Role of brain-derived neurotrophic factor in Huntington's disease

https://doi.org/10.1016/j.pneurobio.2007.01.003Get rights and content

Abstract

Neurotrophic factors are essential contributors to the survival of peripheral and central nervous system (CNS) neurons, and demonstration of their reduced availability in diseased brains indicates that they play a role in various neurological disorders. This paper will concentrate on the role of brain-derived neurotrophic factor (BDNF) in the survival and activity of the neurons that die in Huntington's disease (HD) by reviewing the evidence indicating that it involves profound changes in BDNF levels and that attempts to restore these levels are therapeutically interesting.

BDNF is a small dimeric protein that is widely expressed in adult mammalian brain and has been shown to promote the survival of all major neuronal types affected in Alzheimer's disease (AD) and Parkinson's disease (PD). Furthermore, cortical BDNF production is required for the correct activity of the corticostriatal synapse and the survival of the GABA-ergic medium-sized spiny striatal neurons that die in HD. We will highlight the available data concerning changes in BDNF levels in HD cells, mice and human postmortem samples, describe the molecular evidence underlying this alteration, and review the data concerning the impact of the experimental manipulation of BDNF levels on HD progression. Such studies have revealed a major loss of BDNF protein in the striatum of HD patients which may contribute to the clinical manifestations of the disease. They have also opened up a molecular window into the underlying pathogenic mechanism and new therapeutic perspectives by raising the possibility that one of the mechanisms triggering the reduction in BDNF in HD may also affect the activity of many other neuronal proteins.

Introduction

Huntington's disease (HD) is a fatal, dominantly inherited, neurodegenerative disorder that usually onsets in midlife, and is characterised by psychiatric, cognitive and motor dysfunctions. It is due to an excessive repetition of the CAG trinucleotide in exon 1 of the huntingtin gene (Huntington's Disease Collaborative Research Group, 1993) which causes the production of a protein bearing a polyglutamine expanded tract in its N-terminus. The huntingtin mutation leads to widespread brain neurodegeneration, with cell loss mainly in the striatum and cerebral cortex (Reiner et al., 1988), although neuronal abnormalities are also found in many other brain regions (Rosas et al., 2003).

Results from various laboratories suggest that brain-derived neurotrophic factor (BDNF) is involved in the development of the human disease. BDNF was discovered in 1982 (Barde et al., 1982), as the second in a family of molecules with neurotrophic activities whose first identified member was nerve growth factor (NGF) (Levi-Montalcini and Hamburger, 1951). Although widely expressed in the adult mammalian central nervous system (CNS), BDNF is particularly abundant in the hippocampus and cerebral cortex where it is anterogradely transported to its striatal targets via the corticostriatal afferents (Hofer et al., 1990, Altar et al., 1997, Baquet et al., 2004).

Striatal neurons in the brain require BDNF for their activity and survival. Approximately 95% of striatal BDNF is actually of cortical origin, with the rest being produced in the substantia nigra and delivered to the striatum (Altar et al., 1997, Baquet et al., 2004). BDNF expression increases during brain development, peaks after birth (Maisonpierre et al., 1990, Friedman et al., 1991, Huntley et al., 1992 Friedman et al., 1998; Schecterson and Bothwell, 1992; Kolbeck et al., 1999, Baquet et al., 2004), and does not seem to decline with age, thus suggesting that it plays an essential role in the adult CNS (Lapchak et al., 1993; Narisawa-Saito and Nawa, 1996; Katoh-Semba et al., 1997, Katoh-Semba et al., 1998).

BDNF protein is transduced from a single, highly complex gene that carries a number of alternatively used promoters to generate a developmental, tissue-specific and stimulus-induced pattern of BDNF expression (Timmusk et al., 1993, Timmusk et al., 1994, Timmusk et al., 1995, Aid et al., 2007, Liu et al., 2006). The activity of this large regulatory region becomes progressively more complex and presumably more specialised in humans (Liu et al., 2005).

