelling in vivo. Nature. 1995 Nov 9;378(6553):192–196. doi: 10.1038/378192a0. [DOI] [PubMed] [Google Scholar]
  • 37.Wirth MJ, Brun A, Grabert J, et al. Accelerated dendritic development of rat cortical pyramidal cells and interneurons after biolistic transfection with BDNF and NT4/5. Dev Camb Engl. 2003 Dec;130( 23):5827–5838. doi: 10.1242/dev.00826. [DOI] [PubMed] [Google Scholar]
  • 38.Luikart BW, Nef S, Virmani T, et al. TrkB has a cell-autonomous role in the establishment of hippocampal Schaffer collateral synapses. J Neurosci Off J Soc Neurosci. 2005 Apr 13;25( 15):3774–3786. doi: 10.1523/JNEUROSCI.0041-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lush ME, Ma L, Parada LF. TrkB signaling regulates the developmental maturation of the somatosensory cortex. Int J Dev Neurosci Off J Int Soc Dev Neurosci. 2005 Oct;23(6):523–536. doi: 10.1016/j.ijdevneu.2005.04.003. [DOI] [PubMed] [Google Scholar]
  • 40.Tyler WJ, Pozzo-Miller LD. BDNF enhances quantal neurotransmitter release and increases the number of docked vesicles at the active zones of hippocampal excitatory synapses. J Neurosci Off J Soc Neurosci. 2001 Jun 15;21(12):4249–4258. doi: 10.1523/JNEUROSCI.21-12-04249.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Tyler WJ, Zhang X, Hartman K, et al. BDNF increases release probability and the size of a rapidly recycling vesicle pool within rat hippocampal excitatory synapses. J Physiol. 2006 Aug 1;574(Pt 3):787–803. doi: 10.1113/jphysiol.2006.111310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kang H, Schuman EM. Long-lasting neurotrophin-induced enhancement of synaptic transmission in the adult hippocampus. Science. 1995 Mar 17;267(5204):1658–1662. doi: 10.1126/science.7886457. [DOI] [PubMed] [Google Scholar]
  • 43.Carmignoto G, Pizzorusso T, Tia S, et al. Brain-derived neurotrophic factor and nerve growth factor potentiate excitatory synaptic transmission in the rat visual cortex. J Physiol. 1997 Jan 1;498(Pt 1):153–164. doi: 10.1113/jphysiol.1997.sp021848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Carvalho AL, Caldeira MV, Santos SD, et al. Role of the brain-derived neurotrophic factor at glutamatergic synapses. Br J Pharmacol. 2008 Mar;153( Suppl 1):S310–24. doi: 10.1038/bjp.2008.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Scharfman HE. Brain-derived neurotrophic factor and epilepsy–a missing link? Epilepsy Curr Am Epilepsy Soc. 2005 Jun;5(3):83–88. doi: 10.1111/j.1535-7511.2005.05312.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Humpel C, Wetmore C, Olson L. Regulation of brain-derived neurotrophic factor messenger RNA and protein at the cellular level in pentylenetetrazol-induced epileptic seizures. Neuroscience. 1993 Apr;53(4):909–918. doi: 10.1016/0306-4522(93)90476-V. [DOI] [PubMed] [Google Scholar]
  • 47.Vezzani A, Ravizza T, Moneta D, et al. Brain-derived neurotrophic factor immunoreactivity in the limbic system of rats after acute seizures and during spontaneous convulsions: temporal evolution of changes as compared to neuropeptide Y. Neuroscience. 1999;90( 4):1445–1461. doi: 10.1016/S0306-4522(98)00553-3. [DOI] [PubMed] [Google Scholar]
  • 48.Scharfman HE, Goodman JH, Sollas AL, et al. Spontaneous limbic seizures after intrahippocampal infusion of brain-derived neurotrophic factor. Exp Neurol. 2002 Apr;174(2):201–214. doi: 10.1006/exnr.2002.7869. [DOI] [PubMed] [Google Scholar]