The fact that BDNF has survival promoting activity on the striatal neurons that die in HD has led to the idea that reduced endogenous trophic support may contribute to disease onset and/or progression. This hypothesis has aroused interest in BDNF and/or BDNF mimetics as potential therapeutic agents, and this has been intensified by reports of reduced BDNF levels in the cerebral cortex and striatum of people with HD (Zuccato et al., 2001), as well as in many mouse and cell models of the disease (see Cattaneo et al., 2005 for a review). Furthermore, although no underlying molecular mechanism has been proposed to explain reduced neurotrophic support in other neurological diseases such a Parkinson's disease (PD) or Alzheimer's disease (AD), it is known that the huntingtin mutation in HD reduces the transcriptional activity of the BDNF promoters, thus reducing the transcription of the BDNF gene and decreasing protein production in the cerebral cortex (Zuccato et al., 2001, Zuccato et al., 2003, Zuccato et al., 2005a). Finally, a selective defect in the transport of BDNF protein from cortex to striatum has also been proposed (Gauthier et al., 2004).

The impact of BDNF depletion in HD mice has been tested in experiments in which the levels of endogenous BDNF have been further reduced experimentally, with the consequence of lowering the age of onset and exacerbating the motor dysfunction. The observed neuropathology correlates with morphological alterations in the brain, a finding that confirms the pivotal role of BDNF in the specific degeneration of striatal neurons (Canals et al., 2004). Early experiments in which BDNF was administered in mouse models of HD indicate that BDNF may be beneficial to striatal neurons, although delivery strategies and BDNF bioavailability need to be further improved. More recently, it has been found that administering BDNF to HD mice by means of an osmotic minipump increases the number of striatal enkephalinergic neurons, the most affected population in HD (Canals et al., 2004), thus confirming the possibility that delivering BDNF or increasing endogenous BDNF production may stop or delay the progression of the human disease.

This review discusses the evidence indicating that cortical BDNF is a critical survival factor for the striatal neurons that die in HD. It describes the available data showing the negative influence of BDNF gene inactivation on the performance of striatal neurons in control mice, considers the molecular relationship between huntingtin and BDNF, describes the mechanism by which normal but not mutant huntingtin promotes BDNF production and axonal transport, and discusses the evidence indicating a worse HD phenotype in animal models lacking one BDNF allele. We present the case that the discovery of the mechanisms by which wild-type huntingtin controls the transcription of the BDNF and other neuronal genes may be exploited in drug-screening strategies aimed at identifying compounds capable of acting as huntingtin mimetics. Finally, we summarise attempts to deliver BDNF in HD animal models and the strategies available for increasing its level in human HD.

Section snippets

BDNF, a member of the neurotrophin family

BDNF is an abundant member of the NGF family in mammalian brain. By binding specific receptors such as tyrosine receptor kinase B (TrkB) and p75 neurotrophin receptor (p75NTR), BNDF acts in a paracrine and autocrine manner to control a variety of brain processes, including the growth, development, differentiation and maintenance of neuronal systems, neuronal plasticity, synaptic activity and neurotransmitter-mediated activities (see Chao, 2003, and Binder and Scharfman, 2004, for reviews).

Like

Production of BDNF is stimulated by wild-type huntingtin: physiology and mechanism

We will here review the evidence linking BDNF gene transcription to wild-type huntingtin, the protein whose mutation causes HD, as well as the data demonstrating that a well-known DNA regulatory sequence located within the BDNF promoter represents the first identified downstream molecular target of wild-type huntingtin activity. We will also review the mechanism by which wild-type huntingtin facilitates BDNF gene transcription and summarise the evidence showing that the same mechanism underlies

BDNF is reduced in HD models

A large number of studies have found reduced BDNF levels in HD, which implies that this may put the target striatal neurons and possibly the corticostriatal circuitry at risk (Table 1). This hypothesis is supported by the fact that the neuronal degeneration in human HD mainly affects the striatum and cerebral cortex (Reiner et al., 1988, Rosas et al., 2003), and cortical atrophy may be present even before the onset of symptoms (Rosas et al., 2005). Reduced BDNF levels have also been observed in

BDNF vesicle transport is enhanced by wild-type huntingtin

Huntingtin is predominantly found in the cytoplasm of neurons and is enriched in compartments containing vesicle-associated proteins (DiFiglia et al., 1995). Moreover, wild-type huntingtin is antero- and retrogradely transported in rat sciatic nerve axons, which is consistent with a vesicle association (Block-Galarza et al., 1997). Importantly, huntingtin interacts with the scaffolding proteins involved in axonal transport, such as huntingtin-associated protein 1 (HAP1), and with the dynactin

Evidence of reduced BDNF transport in HD

To establish whether the presence of a polyglutamine expansion in huntingtin affects BDNF transport in HD, BDNF vesicle velocity was measured in heterozygous and homozygous mutant huntingtin knock-in cells in which 109 CAG repeats are inserted in the endogenous huntingtin locus, thus creating a situation similar to that found in HD patients. Further biochemical studies of mutant huntingtin knock-in cells, mice and HD postmortem tissues indicated that the complex driving BDNF vesicles is altered

BDNF levels in HD patients

As described above, analyses of BDNF levels in the striatum of HD mice have generated two different nonmutually exclusive data sets, indicating the possibility that reduced BDNF in striatum is the result of decreased BDNF production in the cortex and decreased transport of this neurotrophin from the cortex to striatum. The first envisions wild-type huntingtin participating in controlling the transcription of the BDNF gene in cerebral cortex by acting at the level of promoter II, thus allowing

BDNF depletion enhances neuropathology and disease onset in HD mice

Previous data indicate that the deficit in striatal BDNF in HD may be due to reduced BDNF gene transcription in the cerebral cortex or reduced BDNF vesicle transport (or both). As described in Section 2.3, the downregulation of endogenous cortical BDNF highlights a link between cortical BDNF levels and specific aspects of HD attributable to decreased BDNF levels (Baquet et al., 2004). To investigate whether the downregulation of endogenous BDNF can influence disease onset and/or progression,

TrkB levels in HD

In order to exert its biological activity, BDNF binds to TrkB receptors, and a number of studies have indicated that TrkB is modulated after neuronal damage: for example, TrkB mRNA is increased in cortical and striatal neurons after excitotoxic lesions (Canals et al., 1999, Checa et al., 2001), and reduced TrkB levels have been associated with neurodegeneration in AD (Connor et al., 1996, Connor et al., 1997, Allen et al., 1999, Ferrer et al., 1999, Soontornniyomkij et al., 1999, Savaskan et

A role for BDNF polymorphisms?

As stated in Section 5.1, a valine-to-methionine substitution at position 66 (Val66Met BDNF) is a known polymorphism of the human BDNF gene that is located in the 5′ pro-BDNF sequence encoding the precursor peptide (pro-BDNF), which is proteolytically cleaved to form the mature protein. This BDNF polymorphism does not affect mature BDNF protein function, but it has recently been shown that it dramatically alters the intracellular trafficking and packaging of pro-BDNF, and consequently the

BDNF: a therapeutic path for Huntington's disease?

A number of studies have shown that BDNF is a potent neurotrophic factor for GABA-ergic striatal neurons. When administered to cultured striatal neurons, it increases their cellular GABA content, the frequency of parvalbumin and calbindin neurons, and the levels of neuropeptide Y and somatostatin (Mizuno et al., 1994; Ventimiglia et al., 1995). In addition, a study by Nakao et al. (1995) showed that BDNF promoted the survival of DARPP-32 positive neurons and, as DARPP-32 is a specific marker of

Conclusion

The findings described in this paper indicate that BDNF is essential for maintaining the corticostriatal pathway, and the cortical BDNF is required for the survival and differentiation of striatal neurons under both physiological and pathological conditions. More importantly, they show that the normal huntingtin protein, whose mutation causes HD, is involved in the physiological control of BDNF synthesis and transport in the brain: it sustains cortical BDNF gene transcription and drives BDNF