  • 49.He X-P, Kotloski R, Nef S, et al. Conditional deletion of TrkB but not BDNF prevents epileptogenesis in the kindling model. Neuron. 2004 Jul 8;43(1):31–42. doi: 10.1016/j.neuron.2004.06.019. [DOI] [PubMed] [Google Scholar]
  • 50.Siegel GJ, Chauhan NB. Neurotrophic factors in Alzheimer’s and Parkinson’s disease brain. Brain Res Brain Res Rev. 2000 Sep;33(2–3):199–227. doi: 10.1016/S0165-0173(00)00030-8. [DOI] [PubMed] [Google Scholar]
  • 51.Howells DW, Porritt MJ, Wong JY, et al. Reduced BDNF mRNA expression in the Parkinson’s disease substantia nigra. Exp Neurol. 2000 Nov;166(1):127–135. doi: 10.1006/exnr.2000.7483. [DOI] [PubMed] [Google Scholar]
  • 52.Takahashi M, Ishikawa K, Sato N, et al. Reduced brain-derived neurotrophic factor (BDNF) mRNA expression and presence of BDNF-immunoreactive granules in the spinocerebellar ataxia type 6 (SCA6) cerebellum. Neuropathol Off J Jpn Soc Neuropathol. 2012 Dec;32(6):595–603. doi: 10.1111/j.1440-1789.2012.01302.x. [DOI] [PubMed] [Google Scholar]
  • 53.Strand AD, Baquet ZC, Aragaki AK, et al. Expression profiling of Huntington’s disease models suggests that brain-derived neurotrophic factor depletion plays a major role in striatal degeneration. J Neurosci Off J Soc Neurosci. 2007 Oct 24;27(43):11758–11768. doi: 10.1523/JNEUROSCI.2461-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ciammola A, Sassone J, Cannella M, et al. Low brain-derived neurotrophic factor (BDNF) levels in serum of Huntington’s disease patients. Am J Med Genet Part B Neuropsychiatr Genet Off Publ Int Soc Psychiatr Genet. 2007 Jun 5;144B(4):574–577. doi: 10.1002/ajmg.b.30501. [DOI] [PubMed] [Google Scholar]
  • 55.Conforti P, Ramos C, Apostol BL, et al. Blood level of brain-derived neurotrophic factor mRNA is progressively reduced in rodent models of Huntington’s disease: restoration by the neuroprotective compound CEP-1347. Mol Cell Neurosci. 2008 Sep;39(1):1–7. doi: 10.1016/j.mcn.2008.04.012. [DOI] [PubMed] [Google Scholar]
  • 56.Autry AE, Monteggia LM. Brain-derived neurotrophic factor and neuropsychiatric disorders. Pharmacol Rev. 2012 Apr;64(2):238–258. doi: 10.1124/pr.111.004580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bergami M, Rimondini R, Santi S, et al. Deletion of TrkB in adult progenitors alters newborn neuron integration into hippocampal circuits and increases anxiety-like behavior. Proc Natl Acad Sci U S A. 2008 Oct 7;105(40):15570–15575. doi: 10.1073/pnas.0803702105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chen Z-Y, Jing D, Bath KG, et al. Genetic variant BDNF (Val66Met) polymorphism alters anxiety-related behavior. Science. 2006 Oct 6;314(5796):140–143. doi: 10.1126/science.1129663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Zuccato C, Marullo M, Vitali B, et al. Brain-derived neurotrophic factor in patients with Huntington’s disease. PloS One. 2011;6(8):e22966. doi: 10.1371/journal.pone.0022966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Spieles-Engemann AL, Steece-Collier K, Behbehani MM, et al. Subthalamic nucleus stimulation increases brain derived neurotrophic factor in the Nigrostriatal system and primary motor cortex. J Park Dis. 2011 May 31;1(1):123–136. [PMC free article] [PubMed] [Google Scholar]
  • 61.Knüsel B, Beck KD, Winslow JW, et al. Brain-derived neurotrophic factor administration protects basal forebrain cholinergic but not nigral dopaminergic neurons from degenerative changes after axotomy in the adult rat brain. J Neurosci Off J Soc Neurosci. 1992 Nov;12(11):4391–4402. doi: 10.1523/JNEUROSCI.12-11-04391.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Nagahara AH, Mateling M, Kovacs I, et al. Early BDNF treatment ameliorates cell loss in the entorhinal cortex of APP transgenic mice. J Neurosci Off J Soc Neurosci. 2013 Sep 25;33(39):15596–15602. doi: 10.1523/JNEUROSCI.5195-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Nagahara AH, Merrill DA, Coppola G, et al. Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat Med. 2009 Mar;15(3):331–337. doi: 10.1038/nm.1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Tsukahara T, Takeda M, Shimohama S, et al. Effects of brain-derived neurotrophic factor on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinsonism in monkeys. Neurosurgery. 1995 Oct;37(4):733–9. doi: 10.1227/00006123-199510000-00018. discussion 739–41. [DOI] [PubMed] [Google Scholar]
  • 65.Drew MR, Denny CA, Hen R. Arrest of adult hippocampal neurogenesis in mice impairs single- but not multiple-trial contextual fear conditioning. Behav Neurosci. 2010 Aug;124(4):446–454. doi: 10.1037/a0020081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Saxe MD, Battaglia F, Wang J-W, et al. Ablation of hippocampal neurogenesis impairs contextual fear conditioning and synaptic plasticity in the dentate gyrus. Proc Natl Acad Sci U S A. 2006 Nov 14;103(46):17501–17506. doi: 10.1073/pnas.0607207103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chai H, Wu W, So K-F, et al. Long-term effects of a single dose of brain-derived neurotrophic factor on motoneuron survival following spinal root avulsion in the adult rat. Neurosci Lett. 1999 Oct 29;274(3):147–150. doi: 10.1016/s0304-3940(99)00671-0. [DOI] [PubMed] [Google Scholar]
  • 68.Chiu AY, Chen EW, Loera S. Distinct neurotrophic responses of axotomized motor neurons to BDNF and CNTF in adult rats. Neuroreport. 1994 Feb 24;5(6):693–696. doi: 10.1097/00001756-199402000-00008. [DOI] [PubMed] [Google Scholar]
  • 69.Yan Q, Matheson C, Lopez OT, et al. The biological responses of axotomized adult motoneurons to brain-derived neurotrophic factor. J Neurosci Off J Soc Neurosci. 1994 Sep;14(9):5281–5291. doi: 10.1523/JNEUROSCI.14-09-05281.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Spires TL, Grote HE, Varshney NK, et al. Environmental enrichment rescues protein deficits in a mouse model of Huntington’s disease, indicating a possible disease mechanism. J Neurosci Off J Soc Neurosci. 2004 Mar 3;24(9):2270–2276. doi: 10.1523/JNEUROSCI.1658-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Nitta A, Murai R, Suzuki N, et al. Diabetic neuropathies in brain are induced by deficiency of BDNF. Neurotoxicol Teratol. 2002 Oct;24( 5):695–701. doi: 10.1016/S0892-0362(02)00220-9. [DOI] [PubMed] [Google Scholar]
  • 72.Lever I, Cunningham J, Grist J, et al. Release of BDNF and GABA in the dorsal horn of neuropathic rats. Eur J Neurosci. 2003 Sep;18( 5):1169–1174. doi: 10.1046/j.1460-9568.2003.02848.x. [DOI] [PubMed] [Google Scholar]
  • 73.Eaton MJ, Blits B, Ruitenberg MJ, et al. Amelioration of chronic neuropathic pain after partial nerve injury by adeno-associated viral (AAV) vector-mediated over-expression of BDNF in the rat spinal cord. Gene Ther. 2002 Oct;9(20):1387–1395. doi: 10.1038/sj.gt.3301814. [DOI] [PubMed] [Google Scholar]
  • 74.Katsu-Jiménez Y, Loria F, Corona JC, et al. Gene transfer of brain derived neurotrophic factor (BDNF) prevents neurodegeneration triggered by frataxin deficiency. Mol Ther J Am Soc Gene Ther. 2016 Feb 5; doi: 10.1038/mt.2016.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tuszynski MH, Mafong E, Meyer S. Central infusions of brain-derived neurotrophic factor and neurotrophin-4/5, but not nerve growth factor and neurotrophin-3, prevent loss of the cholinergic phenotype in injured adult motor neurons. Neuroscience. 1996 Apr;71(3):761–771. doi: 10.1016/0306-4522(95)00440-8. [DOI] [PubMed] [Google Scholar]