Acknowledgements

The authors thanks Drs. Paola Conforti, Manuela Marullo and Dorotea Rigamonti for their comments during the preparation of the manuscript. The work of the authors described in this review is supported by the Huntington's Disease Society of America (USA), Telethon (Italy), The Hereditary Disease Foundation (USA), The HighQ Foundation (USA), Fondazione Cariplo (Italy), Ministero dell’Istruzione dell’Università e della Ricerca (Italy) and NeuroNE (Network of Excellence on Neurodegenerative

References (351)

  • A. Blochl et al.

    Localization of cellular storage compartments and sites of constitutive and activity-dependent release of nerve growth factor (NGF) in primary cultures of hippocampal neurons

    Mol. Cell Neurosci.

    (1996)
  • D. Blum et al.

    Adenosine receptors and Huntington's disease: implications for pathogenesis and therapeutics

    Lancet Neurol.

    (2003)
  • J.M. Canals et al.

    Differential regulation of the expression of nerve growth factor, brain-derived neurotrophic factor, and neurotrophin-3 after excitotoxicity in a rat model of Huntington's disease

    Neurobiol. Dis.

    (1998)
  • J.M. Canals et al.

    The neurotrophin receptors trkA, trkB and trkC are differentially regulated after excitotoxic lesion in rat striatum

    Brain Res. Mol. Brain Res.

    (1999)
  • J. Carmichael et al.

    Glycogen synthase kinase-3beta inhibitors prevent cellular polyglutamine toxicity caused by the Huntington's disease mutation

    J. Biol. Chem.

    (2002)
  • E. Castren et al.

    Brain-derived neurotrophic factor messenger RNA is expressed in the septum, hypothalamus and in adrenergic brain stem nuclei of adult rat brain and is increased by osmotic stimulation in the paraventricular nucleus

    Neuroscience

    (1995)
  • E. Cattaneo et al.

    A short term analysis of the behaviour of conditionally immortalized neuronal progenitors and primary neuroepithelial cells implanted into the fetal rat brain

    Brain Res. Dev. Brain Res.

    (1994)
  • N. Checa et al.

    TrkB and TrkC are differentially regulated by excitotoxicity during development of the basal ganglia

    Exp. Neurol.

    (2001)
  • J.A. Chong et al.

    REST: a mammalian silencer protein that restricts sodium channel gene expression to neurons

    Cell

    (1995)
  • B. Connor et al.

    Trk receptor alterations in Alzheimer's disease

    Brain Res. Mol. Brain Res.

    (1996)
  • B. Connor et al.

    Brain-derived neurotrophic factor is reduced in Alzheimer's disease

    Brain Res. Mol. Brain Res.

    (1997)
  • B.R. Cullen

    Transcription and processing of human microRNA precursors

    Mol. Cell

    (2004)
  • L.P. De Almeida et al.

    Neuroprotective effect of a CNTF-expressing lentiviral vector in the quinolinic acid rat model of Huntington's disease

    Neurobiol. Dis.

    (2001)
  • T. De Cristofaro et al.

    The length of polyglutamine tract, its level of expression, the rate of degradation, and the transglutaminase activity influence the formation of intracellular aggregates

    Biochem. Biophys. Res. Commun.

    (1999)
  • M. DiFiglia et al.

    Huntingtin is a cytoplasmic protein associated with vesicles in human and rat brain neurons

    Neuron

    (1995)
  • E. Di Maria et al.

    No evidence of association between BDNF gene variants and age-at-onset of Huntington's disease

    Neurobiol Dis.

    (2006)
  • A. Dubey et al.

    Effect of age and caloric intake on protein oxidation in different brain regions and on behavioral functions of the mouse

    Arch. Biochem. Biophys.

    (1996)
  • M.F. Egan et al.