  • 76.Ochs G, Penn RD, York M, et al. A phase I/II trial of recombinant methionyl human brain derived neurotrophic factor administered by intrathecal infusion to patients with amyotrophic lateral sclerosis. Amyotroph Lateral Scler Mot Neuron Disord Off Publ World Fed Neurol Res Group Mot Neuron Dis. 2000 Jun;1(3):201–206. doi: 10.1080/14660820050515197. [DOI] [PubMed] [Google Scholar]
  • 77.The BDNF Study Group. A controlled trial of recombinant methionyl human BDNF in ALS: the BDNF study group (Phase III) Neurology. 1999 Apr 22;52(7):1427–1433. doi: 10.1212/wnl.52.7.1427. [DOI] [PubMed] [Google Scholar]
  • 78.Ankeny DP, McTigue DM, Guan Z, et al. Pegylated brain-derived neurotrophic factor shows improved distribution into the spinal cord and stimulates locomotor activity and morphological changes after injury. Exp Neurol. 2001 Jul;170(1):85–100. doi: 10.1006/exnr.2001.7699. [DOI] [PubMed] [Google Scholar]
  • 79.Pardridge WM, Wu D, Sakane T. Combined use of carboxyl-directed protein pegylation and vector-mediated blood-brain barrier drug delivery system optimizes brain uptake of brain-derived neurotrophic factor following intravenous administration. Pharm Res. 1998 Apr;15(4):576–582. doi: 10.1023/A:1011981927620. [DOI] [PubMed] [Google Scholar]
  • 80.Pan W, Banks WA, Fasold MB, et al. Transport of brain-derived neurotrophic factor across the blood-brain barrier. Neuropharmacology. 1998 Dec;37(12):1553–1561. doi: 10.1016/S0028-3908(98)00141-5. [DOI] [PubMed] [Google Scholar]
  • 81.Pardridge WM, Kang YS, Buciak JL. Transport of human recombinant brain-derived neurotrophic factor (BDNF) through the rat blood-brain barrier in vivo using vector-mediated peptide drug delivery. Pharm Res. 1994 May;11(5):738–746. doi: 10.1023/a:1018940732550. [DOI] [PubMed] [Google Scholar]
  • 82.Poduslo JF, Curran GL. Permeability at the blood-brain and blood-nerve barriers of the neurotrophic factors: NGF, CNTF, NT-3, BDNF. Brain Res Mol Brain Res. 1996 Mar;36(2):280–286. doi: 10.1016/0169-328x(95)00250-v. [DOI] [PubMed] [Google Scholar]
  • 83.Rodriguez-Lebron E, Denovan-Wright EM, Nash K, et al. Intrastriatal rAAV-mediated delivery of anti-huntingtin shRNAs induces partial reversal of disease progression in R6/1 Huntington’s disease transgenic mice. Mol Ther J Am Soc Gene Ther. 2005 Oct;12(4):618–633. doi: 10.1016/j.ymthe.2005.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Arregui L, Benítez JA, Razgado LF, et al. Adenoviral astrocyte-specific expression of BDNF in the striata of mice transgenic for Huntington’s disease delays the onset of the motor phenotype. Cell Mol Neurobiol. 2011 Nov;31(8):1229–1243. doi: 10.1007/s10571-011-9725-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Bemelmans A-P, Horellou P, Pradier L, et al. Brain-derived neurotrophic factor-mediated protection of striatal neurons in an excitotoxic rat model of Huntington’s disease, as demonstrated by adenoviral gene transfer. Hum Gene Ther. 1999 Dec 10;10( 18):2987–2997. doi: 10.1089/10430349950016393. [DOI] [PubMed] [Google Scholar]
  • 86.Kells AP, Henry RA, Connor B. AAV-BDNF mediated attenuation of quinolinic acid-induced neuropathology and motor function impairment. Gene Ther. 2008 Jul;15(13):966–977. doi: 10.1038/gt.2008.23. [DOI] [PubMed] [Google Scholar]
  • 87.Goldberg NS, Caesar J, Park A, et al. Neural stem cells rescue cognitive and motor dysfunction in a transgenic model of dementia with Lewy bodies through a BDNF-dependent mechanism. Stem Cell Rep. 2015 Nov 10;5(5):791–804. doi: 10.1016/j.stemcr.2015.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Benraiss A, Toner MJ, Xu Q, et al. Sustained mobilization of endogenous neural progenitors delays disease progression in a transgenic model of Huntington’s disease. Cell Stem Cell. 2013 Jun 6;12( 6):787–799. doi: 10.1016/j.stem.2013.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Alawdi SH, El-Denshary ES, Safar MM, et al. Neuroprotective effect of nanodiamond in Alzheimer’s disease rat model: a pivotal role for modulating NF-κB and STAT3 signaling. Mol Neurobiol. 2016 Feb;20:1–13. doi: 10.1007/s12035-016-9762-0. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 90.Dey ND, Bombard MC, Roland BP, et al. Genetically engineered mesenchymal stem cells reduce behavioral deficits in the YAC 128 mouse model of Huntington’s disease. Behav Brain Res. 2010 Dec 25;214(2):193–200. doi: 10.1016/j.bbr.2010.05.023. [DOI] [PubMed] [Google Scholar]
  • 91.Benraiss A, Bruel-Jungerman E, Lu G, et al. Sustained induction of neuronal addition to the adult rat neostriatum by AAV4-delivered noggin and BDNF. Gene Ther. 2012 May;19(5):483–493. doi: 10.1038/gt.2011.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Lynch G, Kramar EA, Rex CS, et al. Brain-derived neurotrophic factor restores synaptic plasticity in a knock-in mouse model of Huntington’s disease. J Neurosci Off J Soc Neurosci. 2007 Apr 18;27(16):4424–4434. doi: 10.1523/JNEUROSCI.5113-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Martínez-Serrano A, Björklund A. Protection of the neostriatum against excitotoxic damage by neurotrophin-producing, genetically modified neural stem cells. J Neurosci Off J Soc Neurosci. 1996 Aug 1;16(15):4604–4616. doi: 10.1523/JNEUROSCI.16-15-04604.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pérez-Navarro E, Canudas AM, Akerund P, et al. Brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5 prevent the death of striatal projection neurons in a rodent model of Huntington’s disease. J Neurochem. 2000 Nov;75(5):2190–2199. doi: 10.1046/j.1471-4159.2000.0752190.x. [DOI] [PubMed] [Google Scholar]
  • 95••.Kordower JH. AAV2-neurturin for Parkinson’s disease: what lessons have we learned? Methods Mol Biol Clifton NJ. 2016;1382:485–490. doi: 10.1007/978-1-4939-3271-9_32. This article highlights many of the challenges with delivering trophic factor support in a clinical trial. [DOI] [PubMed] [Google Scholar]
  • 96.Joyce N, Annett G, Wirthlin L, et al. Mesenchymal stem cells for the treatment of neurodegenerative disease. Regen Med. 2010 Nov;5( 6):933–946. doi: 10.2217/rme.10.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kidd S, Spaeth E, Dembinski JL, et al. Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem Cells Dayt Ohio. 2009 Oct;27(10):2614–2623. doi: 10.1002/stem.187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98•.Fink KD, Deng P, Torrest A, et al. Developing stem cell therapies for juvenile and adult-onset Huntington’s disease. Regen Med. 2015;10( 5):623–646. doi: 10.2217/rme.15.25. This review covers many of the preclinical studies using mesenchymal stem cell transplantation for in transgenic Huntington’s disease mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Meyerrose T, Olson S, Pontow S, et al. Mesenchymal stem cells for the sustained in vivo delivery of bioactive factors. Adv Drug Deliv Rev. 2010 Sep 30;62(12):1167–1174. doi: 10.1016/j.addr.2010.