    The BDNF val66met polymorphism affects activity-dependent secretion of BDNF and human memory and hippocampal function

    Cell

    (2003)
  • T. Falkenberg et al.

    Increased expression of brain-derived neurotrophic factor mRNA in rat hippocampus is associated with improved spatial memory and enriched environment

    Neurosci. Lett.

    (1992)
  • J.P. Fawcett et al.

    Detection of brain-derived neurotrophic factor in a vesicular fraction of brain synaptosomes

    J. Biol. Chem.

    (1997)
  • I. Ferrer et al.

    Brain-derived neurotrophic factor in Huntington disease

    Brain Res.

    (2000)
  • T. Aid et al.

    Mouse and rat BDNF gene structure and expression revisited

    J. Neurosci. Res.

    (2007)
  • J. Alberch et al.

    Association between BDNF Val66Met polymorphism and age at onset in Huntington disease

    Neurology

    (2005)
  • C.A. Altar et al.

    Anterograde transport of brain-derived neurotrophic factor and its role in the brain

    Nature

    (1997)
  • K.D. Anderson et al.

    Ciliary neurotrophic factor protects striatal output neurons in an animal model of Huntington disease

    Proc. Natl. Acad. Sci. U.S.A.

    (1996)
  • S.E. Andrew et al.

    The relationship between trinucleotide (CAG) repeat length and clinical features of Huntington's disease

    Nat. Genet.

    (1993)
  • M.E. Andres et al.

    CoREST: a functional corepressor required for regulation of neural-specific gene expression

    Proc. Natl. Acad. Sci. U.S.A.

    (1999)
  • E. Arenas et al.

    Effects of BDNF and NT-4/5 on striatonigral neuropeptides or nigral GABA neurons in vivo

    Eur. J. Neurosci.

    (1996)
  • L. Arning et al.

    NR2A and NR2B receptor gene variations modify age at onset in Huntington disease

    Neurogenetics

    (2005)
  • P.A. Bresnahan et al.

    Human fur gene encodes a yeast KEX2-like endoprotease that cleaves pro-beta-NGF in vivo

    J. Cell Biol.

    (1990)
  • A.W. Bruce et al.

    Genome-wide analysis of repressor element 1 silencing transcription factor/neuron-restrictive silencing factor (REST/NRSF) target genes

    Proc. Natl. Acad. Sci. U.S.A.

    (2004)
  • A.J. Bruce-Keller et al.

    Food restriction reduces brain damage and improves behavioral outcome following excitotoxic and metabolic insults

    Ann. Neurol.

    (1999)
  • A.C. Bachoud-Levi et al.

    Neuroprotective gene therapy for Huntington's disease using a polymer encapsulated BHK cell line engineered to secrete human CNTF

    Hum. Gene Ther.

    (2000)
  • A.A. Badawy et al.

    Effects of acute paroxetine administration on tryptophan metabolism and disposition in the rat

    Br. J. Pharmacol.

    (1991)
  • C.D. Bailey et al.

    Tissue transglutaminase contributes to disease progression in the R6/2 Huntington's disease mouse model via aggregate-independent mechanisms

    J. Neurochem.

    (2005)
  • N.S. Bamford et al.

    Dopamine modulates release from corticostriatal terminals

    J. Neurosci.

    (2004)
  • Z.C. Baquet et al.

    Early striatal dendrite deficits followed by neuron loss with advanced age in the absence of anterograde cortical brain-derived neurotrophic factor

    J. Neurosci.

    (2004)
  • Y.A. Barde et al.

    Purification of a new neurotrophic factor from mammalian brain

    EMBO J.

    (1982)
  • P.E. Batchelor et al.

    Activated macrophages and microglia induce dopaminergic sprouting in the injured striatum and express brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor

    J. Neurosci.

    (1999)
  • M.F. Beal

    Neurochemistry and toxin models in Huntington's disease

    Curr. Opin. Neurol.

    (1994)
  • Cited by (469)

    View all citing articles on Scopus
    View full text