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Meyerrose TE, Roberts M, Ohlemiller KK, et al. Lentiviral-transduced human mesenchymal stem cells persistently express therapeutic levels of enzyme in a xenotransplantation model of human disease. Stem Cells Dayt Ohio. 2008 Jul;26(7):1713–1722. doi: 10.1634/stem-cells.2008-0008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Bauer G, Dao MA, Case SS, et al. In vivo biosafety model to assess the risk of adverse events from retroviral and lentiviral vectors. Mol Ther J Am Soc Gene Ther. 2008 Jul;16(7):1308–1315. doi: 10.1038/mt.2008.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Petrou P, Gothelf Y, Argov Z, et al. Safety and clinical effects of Mesenchymal stem cells secreting neurotrophic factor transplantation in patients with amyotrophic lateral sclerosis: results of phase 1/2 and 2a clinical trials. JAMA Neurol. 2016;73(3):337–344. doi: 10.1001/jamaneurol.2015.4321. [DOI] [PubMed] [Google Scholar]
  • 103.Zhao C-P, Zhang C, Zhou S-N, et al. Human mesenchymal stromal cells ameliorate the phenotype of SOD1-G93A ALS mice. Cytotherapy. 2007;9(5):414–426. doi: 10.1080/14653240701376413. [DOI] [PubMed] [Google Scholar]
  • 104.Macias M, Nowicka D, Czupryn A, et al. Exercise-induced motor improvement after complete spinal cord transection and its relation to expression of brain-derived neurotrophic factor and pre-synaptic markers. BMC Neurosci. 2009;10:144. doi: 10.1186/1471-2202-10-144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Rong R, Meng B-L, Jiang N, et al. Roles of BDNF in spinal neuro-plasticity in cats subjected to partial dorsal ganglionectomy. Growth Factors Chur Switz. 2011 Dec;29(6):263–270. doi: 10.3109/08977194.2011.606786. [DOI] [PubMed] [Google Scholar]
  • 106.Vavrek R, Girgis J, Tetzlaff W, et al. BDNF promotes connections of corticospinal neurons onto spared descending interneurons in spinal cord injured rats. Brain J Neurol. 2006 Jun;129(Pt 6):1534–1545. doi: 10.1093/brain/awl087. [DOI] [PubMed] [Google Scholar]
  • 107.Navarro X, Vivó M, Valero-Cabré A. Neural plasticity after peripheral nerve injury and regeneration. Prog Neurobiol. 2007 Jul;82(4):163–201. doi: 10.1016/j.pneurobio.2007.06.005. [DOI] [PubMed] [Google Scholar]
  • 108.Terenghi G. Peripheral nerve regeneration and neurotrophic factors. J Anat. 1999 Jan;194(Pt 1):1–14. doi: 10.1046/j.1469-7580.1999.19410001.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Uchida H, Matsushita Y, Ueda H. Epigenetic regulation of BDNF expression in the primary sensory neurons after peripheral nerve injury: implications in the development of neuropathic pain. Neuroscience. 2013 Jun 14;240:147–154. doi: 10.1016/j.neuroscience.2013.02.053. [DOI] [PubMed] [Google Scholar]
  • 110.Boyd JG, Gordon T. Glial cell line-derived neurotrophic factor and brain-derived neurotrophic factor sustain the axonal regeneration of chronically axotomized motoneurons in vivo. Exp Neurol. 2003 Oct;183(2):610–619. doi: 10.1016/S0014-4886(03)00183-3. [DOI] [PubMed] [Google Scholar]
  • 111.Takemura Y, Imai S, Kojima H, et al. Brain-derived neurotrophic factor from bone marrow-derived cells promotes post-injury repair of peripheral nerve. PloS One. 2012;7(9):e44592. doi: 10.1371/jour-nal.pone.0044592. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES