Abstract
Neurodegenerative diseases exemplified by Alzheimer’s and Huntington disease are characterized by the progressive neuropsychiatric dysfunction and loss of specific neuronal subtypes. Even though there are differences in the exact sites of pathology and clinical profiles only partially overlap, considerable similarities in disease mechanisms and pathogenic pathways can be observed. These shared mechanisms raise the possibility of common therapeutic targets for drug development. Huntington disease with a monogenic cause and the possibility to accurately identify pre-manifest mutation carriers could be exploited as a ‘model’ for Alzheimer’s disease to test the efficacy of therapeutic interventions targeting shared pathogenic pathways.
Introduction
The rapidly aging population in the developed world, spurred on by an increase in life expectancy, has led to increased prevalence of late onset neurodegenerative disorders and imposes an enormous financial and social burden on health care systems as well as society as a whole. In 2010, for example, 6 million people in the US were over the age of 85, and this number is projected to quadruple by 20501.
The clinical symptoms of neurodegenerative disorders such as Alzheimer’s disease (AD) and Huntington disease (HD) are progressive and debilitating. The hallmark of HD is motor disability that features chorea, while the main symptom of AD is dementia. Nevertheless, patients with HD and AD share many clinical manifestations. These include behavioural and psychiatric disturbances (including depression and apathy) in the early stages and cognitive defects that result in forgetfulness, impaired judgement, disorientation and confusion. Cognitive deficits in patients with HD however are usually less severe than in AD; Patients also exhibit difficulty in ambulation and eating at late stages of both diseases, which ultimately lead to death1, 2.
AD is genetically heterogeneous and can be caused by any one or more of several genes as well as environmental factors. Familial AD (FAD), which accounts for less than 1% of all AD cases, are caused by rare mutations in genes encoding for amyloid precursor protein (APP), presenilin (PSEN)-1 and -21, 3. Numerous genes are significantly associated with sporadic late-onset AD. The ε4 allele of apolipoprotein E (APOE) is the single strongest genetic risk factor for sporadic AD4. APP cleavage by the β-secretase BACE1 and the γ-secretase complex, which consists of PSEN1 or PSEN2, anterior pharynx-defective-1 (APH-1), presenilin enhancer-2 (PEN-2) and nicastrin, leads to the generation of the extracellular Aβ peptide. This peptide fragment is prone to aggregate and form amyloid plaques that can be detected in post-mortem brain from AD patients5, 6. In addition to amyloid plaques, neurofibrillary tangles made up of hyperphosphorylated tau aggregates, a microtubule protein, are also observed in post-mortem brain from AD patients5, 6. In contrast, HD is a monogenic disorder with autosomal dominant inheritance and is caused by a CAG repeat that expands to 36 copies or more in the gene encoding the huntingtin (Htt) protein2, resulting in an expansion of the polyglutamine tract. Furthermore, the length of the CAG tract is directly correlated with the disease onset, with longer expansions leading to earlier onset2. β-secretase, γ-secretase and the Aβ peptide are well validated therapeutic targets in AD7, while mutant Htt (mHtt) is a promising target in HD8, 9. However these targets are not shared between AD and HD and will therefore not be the focus of this review.
At a neuropathological level, the diseases are initially characterized by a specific loss of certain neuronal subtypes. In HD, it is the medium-spiny neurons (MSNs) in the striatum that undergo atrophy in early stages of the disease, while in AD, large pyramidal neurons in the CA1 zone of the hippocampus as well as neurons in the basal forebrain and the entorhinal cortex are sites of early disease10-12. The process of neuronal dysfunction and death is progressive, and early changes are followed by a more wide-spread atrophy of the brain10, 13. Considerable progress has been made in the elucidation of mechanisms that lead to neurodegeneration in AD and HD. There is evidence for the aberrant phosphorylation, palmitoylation and acetylation of disease-causing proteins, protein misfolding, failure to clear disease-causing proteins by the ubiquitin-proteasome system or autophagy, and changes in NMDA receptor activity at the synapse. Additional mechanisms include alterations in levels of brain-derived neurotrophic factor (BDNF) and neuronal growth factor (NGF) as well as associated receptors and trafficking pathways,, and increased activity of caspase enzymes in both disorders5, 6, 9, 14 (Table 1).
Table 1. Similarities in pathogenic pathways for AD and HD.
| Pathway | Alteration | References |
|---|---|---|
| Synaptic dysfunction | Increased extrasynaptic NR2B-containing NMDARs |
18, 19 |
| Increased phosphorylation of NMDARs | 20, 22 | |
| Affected neurons have defective LTP | 25, 26 | |
| Increased Ca2+ influx | 18 | |
| Defects in mitochondrial trafficking | 18 | |
| Mitochondrial fragmentation & dysfunction |
||
| Alterations in microglial tryptophan metabolism, leading to increased release of neurotoxic metabolites |
5, 14, 35 | |
| Neurotrophic factor related abnormalities |
Reduced BDNF levels | 39, 40 |
| Increasing BDNF & NGF levels is beneficial in disease models |
51-54 | |
| Increased p75NTR levels and signalling | 75, 172 | |
| Decreased Trk receptor levels and signalling |
75, 172 | |
| Increased Gsk3ß activity | 66, 173 | |
| Altered ERK activity | 174 | |
| Reduced velocity and efficiency of axonal transport of BDNF |
65, 66 | |
| Apoptotic pathways | Increased caspase-6 activity | 89, 90 |
| Caspase-6 cleavage of disease proteins | 89-92 | |
| Preventing caspase cleavage of disease proteins is beneficial in mouse models |
93, 94 | |
| Posttranslational modifications |
Palmitoylation of disease proteins is linked to aggregate formation |
118, 119 |
| Phosphorylation of disease proteins reduces their cleavage by caspases |
105, 106 | |
| HDAC inhibition is beneficial in disease models |
69, 126, 127 | |
| Protein aggregation and clearance mechanisms |
Misfolding and aggregation of disease proteins |
128 |
| UPS impairment | 144, 145 | |
| Impaired autophagy | 161 | |
| Upregulation of autophagy is beneficial in disease models |
153, 155, 156, 158, 162, 163, 165, 167, 168 | |
| Increased expression of Beclin 1 is beneficial in disease models |
Although no treatments are available to slow or halt neuronal degeneration and neuronal death, the analysis of disease pathways has led to the identification of common drug targets for AD and HD. These similarities have not been commonly appreciated by the HD or AD research communities, and could lead to the development of drugs that can be used for both disorders. Here we will describe the pathways and therapeutic targets and rank them with target validation (TV) scores based on a scale developed by the Cure Huntington’s Disease Initiative (CHDI) (http://www.hdresearchcrossroads.org, Table 2, 15), which we apply to both diseases for direct comparison. Potential therapeutic targets are hereby ranked by their degree of validation from 1.0 (a gene that is linked to a disease-relevant biological mechanism) to 6.0 (therapeutic modulation of the target demonstrates efficacy in a phase 3 clinical trial) in AD and HD. Although well-studied targets will automatically rank higher in this system than understudied targets, we believe that our ranking can identify common, well-validated targets as well as knowledge gaps that warrant further studies.
Table 2. Target validation (TV) scoring.
| TV score | Description |
|---|---|
| 1.0 | A gene that is conceivably linked to a disease-relevant biological mechanism |
| 2.0 | A gene that is associated with the disease and binds to a disease protein OR has an altered expression pattern in the disease OR has an altered cellular distribution in the disease |
| 2.5 | A gene that shows an altered pathway or functional activity in the disease |
| 3.0 | A gene that shows a causal relationship with the disease when manipulated genetically or pharmacologically in an in vitro or non-rodent lower organismal model of the disease |
| 3.5 | A gene that when genetically manipulated, modifies the disease phenotype in a rodent model of the disease or shows disease-like effects in a normal rodent, OR is identified as a genetic modifier of the disease phenotype in humans by a linkage/association study |
| 4.0 | A therapeutically relevant drug or genetic intervention, that is highly specific for this gene target, improves the disease phenotype in a rodent model of the disease |
| 4.5 | Manipulation of this gene target improves the disease phenotype in a larger, non-rodent mammalian model of the disease |
| 5.0 | A drug or gene therapy known to modulate this gene target demonstrates positive outcome in a Phase 2 clinical trial efficacy in a Phase 3 clinical trial |
| 6.0 | A drug or gene therapy known to modulate this gene target demonstrates efficacy in a Phase 3 clinical trial |
For most targets, it is unknown whether their modulation will be disease-modifying or only lead to the alleviation of symptoms, and the TV score does not discriminate between these possibilities. To clarify this issue, long-term clinical studies are necessary, and we believe that these could initially be conducted in individuals carrying the mutation causing HD, since this population is well defined and alterations in the course of the disease can be determined at very early stages16. HD is the commonest purely genetic neurodegenerative disorders, while AD is the most common neurodegenerative disease overall1, 2. Since many of the pathways and targets described here are also shared with other diseases such as Parkinson’s disease, dementia with Lewy bodies or ataxias, HD could serve as a general model for neurodegeneration, where well-defined animal models are available for preclinical studies and endpoints for human clinical trials are well established16, 17. The “Enroll-HD” study that is currently underway will provide a registry of HD mutationcarriers worldwide and serve as a resource for recruiting patients to clinical trials (http://www.enroll-hd.org).
Targeting synaptic dysfunction in HD and AD
Excitotoxicity has been implicated in neuronal death in HD and AD. The N-methyl-D-aspartate (NMDA) subtype of glutamate receptors is thought to be a major contributor to excitotoxic cell death because of its high permeability to calcium (Ca2+)18. The NR2A and NR2B subunits of NMDAR differ in their sensitivities to agonists and antagonists, their channel gating properties, and in their localization. NR2A-containing receptors are generally found in the synapse, while NR2B-containing receptors are predominantly localized at extrasynaptic sites18. Enhanced activation of extrasynaptic, NR2B-containing NMDAR is common in both HD and AD.
Aberrant extrasynaptic NMDA receptor activity in HD and AD
The presence of mHtt and APP alters the subcellular distribution of NMDAR18, 19. In mouse models of both diseases, deficits in trafficking mechanisms increase cell surface levels of NR2B and decrease NR2B internalization18. Increased levels of NR2B on the cell surface results in enhanced NMDAR currents in mouse models of HD and AD19. Both wild type Htt and tau, a neuronal protein that stabilizes microtubules, function in NR2B trafficking as well as the recruitment of kinases to NMDARs at the cell surface. MHtt disrupts these processes, resulting in altered tyrosine phosphorylation of NR2B and enhanced NMDAR-mediated toxicity20. Similarly, Aβ-induced NMDAR dysfunction is mediated by Fyn, a tyrosine kinase that phosphorylates NR2B21, 22 and mediates its insertion into the plasma membrane23 increasing the levels of NR2B on the cell surface. The activation of extrasynaptic NR2B-containing NMDARs also dephosphorylates and inhibits the CREB/PGC1α signalling pathway, which makes cells expressing mHtt more susceptible to cell death24 (Figure 1). Enhanced activation of extrasynaptic NMDAR further leads to excessive influx of Ca2+ into the cell, which results in inappropriate activation of enzymes (i.e., calpains and other Ca2+-regulated enzymes) and mitochondrial dysfunction, and leads to apoptosis18.
Figure 1. Therapeutic targets in synaptic dysfunction pathway.
NMDAR receptor signalling can be triggered by glutamate released from the presynaptic neuron or through the NMDAR agonist quinolinic acid (QA), a product of tryptophan metabolic pathways in microglia. While glutamate signalling through synaptic NMDARs promotes neuronal survival, QA leads to excessive NMDAR activation and synaptic dysfunction. The levels of QA generation can be altered through inhibition of kynurenine 3-monooxygenase, an enzyme in the tryptophan metabolic pathway. An imbalance in synaptic and extrasynaptic NMDARs is seen in both HD and AD. Htt and Tau are both involved in regulating the trafficking of NR2B to the extrasynaptic membrane. NR2B-containing NMDARs are hyperphosphorylated as mHtt and Tau increase recruitment of kinases to NR2B-containing NMDARs. An increase in both the levels and phosphorylation of NR2B-containing NMDARs leads to an increased extrasynaptic NMDA current that triggers cell death through multiple pathways. Two major pathways leading to cell death in HD and AD are represented: 1) inhibition of CREB phosphorylation that inhibits pro-survival pathways and 2) increases in Ca2+ influx, leading to mitochondrial dysfunction. Therapeutic targets are highlighted in red; Drugs are highlighted in green.
Activation of extrasynaptic NMDARs may also underlie the cognitive impairment that is observed in patients with HD and AD. Long term potentiation (LTP), a measure of synaptic plasticity, is thought to be a mechanism underlying learning and memory. In mouse models of AD and HD, neurons derived from affected brain regions have defective LTP 25, 26. Furthermore, the activation of extrasynaptic NMDARs in a mouse model of AD promotes ß-secretase cleavage of APP, which results in increased levels of Aβ that are directly correlated with the severity of the cognitive deficit27. The presence of Aβ oligomers, early intermediates in the Aβ aggregation pathway, has in turn been associated with increased activation of extrasynaptic NMDARs, which could cause a feedback loop leading to synaptic dysfunction28. However, the strongest evidence for the role of extrasynaptic NMDAR in cognitive impairment is the effectiveness of memantine, an NMDAR blocker (see below), in rodent models of HD and AD as well as in AD patients24, 29-31.
Therapeutic approaches
Inhibition of extrasynaptic NMDARs can be achieved using memantine treatment. At a low dose (1 mg/kg = 3 mg/m2 human equivalent dose (HED)), memantine blocks extrasynaptic but not synaptic NMDAR activity, whereas high doses (30 mg/kg = 90 mg/m2 HED) inhibit both synaptic and extrasynaptic NMDARs (Figure 1). A low dose of memantine rescues neuropathological and behavioural phenotypes as well as electrophysiological abnormalities in models of HD. By contrast, high dose memantine worsens these symptoms24, 29. Similarly, administration of a low memantine dose corrected defects in learning and memory tasks, such as performance in the Morris water maze and passive avoidance learning in a mouse model of AD30.
Memantine treatment has also been tested in patients with AD and is now approved by the FDA for this indication (Namenda XR, Forest Laboratories, NDA no. 022525). In clinical trials, patients with AD who received 28 mg of memantine (extended release formulation) for 24 weeks showed significantly reduced decline in cognitive symptoms in two measures of cognitive performance compared to patients receiving placebo (FDA approval review, http://www.accessdata.fda.gov/drugsatfda_docs/nda/2010/022525Orig1s000MedR.pdf). Longer studies are needed to determine whether the effects of memantine are purely symptomatic or disease modifying. Nevertheless, the improvements seen in patients with AD are encouraging and NR2B receptors are currently the best validated of the shared therapeutic targets between AD and HD, with TV scores of 6 and 4, respectively (Suppl Table S1). In view of open label studies showing some potential benefit, clinical trials with memantine in HD patients might be considered.32-34
Non-neuronal contributions to excitotoxic pathways
Another striking similarity between AD and HD is the presence of activated microglia as markers of inflammation5, 14. Microglia produce quinolinic acid (QA), a selective NMDAR agonist and a metabolite of the tryptophan degradation pathway, that elicits symptoms reminiscent of HD when injected into the striatum, and symptoms similar to AD when injected into the nucleus basalis of rodents35. A recent study furthermore demonstrates that inhibiting kynurenine 3-monooxygenase, an enzyme in the pathway generating QA, can ameliorate excitotoxicity and disease phenotypes in mouse models of both AD and HD35.
Modulating neurotrophic factors and receptors in HD and AD
Neurotrophic factors, including neural growth factor (NGF) and brain-derived neurotrophic factor (BDNF), are secreted peptides that have prominent roles in neuronal development, health and function of the brain regions that are most affected in patients with AD and HD36. NGF specifically binds tyrosine protein kinase A (TrkA) receptors, while BDNF recognizes TrkB receptors to activate downstream signalling pathways. In addition, both bind the p75 neurotrophin receptor (p75NTR) with lower affinity, which is associated with pro-death signalling following neuronal injury36 (Figure 2). The unprocessed, precursor forms of NGF and BDNF (pro-NGF and pro-BDNF, respectively) also bind p75NTR, leading to apoptosis36. Alterations in the levels of NGF and BDNF, the levels and localization of cognate receptors and changes in the pathways activated by these neurotrophins have been implicated in the pathogenesis of HD and AD (Figure 2, Suppl Table S1).
Figure 2. Therapeutic targets in the neurotrophin pathway.
BDNF and NGF levels are reduced in both HD and AD. Presynaptically, Htt, APP and Tau influence trafficking mechanisms via interaction with HAP1, kinesin and dynein/dynactin, influencing their binding to microtubules. Pro-BDNF and pro-NGF are processed by unknown proteases to generate mature BDNF and NGF. Furthermore, TrkA and TrkB receptor levels are decreased in AD and HD, respectively. Signalling through Trk receptors is reduced, thereby increasing Gsk3ß activity and enhancing cell death pathways. Increased p75NTR levels and signalling mediated by pro-BDNF and pro-NGF also triggers cell death. Therapeutics examined to date include: Epothilone D, BDNF, NGF, glucocorticoids and Lithium. Therapeutic targets are highlighted in red; Drugs are highlighted in green.
Increasing BDNF as a therapeutic approach
Reduced levels of BDNF are observed in the striatum and the hippocampus in patients suffering from HD and AD, respectively37, 38. Polymorphisms in the gene encoding BDNF have been positively associated with age of onset of AD in some studies39, 40,41, although this association is controversial42-44. Similarly, data on an association between BDNF polymorphisms and age of onset in HD has also been conflicting45,46.
The ability to increase BDNF levels through exercise is well documented47. Using APP/PSEN1 transgenic mice, it has been shown recently that exercise increases BDNF levels and prevents decline in spatial learning and memory through the improvement of LTP in the hippocampus48. The beneficial effects of BDNF overexpression using genetically-modified mesenchymal stem cell transplantation, viral-mediated expression or intracranial injections of BDNF in different rodent models of HD and AD validate BDNF as a therapeutic target and led to a TV score of 4.5 and 4.0 for AD and HD, respectively (Suppl Table S1). Overexpression of BDNF protects against neurotoxicity, prevents loss of neurons, corrects motor dysfunction, improves procedural learning and corrects synaptic plasticity49-51. Increasing BDNF in a non-human primate model of AD also improved hippocampal learning and ameliorated neuronal death with no observable adverse effects52.
Pharmacological agents that are currently used or are undergoing testing for other clinical applications, such as lithium or the ampakine CX929, can increase BDNF levels by increasing its expression and promoting its trafficking and improve phenotypes in animal models of HD and patients suffering from AD53, 54. A Phase II clinical trial studying the effect of lithium treatment in patients with AD is currently underway (ClinicalTrials.gov identifier: NCT00088387), and in a small cohort of patients lithium reduced the progression from mild cognitive impairment to full AD55. In addition to increasing BDNF levels, lithium also inhibits the hyperphosphorylation of Tau mediated by GSK3β and has additional beneficial effects in the activation of pro-survival CREB signalling and the activation of autophagy 56-59(Figure 2, and see below). Due to these multiple effects, lithium is a suboptimal tool compound to investigate specific pathways, but remains a potential therapeutic.
Trafficking defects in HD and AD: Targeting microtubule-associating proteins
Striatal MSNs rely on BDNF release from innervating cortical neurons for their normal functioning and survival, which makes axonal trafficking of BDNF from the cell body of cortical neurons to the synapse an important process60. Defects in intracellular trafficking have been suggested as a cause for the reduced BDNF levels in the brains of patients with HD or AD60, 61. Interestingly, polymorphisms in the gene encoding BDNF that are associated with an increased risk of AD and reduced age of onset of HD impair the binding of pro-BDNF to the huntingtin-associated protein-1 (HAP1), which is necessary for the intracellular trafficking of pro-BDNF60, 62. These polymorphisms could therefore lead to reduced BDNF levels in both diseases (Figure 2).
Htt and Tau are predominantly found in the cytoplasm where they colocalize with microtubules and vesicular structures and associate with proteins that are involved in intracellular trafficking61, 63. Wild type Htt interacts with HAP1 that associates with proteins essential for intracellular trafficking, such as kinesin and p150Glued (a subunit of dynactin)60, 63. Similarly, Tau, a microtubule-associated protein, regulates axonal transport by inhibiting the motor activity of kinesin and dynein (Figure 2, Suppl Table S1)63, 64.
The velocity and efficiency of the transport of BDNF-containing vesicles are reduced in the presence of mHtt, PSEN1 mutations and hyperphosphorylated Tau60, 61. In both neuronal cultures and post-mortem extracts of brains from patients with HD, mHtt shows increased binding to HAP1, which in turn disrupts HAP1 binding to kinesin and reduces the HAP1/p150Glued interaction, thereby reducing the transport of BDNF-containing vesicles60. Interestingly, HAP1 deficiency also affects kinesin-dependent transport of APP, which suggests a role for HAP1 in the pathogenesis of AD65. Mutations in PSEN1 that are associated with FAD deregulate the glycogen synthase kinase-3 ß (Gsk3ß), leading to hyper-phosphorylation of Tau and its detachment from microtubules, which results in cytoskeletal collapse and deficits in axonal transport that contribute to the pathogenesis of AD 61, 66, 67 (Figure 2).
Although there are currently no therapies to ameliorate trafficking deficits to increase BDNF levels, this pathway has many potential therapeutic targets, such as HAP1, kinesin, dynein/dynactin, and microtubules, which could be modified for the treatment of HD and AD.
Inhibitors of Histone deacetylases (HDACs), which are currently being used as cancer therapeutics 68, influence axonal transport through increased acetylation of α-tubulin, enhancing its binding to kinesin4, 69. The modulation of HAP1 binding to mHtt would likely be harder to achieve, since the strengthening of a protein-protein interaction is generally not considered a druggable intervention. However, microtubule (MT)-binding/stabilizing drugs, such as paclitaxel and epothilones that are also currently used as cancer therapy70-72, could be tested in models of HD and AD to better validate therapeutic targets in axonal transport pathways (Suppl Table S1). Recently, epothilone D, a MT-binding/stabilizing agent from the epothilone natural product family, was shown to penetrate the blood-brain barrier and increase MT stability in the CNS of Tau transgenic mice71. The identification and evaluation of MT-binding/stabilizing agents is therefore a promising research area for the treatment of neurodegenerative disorders with impaired axonal transport such as HD and AD.
Increasing NGF is beneficial
A reduction in the levels of mature NGF is observed in the basal forebrain of aged animals and in rodent models of AD and patients with AD73. However, the levels of NGF in human patients with HD have not been reported. Nevertheless, overexpression of NGF in both model systems of HD and AD has beneficial effects that are similar to BDNF overexpression. Rat models of AD and mouse models of HD transplanted with NGF-loaded microspheres in the forebrain show significant improvement in learning and memory tasks and better survival of cholinergic neurons74, 75. Intrastriatal transplantation of genetically engineered mesenchymal stem cells expressing NGF and administration of an amplifier of NGF into transgenic mouse models of HD slow neurodegenerative processes and rescue behavioural deficits50.
Human therapeutic trials that assess the effects of increasing NGF levels in patients with AD are underway. In a phase I clinical study, the implantation of modified fibroblasts expressing human NGF in the basal forebrain of patients with AD resulted in a decrease in the rate of cognitive decline76. Currently, therapeutic options that are less invasive are being explored, including the intranasal delivery of recombinant NGF, which rescued behavioural and neuropathological defects in a mouse model of AD77. NGF, like BDNF, is therefore a highly validated target for AD and HD with TV scores of 4.5 and 4.0, respectively (Suppl Table S1).
Activating Trk and reducing p75NTR signalling as a therapeutic strategy
In patients with AD and HD and animal models, there is an increase in p75NTR levels and a decrease in TrkA and TrkB levels, respectively73, 78. Neurotrophin-mediated cell death that occurs through activation of p75NTR is mediated through caspase-679, which emerges as a therapeutic target for both AD and HD in multiple studies (Suppl Table S1, see below).
Neurotrophin signalling through Trk receptors activates pro-survival pathways, such as the PI3K/Akt signalling pathway. However, reduced signalling through Trk receptors results in increased Gsk3ß activity, which has been implicated in the prevention of LTP, induction of microglia-mediated inflammation and promotion of neuronal death in models of AD and HD57, 80. In AD, increased Gsk3ß signalling also increases Tau phosphorylation and amyloid production, while in HD, increased Gsk3β activity enhances the toxicity of mHtt57, 80.
Reduced NGF signalling also results in the increased production and intracellular aggregation of Aß peptides, mediated through increased transcription of APP and increased processing of APP by ß- and γ-secretases81. The pathogenic effects of altered Trk and p75NTR signalling in HD are not well-characterised (Suppl Table S1).
Glucocorticoids (GCs) have protective effects in the CNS and this protection is dependent on an increase in Trk receptor activity82. Such treatments may be beneficial in HD and AD, but this remains to be shown in the relevant disease models, which is reflected by currently low target validation scores for Trk receptors (Figure 2, Suppl Table S1).
Targeting apoptotic pathways in HD and AD
The caspase proteases are best known for their roles in apoptotic cell death, where they are activated in a signalling cascade involving either extrinsic signals mediated through death receptors or intrinsic pathways initiated by DNA or mitochondrial damage83. Both pathways culminate in the activation of one or more members of the so called executioner caspase subfamily (caspases-3, -7 and -6), which cleave cytoskeletal proteins as well as pro-survival and antiapoptotic factors and thereby commit the cell to death83. However, in addition to this classical role, caspases are also involved in non-apoptotic processes84. In the brain in particular, activation of the ‘executioner’ caspases has been observed in events associated with learning and memory such as synaptic plasticity and LTP, as well as the developmental pruning of axons85-88. These findings challenge the view of caspases as the final executioners of cell death and suggest that the role of these enzymes in neurodegeneration might be more upstream in signalling pathways, where they could mediate early neuronal dysfunction.
Preventing caspase-6 cleavage is beneficial in AD and HD
Caspase-6 is activated in brain tissue from pre-symptomatic individuals carrying the HD mutation and in individuals with only mild cognitive impairment that could progress into AD, supporting the idea of caspase activation as an early event in the pathogenesis of neurodegenerative disease89, 90. These findings are paralleled by results that show the aberrant cleavage of caspase-6 substrates in brain tissue from patients suffering from HD or AD90-92. Interestingly, the disease-causing proteins mHtt, Tau and APP are substrates for caspase-6, and the abrogation of mHtt or APP cleavage in mouse models is protective and leads to a dramatic improvement of the disease phenotype93, 94.
Therapeutic approaches
While the findings discussed above make caspase-6 an interesting and well-validated target in the apoptotic pathway for both diseases (Suppl Table S2), it may not easily be tractable by small molecule inhibitors. The inhibition of ‘executioner’ caspases by compounds such as IDN-6556 (Pfizer), compound 34 (Sunesis) or M867 (Merck-Frosst) may also have some liabilities including the concern of carcinogenic side-effects95, 96. An alternative would be to inhibit activators of caspase-6. However the exact mechanism underlying aberrant caspase activation is not understood in either AD or HD. Activation could occur through the activation of ‘initiator’ caspases such as caspase-9 via death or neurotrophic receptors (DR6, p75NTR, TrkB or TrkA). As described above, alterations in these receptors occur in HD and AD, and neuronal death mediated through p75NTR in a seizure model is dependent on the downstream activation of caspase-679 (Figure 3).
Figure 3. Therapeutic targets in the apoptotic pathway.
The aberrant activation of caspase-6 that is seen in animal models and patients of both HD and AD could be mediated through multiple pathways: 1) Excitotoxicity, as described above, leads to an excessive influx of Ca2+, which can depolarize mitochondria and lead to caspase activation. 2) Aberrant signalling through trophic receptors or death receptors as outlined can result a release of cytochrome c and Smac/Diablo from mitochondria, which activate caspase-9 and inhibit Bcl-2 as well as IAP proteins. These events lead to a subsequent activation of caspase-6 by both blocking of inhibitory pathways as well as direct activation through caspase-9. 3) Caspase-6 and -3 could furthermore be activated through the intrinsic pathway, since evidence for DNA and mitochondrial damage, ER stress and protein misfolding as well as oxidative stress is found in AD and HD patients and animal models. Degradation of caspase-6 through the proteasome reduces its activity. Therapeutic targets are highlighted in red.
The intrinsic apoptotic pathway is another well-described paradigm for caspase activation and it can be triggered by ER stress, protein misfolding (see below), DNA and mitochondrial damage or oxidative stress83. mHtt and Aß may directly alter mitochondrial function in HD and AD. Htt can be found at the mitochondrial membrane97 and in the presence of mHtt oligomers, expression levels of mitochondrial fusion and fission proteins are altered. These changes in expression levels may be responsible for decreased mitochondrial function by causing structural abnormalities in and defective trafficking of mitochondria in HD97. In AD, mitochondria, especially synaptic mitochondria, show age-dependent Aß accumulation98, 99, which enhances the permeability of the mitochondrial permeability transition pore and causes the generation of reactive oxygen species99. Clinical trials of antioxidants that might ameliorate oxidative stress, DNA and mitochondrial damage, have not yielded conclusive results in patients with either AD or HD100, 101. However a better understanding of endogenous antioxidant levels in patients before and after treatment might help to stratify potential responders from non-responders100.
Caspases can also be activated by disturbances of developmental and synaptic plasticity signals such as axonal pruning or LTP85-88, 102. In these non-apoptotic paradigms, caspase activation is usually contained in a well-defined subcellular compartment such as the synapse, and the active executioner caspases are kept under tight control by endogenous inhibitor proteins or through degradation by the proteasome88, 103. This insures that the activation of enzymes such as caspase-3, which are usually highly efficient in amplifying the cell death cascade, remains locally contained and does not trigger the demise of the whole cell. Under pathological conditions, toxic proteins or fragments thereof (such as mHtt, Aß) can cause proteasomal dysfunction, which might lead to an impaired degradation and thus accumulation of active caspase-6 and -3103. The resulting increase in caspase activity could then lead to an amplification of the apoptotic cascade and commit the cell to death, even though the initial caspase activating signal was meant to only trigger non-apoptotic functions.
Further studies are needed to better understand apoptotic pathways in AD and HD and to better validate therapeutic targets that are further upstream in the signalling cascade.
Targeting posttranslational modifications of disease-causing proteins
Although the proteolytic cleavage of mHtt and APP can be considered a relevant posttranslational modification (PTM) in the pathogenesis of both disorders, additional, reversible PTMs of mHtt, Aß and Tau have been described extensively6, 9, 104 and lead to considerable overlap in potential therapeutic strategies that will be discussed below.
Kinases and phosphatases
Both mHtt and APP are phosphorylated at multiple sites, and phosphorylation at certain amino acids (S421 for mHtt, T668 for APP) reduces the amount of caspase cleavage at adjacent sites as well as the toxicity that is associated with this process105, 106. mHtt is phosphorylated at S421 by Akt, and its dephosphorylation depends on the phosphatases PP1/PP2A107, 108, whereas APP is phosphorylated by Cdk5 and Gsk3β in neurons104 (Suppl Table S3). Gsk3β is also the major kinase phosphorylating Tau in AD, which leads to the formation of insoluble Tau aggregates (Tau tangles) and subsequent disruption of the microtubule system67. The phosphatases that are responsible for the dephosphorylation of APP or Tau are unknown.
Therapeutic strategies
Inhibition of Gsk3 using lithium is protective in models of AD, which has been partly attributed to a decrease of Tau hyperphosphorylation and improved BDNF trafficking (see above) as well as a reduction in APP cleavage and Aß accumulation, since the GSK3α isoform has been implicated in processing of APP by γ-secretase67, 109. Lithium treatment has also shown benefits in mouse models of HD57, 110, and levels of GSK3β are increased prior to symptom onset in an HD model111.The treatment of primary neurons derived from these mice with GSK3β inhibitors reduces neuronal death111 (Suppl Table S3).
Sodium valproate, which is widely used as a mood stabilizer and antiepileptic drug, has also shown promising neuroprotective effects in several models of AD112. It inhibits GSK3β, but has additional effects by acting as a transcriptional modulator through the inhibition of HDACs, and by reducing excitotoxicity110, 113. While valproate might not be suited for the management of agitation in dementia114, longer-term studies are necessary to be able to judge its neuroprotective effects.
PP1/PP2A phosphatase inhibition is a possible strategy to boost phosphorylation of mHtt in HD and reduce its toxicity108. As illustrated by the low TV score for phosphatases (Suppl Table S3), further validation studies are needed before phosphatase inhibition can be pursued as a therapeutic option. Of note, the identity of the PP1/PP2A regulatory subunits mediating Htt dephosphorylation remains unknown, although their inhibition might be more successful than the development of active site inhibitors, since the active phosphatase subunits are very promiscuous in terms of substrate specificity115. Upregulation of Akt activity on the other hand could be achieved through the modulation of the upstream IGF-1 and PI3K signalling pathway107. This pathway also emerges as a target in the upregulation of autophagy in both AD and HD.
Palmitoylation: Palmitoyl transferases and Thioesterases as targets in AD and HD
Palmitoylation116-119, a PTM that attaches a single palmitate moiety to a cysteine residue, is important in synaptic transmission and neuronal plasticity120. In AD, palmitoylation of BACE1 reduces the shedding of the ectodomain of the protease, which in turn reduces the generation of Aß119. In HD, the Htt protein itself is palmitoylated by the palmitoyl transferases HIP14 and HIP14L117, and acts as a cofactor for the palmitoylation of other HIP14 substrates121. The interaction of mHtt with HIP14 is reduced in a mouse model of HD118. The resulting decrease in the palmitoylation of multiple synaptic substrates could lead to the phenotype observed in HD122 (Suppl Table S3). Furthermore, the ablation of HIP14 leads to a phenotype similar to HD in a mouse model123.
An increase of palmitoyl transferase activity or the inhibition of thioesterases could therefore be beneficial by reducing Aß generation in AD as well as normalizing the aberrant palmitoylation of numerous synaptic substrates in HD. However, the lack of information on the palmitoyl transferase(s) involved in BACE1 palmitoylation and on the thioesterases depalmitoylating Htt or BACE1 warrants further studies before targeted therapies to correct palmitoylation defects can be developed.
Aberrant protein acetylation and HDACs in HD and AD
In both AD and HD, transcriptional dysregulation that is linked to altered histone acetylation as well as defective axonal transport due to a lack of tubulin acetylation69, 124. These defects could account for a variety of the disease phenotypes associated with AD and HD, such as the altered expression and transport of neurotrophic factors125, and might therefore be upstream events in the pathogenic pathways of both disorders.
As a therapeutic strategy to increase acetylation, the inhibition of HDACs by compounds such as phenyl butyrate, trichostatin A or Suberoylanilide hydroxamic acid (SAHA) is protective in cellular and animal models of both AD and HD69, 126, 127 (Suppl Table S3). HDAC inhibitors such as SAHA (Vorinostat, Merck) or valproic acid are currently undergoing Phase II/III clinical trials for different types of cancer, which together with the high degree of target validation for HDACs makes them good candidates for clinical trials in neurodegenerative disease.
Protein aggregation and clearance mechanisms in AD and HD
Protein aggregation is a prime neuropathological hallmark of AD and HD that has been recognized for many decades. In HD, this involves the deposition of intracellular (and intranuclear) aggregates of mHtt, whereas in AD, there is accumulation of extracellular Aß plaques and intracellular Tau tangles128. The accumulation of misfolded proteins that have amyloid characteristics (a tertiary structure that is rich in β-sheets and can be visualized by staining with dyes such as Congo red and Thioflavin S) is thought to result from improper folding of the mutant proteins as well as insufficient clearance mechanisms129. Endogenous chaperones, in particular the heat-shock proteins, prevent misfolding and aggregation of mHtt, Tau and Aβ130, and their overexpression is protective against neurotoxic insults such as excitotoxicity131 (Suppl Table S4).
Targeting protein misfolding directly and/or through chaperones
Large screening campaigns have led to the identification of a variety of compounds that directly interfere with Aß, mHtt and Tau protein misfolding and aggregation in cell-free systems, as well as compounds that modulate the activity of heat-shock proteins and other chaperones132-138 (Figure 4). However, for both classes of compounds, the translation of therapeutic efficacy into mouse models has been difficult. Geldanamycin, a compound that is highly efficient in the upregulation of chaperone proteins and can thus reduce amyloid formation in cell culture, has the drawback of substantial toxicity and low blood-brain-barrier (BBB) penetration. Another example is (-) epigallocatechin gallate, a compound that provides benefits to patients suffering from systemic amyloidoses by reducing the amount of aggregated protein in tissues such as the heart139. It stabilizes a non-toxic form with low β-sheet content of both mHtt and Aß134, 135, and although some studies report beneficial effects in animal models of AD140, delivery to the central nervous system is difficult141. Lack of translation of the effects seen in cell culture into mouse models has also been attributed to the high drug-to-amyloid protein ratios that need to be achieved to directly influence aggregation.
Figure 4. Therapeutic targets in the protein misfolding pathway.
The disease proteins mHtt, Tau and Aβ undergo misfolding and aggregation, which is a multi-step process involving misfolded monomers, oligomers and large aggregate structures. Endogenous chaperone proteins of the Hsp family can inhibit aggregation and are likely able to interfere at different steps. Hsp’s can be transcriptionally upregulated through Heat shock factor 1 (Hsf-1), which in turn is activated by compounds such as Celastrol or Geldanamycin that are known to reduce protein aggregation and toxicity in cell culture and Drosophila models of AD and HD. Chemical compounds that directly interfere with the aggregation cascade are often identified as inhibitors of aggregate formation, and with the exception of EGCG it is unknown for most compounds at which step they interfere. Therapeutic targets are highlighted in red; Drugs are highlighted in green.
A major limitation to the development of aggregation inhibitors is the fact that the protein misfolding and aggregation pathway is incompletely understood. While early studies focused on preventing the formation of large amyloid aggregates, it is now clear that misfolding events occurring early in the amyloidogenic process lead to the generation of monomers or small oligomers that can mediate neurotoxicity and degradation142, 143. Due to difficulties in detecting these smaller toxic assemblies, the exact mechanism of action for many aggregation inhibitors such as the benzothiazoles, C2-8 or triazines remains unknown, and while they prevent the formation of large Aß plaques and mHtt aggregates132, 133, 136, they might not interfere with initial misfolding steps and thus not have a significant impact on pathogenesis (Figure 4).
Proof for the therapeutic benefits of inhibitors of protein aggregation in mammalian models of AD and HD is therefore necessary to increase the TV score (Suppl Table S4), and such studies would also answer the question whether the prevention of protein aggregation alone is sufficient to slow down the pathogenesis in AD and HD.
Increasing the clearance of misfolded proteins: The ubiquitin-proteasome system as a target
Intracellular aggregates of both mHtt and Tau contain high levels of ubiquitin, which led to the hypothesis that proteasomal degradation of ubiquitinated proteins could be impaired in HD and AD, and might lead to the accumulation of amyloid aggregates144, 145. There is conflicting evidence for an impairment of the ubiquitin-proteasome system (UPS) in HD and AD. A recent time-lapse study of single neurons that expressed mHtt showed that the UPS is impaired before the formation of visible aggregates. However UPS function normalizes at later time points in the presence of aggregates146. Recent studies further show that the UPS has an important function in the degradation of signalling molecules at the synapse, and contributes to synaptic plasticity147. Since synaptic dysfunction occurs early in the pathogenesis of neurodegenerative disease, it is possible that localized UPS impairment plays an important role, even if overall UPS function at other subcellular localizations remains intact148.
Although the UPS is a popular target in cancer, where its activity is reduced by compounds such as bortezomib (Velcade, Millenium Pharmaceuticals)149, upregulation of proteasomal degradation by a small molecule has only recently been reported150. It will be interesting to follow the further development of such compounds and monitor their effects in models of HD and AD, as they are valuable tools for the further validation of the UPS as a therapeutic target.
Lysosomal proteases as therapeutic targets in AD and HD
In addition to the UPS, macroautophagy is an important pathway for the elimination of toxic or misfolded proteins. Protein degradation in the autophagic pathway takes place upon fusion of the autophagosome with lysosomes, and these vesicles are known sites of pathology in AD and HD (Figure 5). Aβ can be generated from APP in autophagic vesicles through cleavage by γ-secretase, and these vesicles accumulate in dystrophic neurites in AD patients and mouse models151. PSEN1 is not only an essential part of the γ-secretase complex in lysosomes, but also necessary to maintain an acidic pH in these vesicles, which in turn is required to guarantee the degradation of protein cargo152. PSEN1 mutations that are linked to FAD impair this function, leading to the accumulation of vesicles that contain Aβ152.
Figure 5. Therapeutic targets in the autophagy pathway.
Upregulation of autophagy is an efficient way to clear disease proteins in AD and HD, and therapeutic interventions have been tested against a variety of different targets. The balance between inhibition of autophagy by Akt and activation by AMPK has been shifted through treatment with resveratrol analogs, which increase AMPK activity. Inhibition of GSK3β through Lithium prevents excessive Tau phosphorylation and allows pro-survival CREB signalling (see above), autophagy is increased through inhibition of the IMPase pathway. Inhibition of mTOR with rapamycin and analog compounds lifts the block on autophagosome formation and is beneficial in both AD and HD mouse models. An mTOR independent pathway activating autophagy can be triggered by treatment with Valproate and is mediated a decrease in Inositol and IP3 levels. The Beclin 1-PI3K type III complex promotes autophagosome formation and loss or inhibition of either protein is detrimental in mouse and cell culture models of AD and HD. Autophagosomes not only engulf aggregates of disease proteins present in the cytoplasm but are also capable of generating Aβ through γ-secretase cleavage. Upon fusion with the lysosome, the autophagosome becomes acidic and lysosomal proteases such as the cathepsins degrade the disease proteins. Deletion of the endogenous cathepsin inhibitor cystatin B is beneficial in an AD mouse model. Furthermore mHtt interferes with lysosome function by impairing vesicle transport from the Golgi, resulting in a decrease of proteases in the lysosome. PSEN1 deletion or mutations interfere with the acidification of the lysosome, thus reducing the activity of lysosomal proteases and rendering autophagic protein degradation less efficient.
Proteases of the cathepsin family finally break down cargo proteins after fusion of the autophagosome with the lysosome (Figure 5). The genetic deletion of an endogenous inhibitor of cathepsins, cystatin B, ameliorates the memory deficits and Aβ aggregate load in a mouse model of AD153, which indicates that lysosomal proteases are able to clear disease-causing proteins in AD and that their upregulation should be beneficial (Suppl Table S4). In HD, it was proposed that the expression of mHtt impairs vesicular transport from the Golgi to the lysosomes and thus leads to a reduction in lysosomal cathepsins154 (Figure 5), which would result in inefficient autophagy in HD. Cystatin B knockdown in models of HD and studies with cathepsin inhibitors in animal models for both AD and HD could help to further confirm and validate these therapeutic targets.
Targeting the PI3 kinase and beclin pathway
The Beclin1/PI3 kinase type III (PI3K) complex influences autophagy through the modulation of autophagosome formation, maturation and degradation155 (Figure 5). In a mouse model of AD, loss of Beclin1 leads to the accumulation of Aß plaques and lysosomes as well as neuronal loss155. A reduction of levels of Beclin1 leads to the accumulation of mHtt and reduced the viability of animals in a mouse model of HD156. The autophagic response initiated by the exposure of cultured cells to Aß is mediated by PI3K157, and a similar induction of autophagy through PI3K occurs in cell culture models of HD158. On the other hand, blocking autophagy through the pharmacological inhibition of PI3K by 3-methyladenine increases the levels of mHtt and reduces viability159.
Preservation or upregulation of the function of PI3K and Beclin1, which normally declines with aging156, is therefore a promising strategy to increase the degradation of mHtt and Aß (Suppl Table S4), although pharmacological agents to achieve this effect are currently not available. Beclin1 gene transfer has been used successfully to activate autophagy in a mouse model of Parkinson’s disease, and similar strategies could be tested in models for AD and HD160.
Targeting mTOR to modulate autophagy in AD and HD
mTOR, the mammalian target of rapamycin, negatively regulates autophagy through the phosphorylation of its target proteins ULK1 and Atg13161, which prevents the formation of autophagosomes161 (Figure 5). The mTOR inhibitors rapamycin (sirolimus) and its analogs everolimus and temsirolimus increase the autophagic clearance of mHtt and Aß, which was associated with decreased toxicity in cellular models and rescue of different disease phenotypes in animal models of AD and HD162, 163 (Suppl Table S4). Everolimus (Afinitor, Novartis), temsirolimus (Torisel, Wyeth Pharmaceuticals) and sirolimus (Rapamune, Wyeth Pharmaceuticals) are FDA-approved drugs for the prevention of organ transplant rejection as well as for the treatment of different forms of cancer, and pre-clinical trials have been conducted for a number of autoimmune and infectious diseases (reviewed in164). However, haematological side-effects are common and often lead to the termination of treatment164, which makes the long-term use of these drugs for HD or AD unlikely.
An additional pathway for the induction of autophagy has been described, that is independent of mTOR and mediated through the inhibition of Inositol monophosphatase (IMPase) and the subsequent reduction in Inositol or inositol-3-phosphate (IP3) levels165. Therapeutically, this inhibition can be achieved through the treatment with lithium or valproate and is beneficial in Drosophila and Zebrafish models of HD165 (Figure 5). Lithium has additional effects on neurotrophic factors and tau phosphorylation, and while an initial short-term trial in AD patients did not result in altered levels of phospho-Tau166, a potential disease-modifying effect has been shown recently in a more long-term study55. Another recently described pharmacophore is the so-called small molecule enhancers of rapamycin (SMER) series of compounds that induce neuronal autophagy independent of mTOR167. SMERs induce autophagy in mammalian cells independently or downstream of mTOR and were protective in a Drosophila model for HD168.
In summary, the autophagic pathway contains a number of well validated targets with TV scores of 3 or higher that are involved in the pathogenesis of AD and HD. While the inhibition of mTOR has been studied in great detail, the application of currently available compounds is hampered by severe side-effects. If no alternatives can be developed, other avenues such as the stabilization of Beclin1 could be pursued to increase the autophagic clearance of disease-causing proteins in AD and HD.
Moving towards clinical trials- the power of predictive testing
Clinical trials in AD have often been criticized for only including patients in late stages of the disease, where substantial neurodegeneration has already occurred, or for including patients that suffer from forms of dementia other than AD169. The development of biomarkers to reliably diagnose AD in early disease stages and monitor disease progression has made significant progress, and CSF levels of total Tau, phosphorylated Tau and Aß yield reasonable sensitivity (95%) and specificity (83%)170. However, CSF studies still show considerable variability and the accurate identification of patients with early stage AD is difficult171.
The monogenic cause for HD, however, makes it possible to determine the effect of therapeutic interventions on the onset of symptoms in patients that do not yet show significant neuronal loss. A more homogeneous disease phenotype, clinical endpoints and longitudinal markers of disease progression that are currently being established allow for the assessment of disease-modifying effects of therapeutics in a timeframe of 1-2 years16 in HD. Effective compounds could then be prioritized for clinical trials in AD patients, whereas drugs that do not show effects in HD patients could be ranked lower in priority. However, the challenge for clinical trials in AD remains in the development of reliable biomarkers that correlate with early stages of the disease. Currently, interventions are being tested in patients with mild cognitive impairment and their progression to clinically manifest AD is being monitored55. Therapeutics that have been beneficial in clinical trials of patients with HD and for which early proof of concept for the modulation of pathways perturbed in both disorders has thus been shown, could similarly be assessed.
Despite the numerous common therapeutic targets identified for AD and HD, the question remains why the phenotype of AD and HD is largely different, with different brain areas affected, different sets of proteins misfolded and accumulated as well as a different clinical appearance. The most obvious explanation is the difference in underlying genetic mutations: HD is defined by the expression of mHtt protein, whereas AD is largely of unknown etiology or in rare cases caused by mutations in APP, PSEN1 or PSEN2. Differences in expression patterns as well as alterations in protein-protein interactions of these critical proteins could account for the susceptibility of distinct neuronal subpopulations, which are in turn correlated with the major clinical symptoms of chorea in HD or dementia in AD. While mHtt, APP, PSEN1 and PSEN2 are therapeutic targets in their own right, their dysfunction triggers similar pathogenic pathways that are shared between AD and HD and that comprise promising therapeutic targets. The modulation of these targets will likely be possible with the same therapeutic strategy for both diseases, and also similar side-effects from these interventions would be expected for both patient populations.
Overall, the convergence of pathways that are shared between AD and HD should encourage greater interaction between scientists working in each of these diseases as lessons from one may have significance for the other.
Supplementary Material
Table 3. Comparison of TV scores for top targets in AD and HD.
| Target | TV score (AD) | TV score (HD) |
|---|---|---|
| NR2B | 6.0 | 4.0 |
| BDNF | 4.5 | 4.0 |
| NGF | 4.5 | 4.0 |
| HDACs | 4.0 | 4.0 |
| Kynurenine 3-Monooxygenase | 4.0 | 4.0 |
| HAP1 | 2.5 | 3.5 |
| Caspase-6 | 3.5 | 3.5 |
| Caspase-9 | 3.5 | 2.5 |
| Bcl-2 | 3.5 | 3.5 |
| GSK3β | 3.5 | 3.0 |
| Hsps | 3.5 | 3.5 |
| Cystatin B | 3.5 | not characterized |
| Cathepsins | 3.5 | 2.0 |
| Beclin 1 | 3.5 | 3.0 |
| mTOR | 3.5 | 3.5 |
| IAPs | 2.0 | 3.0 |
| HIP14 | not characterized | 3.0 |
| misfolded proteins | 3.0 | 3.0 |
| PI3 kinase type III | 2.0 | 3.0 |
References
- 1.Alzheimer’s Association. Thies W, Bleiler L. Alzheimer’s disease facts and figures. Alzheimers Dement. 2011;7:208–44. doi: 10.1016/j.jalz.2011.02.004. 2011. [DOI] [PubMed] [Google Scholar]
- 2.Novak MJ, Tabrizi SJ. Huntington’s disease. BMJ. 2010;340:c3109. doi: 10.1136/bmj.c3109. [DOI] [PubMed] [Google Scholar]
- 3.Mihaescu R, et al. Translational research in genomics of Alzheimer’s disease: a review of current practice and future perspectives. J Alzheimers Dis. 2010;20:967–80. doi: 10.3233/JAD-2010-1410. [DOI] [PubMed] [Google Scholar]
- 4.Zetzsche T, Rujescu D, Hardy J, Hampel H. Advances and perspectives from genetic research: development of biological markers in Alzheimer’s disease. Expert Rev Mol Diagn. 2010;10:667–90. doi: 10.1586/erm.10.48. [DOI] [PubMed] [Google Scholar]
- 5.Bi X. Alzheimer disease: update on basic mechanisms. J Am Osteopath Assoc. 2010;110:S3–9. [PubMed] [Google Scholar]
- 6.Crews L, Masliah E. Molecular mechanisms of neurodegeneration in Alzheimer’s disease. Hum Mol Genet. 2010;19:R12–20. doi: 10.1093/hmg/ddq160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzherimer’s disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov. 2011 doi: 10.1038/nrd3505. AOP, published online 19 August 2011; doi:10.1038/nrd3505. [DOI] [PubMed] [Google Scholar]
- 8.Southwell AL, Patterson PH. Gene therapy in mouse models of huntington disease. Neuroscientist. 2011;17:153–62. doi: 10.1177/1073858410386236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ehrnhoefer DE, Sutton L, Hayden MR. Small Changes, Big Impact: Posttranslational Modifications and Function of Huntingtin in Huntington Disease. Neuroscientist. 2011 doi: 10.1177/1073858410390378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Albin RL, et al. Abnormalities of striatal projection neurons and N-methyl-D-aspartate receptors in presymptomatic Huntington’s disease. N Engl J Med. 1990;322:1293–8. doi: 10.1056/NEJM199005033221807. [DOI] [PubMed] [Google Scholar]
- 11.Price JL, et al. Neuron number in the entorhinal cortex and CA1 in preclinical Alzheimer disease. Arch Neurol. 2001;58:1395–402. doi: 10.1001/archneur.58.9.1395. [DOI] [PubMed] [Google Scholar]
- 12.Tagliavini F, Pilleri G. Neuronal counts in basal nucleus of Meynert in Alzheimer disease and in simple senile dementia. Lancet. 1983;1:469–70. doi: 10.1016/s0140-6736(83)91462-9. [DOI] [PubMed] [Google Scholar]
- 13.Mouton PR, Martin LJ, Calhoun ME, Dal Forno G, Price DL. Cognitive decline strongly correlates with cortical atrophy in Alzheimer’s dementia. Neurobiol Aging. 1998;19:371–7. doi: 10.1016/s0197-4580(98)00080-3. [DOI] [PubMed] [Google Scholar]
- 14.Zuccato C, Valenza M, Cattaneo E. Molecular mechanisms and potential therapeutical targets in Huntington’s disease. Physiol Rev. 2010;90:905–81. doi: 10.1152/physrev.00041.2009. [DOI] [PubMed] [Google Scholar]
- 15.Kwak SP, Wang JKT, Howland DS. In: Neurobiology of Huntington’s Disease: Applications to Drug Discovery. Lo DC, Hughes RE, editors. CRC Press; Boca Raton, FL: 2011. [PubMed] [Google Scholar]
- 16.Tabrizi SJ, et al. Biological and clinical changes in premanifest and early stage Huntington’s disease in the TRACK-HD study: the 12-month longitudinal analysis. Lancet Neurol. 2011;10:31–42. doi: 10.1016/S1474-4422(10)70276-3. [DOI] [PubMed] [Google Scholar]
- 17.Ehrnhoefer DE, Butland SL, Pouladi MA, Hayden MR. Mouse models of Huntington disease: variations on a theme. Dis Model Mech. 2009;2:123–9. doi: 10.1242/dmm.002451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Fernandes HB, Raymond LA. NMDA Receptors and Huntington’s Disease. 2009. [Google Scholar]
- 19.Hoe HS, et al. The effects of amyloid precursor protein on postsynaptic composition and activity. J Biol Chem. 2009;284:8495–506. doi: 10.1074/jbc.M900141200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Song C, Zhang Y, Parsons CG, Liu YF. Expression of polyglutamine-expanded huntingtin induces tyrosine phosphorylation of N-methyl-D-aspartate receptors. J Biol Chem. 2003;278:33364–9. doi: 10.1074/jbc.M304240200. [DOI] [PubMed] [Google Scholar]
- 21.Roberson ED, et al. Amyloid-beta/Fyn-induced synaptic, network, and cognitive impairments depend on tau levels in multiple mouse models of Alzheimer’s disease. J Neurosci. 2011;31:700–11. doi: 10.1523/JNEUROSCI.4152-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ittner LM, et al. Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer’s disease mouse models. Cell. 2010;142:387–97. doi: 10.1016/j.cell.2010.06.036. [DOI] [PubMed] [Google Scholar]
- 23.Hu JL, Liu G, Li YC, Gao WJ, Huang YQ. Dopamine D1 receptor-mediated NMDA receptor insertion depends on Fyn but not Src kinase pathway in prefrontal cortical neurons. Mol Brain. 2010;3:20. doi: 10.1186/1756-6606-3-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Okamoto S, et al. Balance between synaptic versus extrasynaptic NMDA receptor activity influences inclusions and neurotoxicity of mutant huntingtin. Nat Med. 2009;15:1407–13. doi: 10.1038/nm.2056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Sydow A, et al. Tau-induced defects in synaptic plasticity, learning, and memory are reversible in transgenic mice after switching off the toxic Tau mutant. J Neurosci. 2011;31:2511–25. doi: 10.1523/JNEUROSCI.5245-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kung VW, Hassam R, Morton AJ, Jones S. Dopamine-dependent long term potentiation in the dorsal striatum is reduced in the R6/2 mouse model of Huntington’s disease. Neuroscience. 2007;146:1571–80. doi: 10.1016/j.neuroscience.2007.03.036. [DOI] [PubMed] [Google Scholar]
- 27.Bordji K, Becerril-Ortega J, Nicole O, Buisson A. Activation of extrasynaptic, but not synaptic, NMDA receptors modifies amyloid precursor protein expression pattern and increases amyloid-ss production. J Neurosci. 2010;30:15927–42. doi: 10.1523/JNEUROSCI.3021-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Li S, et al. Soluble A{beta} Oligomers Inhibit Long-Term Potentiation through a Mechanism Involving Excessive Activation of Extrasynaptic NR2B-Containing NMDA Receptors. J Neurosci. 2011;31:6627–38. doi: 10.1523/JNEUROSCI.0203-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Milnerwood AJ, et al. Early increase in extrasynaptic NMDA receptor signaling and expression contributes to phenotype onset in Huntington’s disease mice. Neuron. 2010;65:178–90. doi: 10.1016/j.neuron.2010.01.008. [DOI] [PubMed] [Google Scholar]
- 30.Filali M, Lalonde R, Rivest S. Subchronic memantine administration on spatial learning, exploratory activity, and nest-building in an APP/PS1 mouse model of Alzheimer’s disease. Neuropharmacology. 2011;60:930–6. doi: 10.1016/j.neuropharm.2011.01.035. [DOI] [PubMed] [Google Scholar]
- 31.Schulz JB, et al. Sustained effects of once-daily memantine treatment on cognition and functional communication skills in patients with moderate to severe Alzheimer’s disease: results of a 16-week open-label trial. J Alzheimers Dis. 2011;25:463–75. doi: 10.3233/JAD-2011-101929. [DOI] [PubMed] [Google Scholar]
- 32.Beister A, et al. The N-methyl-D-aspartate antagonist memantine retards progression of Huntington’s disease. J Neural Transm Suppl. 2004:117–22. doi: 10.1007/978-3-7091-0579-5_14. [DOI] [PubMed] [Google Scholar]
- 33.Hjermind LE, Law I, Jonch A, Stokholm J, Nielsen JE. Huntington’s Disease: Effect of Memantine on FDG-PET Brain Metabolism? J Neuropsychiatry Clin Neurosci. 2011;23:206–10. doi: 10.1176/jnp.23.2.jnp206. [DOI] [PubMed] [Google Scholar]
- 34.Ondo WG, Mejia NI, Hunter CB. A pilot study of the clinical efficacy and safety of memantine for Huntington’s disease. Parkinsonism Relat Disord. 2007;13:453–4. doi: 10.1016/j.parkreldis.2006.08.005. [DOI] [PubMed] [Google Scholar]
- 35.Zwilling D, et al. Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell. 2011;145:863–74. doi: 10.1016/j.cell.2011.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hennigan A, O’Callaghan RM, Kelly AM. Neurotrophins and their receptors: roles in plasticity, neurodegeneration and neuroprotection. Biochem Soc Trans. 2007;35:424–7. doi: 10.1042/BST0350424. [DOI] [PubMed] [Google Scholar]
- 37.Ferrer I, Goutan E, Marin C, Rey MJ, Ribalta T. Brain-derived neurotrophic factor in Huntington disease. Brain Res. 2000;866:257–61. doi: 10.1016/s0006-8993(00)02237-x. [DOI] [PubMed] [Google Scholar]
- 38.Connor B, et al. Brain-derived neurotrophic factor is reduced in Alzheimer’s disease. Brain Res Mol Brain Res. 1997;49:71–81. doi: 10.1016/s0169-328x(97)00125-3. [DOI] [PubMed] [Google Scholar]
- 39.Voineskos AN, et al. The brain-derived neurotrophic factor Val66Met polymorphism and prediction of neural risk for Alzheimer disease. Arch Gen Psychiatry. 2011;68:198–206. doi: 10.1001/archgenpsychiatry.2010.194. [DOI] [PubMed] [Google Scholar]
- 40.Bian JT, Zhang JW, Zhang ZX, Zhao HL. Association analysis of brain-derived neurotrophic factor (BDNF) gene 196 A/G polymorphism with Alzheimer’s disease (AD) in mainland Chinese. Neurosci Lett. 2005;387:11–6. doi: 10.1016/j.neulet.2005.07.009. [DOI] [PubMed] [Google Scholar]
- 41.Ventriglia M, et al. Association between the BDNF 196 A/G polymorphism and sporadic Alzheimer’s disease. Mol Psychiatry. 2002;7:136–7. doi: 10.1038/sj.mp.4000952. [DOI] [PubMed] [Google Scholar]
- 42.Desai P, Nebes R, DeKosky ST, Kamboh MI. Investigation of the effect of brain-derived neurotrophic factor (BDNF) polymorphisms on the risk of late-onset Alzheimer’s disease (AD) and quantitative measures of AD progression. Neurosci Lett. 2005;379:229–34. doi: 10.1016/j.neulet.2005.01.008. [DOI] [PubMed] [Google Scholar]
- 43.Bagnoli S, et al. Brain-derived neurotrophic factor genetic variants are not susceptibility factors to Alzheimer’s disease in Italy. Ann Neurol. 2004;55:447–8. doi: 10.1002/ana.10842. [DOI] [PubMed] [Google Scholar]
- 44.Combarros O, Infante J, Llorca J, Berciano J. Polymorphism at codon 66 of the brain-derived neurotrophic factor gene is not associated with sporadic Alzheimer’s disease. Dement Geriatr Cogn Disord. 2004;18:55–8. doi: 10.1159/000077736. [DOI] [PubMed] [Google Scholar]
- 45.Alberch J, et al. Association between BDNF Val66Met polymorphism and age at onset in Huntington disease. Neurology. 2005;65:964–5. doi: 10.1212/01.wnl.0000175977.57661.b1. [DOI] [PubMed] [Google Scholar]
- 46.Kishikawa S, et al. Brain-derived neurotrophic factor does not influence age at neurologic onset of Huntington’s disease. Neurobiol Dis. 2006;24:280–5. doi: 10.1016/j.nbd.2006.07.008. [DOI] [PubMed] [Google Scholar]
- 47.Neeper SA, Gomez-Pinilla F, Choi J, Cotman C. Exercise and brain neurotrophins. Nature. 1995;373:109. doi: 10.1038/373109a0. [DOI] [PubMed] [Google Scholar]
- 48.Liu HL, Zhao G, Cai K, Zhao HH, Shi LD. Treadmill exercise prevents decline in spatial learning and memory in APP/PS1 transgenic mice through improvement of hippocampal long-term potentiation. Behav Brain Res. 2011;218:308–14. doi: 10.1016/j.bbr.2010.12.030. [DOI] [PubMed] [Google Scholar]
- 49.Arancibia S, et al. Protective effect of BDNF against beta-amyloid induced neurotoxicity in vitro and in vivo in rats. Neurobiol Dis. 2008;31:316–26. doi: 10.1016/j.nbd.2008.05.012. [DOI] [PubMed] [Google Scholar]
- 50.Dey ND, et al. Genetically engineered mesenchymal stem cells reduce behavioral deficits in the YAC 128 mouse model of Huntington’s disease. Behav Brain Res. 2010;214:193–200. doi: 10.1016/j.bbr.2010.05.023. [DOI] [PubMed] [Google Scholar]
- 51.Blurton-Jones M, et al. Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc Natl Acad Sci U S A. 2009;106:13594–9. doi: 10.1073/pnas.0901402106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Nagahara AH, et al. Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer’s disease. Nat Med. 2009;15:331–7. doi: 10.1038/nm.1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Simmons DA, et al. Up-regulating BDNF with an ampakine rescues synaptic plasticity and memory in Huntington’s disease knockin mice. Proc Natl Acad Sci U S A. 2009;106:4906–11. doi: 10.1073/pnas.0811228106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Leyhe T, et al. Increase of BDNF serum concentration in lithium treated patients with early Alzheimer’s disease. J Alzheimers Dis. 2009;16:649–56. doi: 10.3233/JAD-2009-1004. [DOI] [PubMed] [Google Scholar]
- 55.Forlenza OV, et al. Disease-modifying properties of long-term lithium treatment for amnestic mild cognitive impairment: randomised controlled trial. Br J Psychiatry. 2011;198:351–6. doi: 10.1192/bjp.bp.110.080044. [DOI] [PubMed] [Google Scholar]
- 56.Toledo EM, Inestrosa NC. Activation of Wnt signaling by lithium and rosiglitazone reduced spatial memory impairment and neurodegeneration in brains of an APPswe/PSEN1DeltaE9 mouse model of Alzheimer’s disease. Mol Psychiatry. 2010;15:272–85. 228. doi: 10.1038/mp.2009.72. [DOI] [PubMed] [Google Scholar]
- 57.Carmichael J, Sugars KL, Bao YP, Rubinsztein DC. Glycogen synthase kinase-3beta inhibitors prevent cellular polyglutamine toxicity caused by the Huntington’s disease mutation. J Biol Chem. 2002;277:33791–8. doi: 10.1074/jbc.M204861200. [DOI] [PubMed] [Google Scholar]
- 58.Rockenstein E, et al. Neuroprotective effects of regulators of the glycogen synthase kinase-3beta signaling pathway in a transgenic model of Alzheimer’s disease are associated with reduced amyloid precursor protein phosphorylation. J Neurosci. 2007;27:1981–91. doi: 10.1523/JNEUROSCI.4321-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Quiroz JA, Machado-Vieira R, Zarate CA, Jr., Manji HK. Novel insights into lithium’s mechanism of action: neurotrophic and neuroprotective effects. Neuropsychobiology. 2010;62:50–60. doi: 10.1159/000314310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gauthier LR, et al. Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell. 2004;118:127–38. doi: 10.1016/j.cell.2004.06.018. [DOI] [PubMed] [Google Scholar]
- 61.Peethumnongsin E, et al. Convergence of presenilin- and tau-mediated pathways on axonal trafficking and neuronal function. J Neurosci. 2010;30:13409–18. doi: 10.1523/JNEUROSCI.1964-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Wu LL, Fan Y, Li S, Li XJ, Zhou XF. Huntingtin-associated protein-1 interacts with pro-brain-derived neurotrophic factor and mediates its transport and release. J Biol Chem. 2010;285:5614–23. doi: 10.1074/jbc.M109.073197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Harjes P, Wanker EE. The hunt for huntingtin function: interaction partners tell many different stories. Trends Biochem Sci. 2003;28:425–33. doi: 10.1016/S0968-0004(03)00168-3. [DOI] [PubMed] [Google Scholar]
- 64.Dixit R, Ross JL, Goldman YE, Holzbaur EL. Differential regulation of dynein and kinesin motor proteins by tau. Science. 2008;319:1086–9. doi: 10.1126/science.1152993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.McGuire JR, Rong J, Li SH, Li XJ. Interaction of Huntingtin-associated protein-1 with kinesin light chain: implications in intracellular trafficking in neurons. J Biol Chem. 2006;281:3552–9. doi: 10.1074/jbc.M509806200. [DOI] [PubMed] [Google Scholar]
- 66.Lazarov O, et al. Impairments in fast axonal transport and motor neuron deficits in transgenic mice expressing familial Alzheimer’s disease-linked mutant presenilin 1. J Neurosci. 2007;27:7011–20. doi: 10.1523/JNEUROSCI.4272-06.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Sereno L, et al. A novel GSK-3beta inhibitor reduces Alzheimer’s pathology and rescues neuronal loss in vivo. Neurobiol Dis. 2009;35:359–67. doi: 10.1016/j.nbd.2009.05.025. [DOI] [PubMed] [Google Scholar]
- 68.Marks PA. The clinical development of histone deacetylase inhibitors as targeted anticancer drugs. Expert Opin Investig Drugs. 2010;19:1049–66. doi: 10.1517/13543784.2010.510514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Dompierre JP, et al. Histone deacetylase 6 inhibition compensates for the transport deficit in Huntington’s disease by increasing tubulin acetylation. J Neurosci. 2007;27:3571–83. doi: 10.1523/JNEUROSCI.0037-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Butler D, Bendiske J, Michaelis ML, Karanian DA, Bahr BA. Microtubule-stabilizing agent prevents protein accumulation-induced loss of synaptic markers. Eur J Pharmacol. 2007;562:20–7. doi: 10.1016/j.ejphar.2007.01.053. [DOI] [PubMed] [Google Scholar]
- 71.Brunden KR, et al. The characterization of microtubule-stabilizing drugs as possible therapeutic agents for Alzheimer’s disease and related tauopathies. Pharmacol Res. 2011;63:341–51. doi: 10.1016/j.phrs.2010.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Iwase H, et al. A Phase II Multi-Center Study of Triple Therapy with Paclitaxel, S-1 and Cisplatin in Patients with Advanced Gastric Cancer. Oncology. 2011;80:76–83. doi: 10.1159/000328746. [DOI] [PubMed] [Google Scholar]
- 73.Schulte-Herbruggen O, Jockers-Scherubl MC, Hellweg R. Neurotrophins: from pathophysiology to treatment in Alzheimer’s disease. Curr Alzheimer Res. 2008;5:38–44. doi: 10.2174/156720508783884620. [DOI] [PubMed] [Google Scholar]
- 74.Gu H, Long D, Song C, Li X. Recombinant human NGF-loaded microspheres promote survival of basal forebrain cholinergic neurons and improve memory impairments of spatial learning in the rat model of Alzheimer’s disease with fimbria-fornix lesion. Neurosci Lett. 2009;453:204–9. doi: 10.1016/j.neulet.2009.02.027. [DOI] [PubMed] [Google Scholar]
- 75.Menei P, et al. Intracerebral implantation of NGF-releasing biodegradable microspheres protects striatum against excitotoxic damage. Exp Neurol. 2000;161:259–72. doi: 10.1006/exnr.1999.7253. [DOI] [PubMed] [Google Scholar]
- 76.Tuszynski MH, et al. A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med. 2005;11:551–5. doi: 10.1038/nm1239. [DOI] [PubMed] [Google Scholar]
- 77.Covaceuszach S, et al. Development of a non invasive NGF-based therapy for Alzheimer’s disease. Curr Alzheimer Res. 2009;6:158–70. doi: 10.2174/156720509787602870. [DOI] [PubMed] [Google Scholar]
- 78.Zuccato C, et al. Systematic assessment of BDNF and its receptor levels in human cortices affected by Huntington’s disease. Brain Pathol. 2008;18:225–38. doi: 10.1111/j.1750-3639.2007.00111.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Troy CM, Friedman JE, Friedman WJ. Mechanisms of p75-mediated death of hippocampal neurons. Role of caspases. J Biol Chem. 2002;277:34295–302. doi: 10.1074/jbc.M205167200. [DOI] [PubMed] [Google Scholar]
- 80.Peineau S, et al. LTP inhibits LTD in the hippocampus via regulation of GSK3beta. Neuron. 2007;53:703–17. doi: 10.1016/j.neuron.2007.01.029. [DOI] [PubMed] [Google Scholar]
- 81.Fombonne J, Rabizadeh S, Banwait S, Mehlen P, Bredesen DE. Selective vulnerability in Alzheimer’s disease: amyloid precursor protein and p75(NTR) interaction. Ann Neurol. 2009;65:294–303. doi: 10.1002/ana.21578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Jeanneteau F, Garabedian MJ, Chao MV. Activation of Trk neurotrophin receptors by glucocorticoids provides a neuroprotective effect. Proc Natl Acad Sci U S A. 2008;105:4862–7. doi: 10.1073/pnas.0709102105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Moffitt KL, Martin SL, Walker B. From sentencing to execution--the processes of apoptosis. J Pharm Pharmacol. 2010;62:547–62. doi: 10.1211/jpp.62.05.0001. [DOI] [PubMed] [Google Scholar]
- 84.Feinstein-Rotkopf Y, Arama E. Can’t live without them, can live with them: roles of caspases during vital cellular processes. Apoptosis. 2009;14:980–95. doi: 10.1007/s10495-009-0346-6. [DOI] [PubMed] [Google Scholar]
- 85.Nikolaev A, McLaughlin T, O’Leary DD, Tessier-Lavigne M. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature. 2009;457:981–9. doi: 10.1038/nature07767. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 86.Park KJ, Grosso CA, Aubert I, Kaplan DR, Miller FD. p75NTR-dependent, myelin-mediated axonal degeneration regulates neural connectivity in the adult brain. Nat Neurosci. 2010;13:559–66. doi: 10.1038/nn.2513. [DOI] [PubMed] [Google Scholar]
- 87.Schoenmann Z, et al. Axonal degeneration is regulated by the apoptotic machinery or a NAD+− sensitive pathway in insects and mammals. J Neurosci. 2010;30:6375–86. doi: 10.1523/JNEUROSCI.0922-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Li Z, et al. Caspase-3 activation via mitochondria is required for long-term depression and AMPA receptor internalization. Cell. 2010;141:859–71. doi: 10.1016/j.cell.2010.03.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Albrecht S, et al. Activation of caspase-6 in aging and mild cognitive impairment. Am J Pathol. 2007;170:1200–9. doi: 10.2353/ajpath.2007.060974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Graham RK, et al. Cleavage at the 586 amino acid caspase-6 site in mutant huntingtin influences caspase-6 activation in vivo. J Neurosci. 2010;30:15019–29. doi: 10.1523/JNEUROSCI.2071-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Guo H, et al. Active caspase-6 and caspase-6-cleaved tau in neuropil threads, neuritic plaques, and neurofibrillary tangles of Alzheimer’s disease. Am J Pathol. 2004;165:523–31. doi: 10.1016/S0002-9440(10)63317-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Halawani D, et al. Identification of Caspase-6-mediated processing of the valosin containing protein (p97) in Alzheimer’s disease: a novel link to dysfunction in ubiquitin proteasome system-mediated protein degradation. J Neurosci. 2010;30:6132–42. doi: 10.1523/JNEUROSCI.5874-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Galvan V, et al. Reversal of Alzheimer’s-like pathology and behavior in human APP transgenic mice by mutation of Asp664. Proc Natl Acad Sci U S A. 2006;103:7130–5. doi: 10.1073/pnas.0509695103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Graham RK, et al. Cleavage at the caspase-6 site is required for neuronal dysfunction and degeneration due to mutant huntingtin. Cell. 2006;125:1179–91. doi: 10.1016/j.cell.2006.04.026. [DOI] [PubMed] [Google Scholar]
- 95.Ghavami S, et al. Apoptosis and cancer: mutations within caspase genes. J Med Genet. 2009;46:497–510. doi: 10.1136/jmg.2009.066944. [DOI] [PubMed] [Google Scholar]
- 96.Philchenkov A, Zavelevich M, Kroczak TJ, Los M. Caspases and cancer: mechanisms of inactivation and new treatment modalities. Exp Oncol. 2004;26:82–97. [PubMed] [Google Scholar]
- 97.Shirendeb U, et al. Abnormal mitochondrial dynamics, mitochondrial loss and mutant huntingtin oligomers in Huntington’s disease: implications for selective neuronal damage. Hum Mol Genet. 2011;20:1438–55. doi: 10.1093/hmg/ddr024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Du H, et al. Early deficits in synaptic mitochondria in an Alzheimer’s disease mouse model. Proc Natl Acad Sci U S A. 2010;107:18670–5. doi: 10.1073/pnas.1006586107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Pagani L, Eckert A. Amyloid-Beta interaction with mitochondria. Int J Alzheimers Dis. 2011;2011:925050. doi: 10.4061/2011/925050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Pratico D. Evidence of oxidative stress in Alzheimer’s disease brain and antioxidant therapy: lights and shadows. Ann N Y Acad Sci. 2008;1147:70–8. doi: 10.1196/annals.1427.010. [DOI] [PubMed] [Google Scholar]
- 101.Stack EC, Matson WR, Ferrante RJ. Evidence of oxidant damage in Huntington’s disease: translational strategies using antioxidants. Ann N Y Acad Sci. 2008;1147:79–92. doi: 10.1196/annals.1427.008. [DOI] [PubMed] [Google Scholar]
- 102.Jo J, et al. Abeta(1-42) inhibition of LTP is mediated by a signaling pathway involving caspase-3, Akt1 and GSK-3beta. Nat Neurosci. 2011 doi: 10.1038/nn.2785. [DOI] [PubMed] [Google Scholar]
- 103.Gray DC, Mahrus S, Wells JA. Activation of specific apoptotic caspases with an engineered small-molecule-activated protease. Cell. 2010;142:637–46. doi: 10.1016/j.cell.2010.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Schettini G, Govoni S, Racchi M, Rodriguez G. Phosphorylation of APP-CTF-AICD domains and interaction with adaptor proteins: signal transduction and/or transcriptional role--relevance for Alzheimer pathology. J Neurochem. 2010;115:1299–308. doi: 10.1111/j.1471-4159.2010.07044.x. [DOI] [PubMed] [Google Scholar]
- 105.Taru H, Yoshikawa K, Suzuki T. Suppression of the caspase cleavage of beta-amyloid precursor protein by its cytoplasmic phosphorylation. FEBS Lett. 2004;567:248–52. doi: 10.1016/j.febslet.2004.04.077. [DOI] [PubMed] [Google Scholar]
- 106.Warby SC, et al. Phosphorylation of huntingtin reduces the accumulation of its nuclear fragments. Mol Cell Neurosci. 2009;40:121–7. doi: 10.1016/j.mcn.2008.09.007. [DOI] [PubMed] [Google Scholar]
- 107.Humbert S, et al. The IGF-1/Akt pathway is neuroprotective in Huntington’s disease and involves Huntingtin phosphorylation by Akt. Dev Cell. 2002;2:831–7. doi: 10.1016/s1534-5807(02)00188-0. [DOI] [PubMed] [Google Scholar]
- 108.Metzler M, et al. Phosphorylation of huntingtin at Ser421 in YAC128 neurons is associated with protection of YAC128 neurons from NMDA-mediated excitotoxicity and is modulated by PP1 and PP2A. J Neurosci. 2010;30:14318–29. doi: 10.1523/JNEUROSCI.1589-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Phiel CJ, Wilson CA, Lee VM, Klein PS. GSK-3alpha regulates production of Alzheimer’s disease amyloid-beta peptides. Nature. 2003;423:435–9. doi: 10.1038/nature01640. [DOI] [PubMed] [Google Scholar]
- 110.Chiu CT, Liu G, Leeds P, Chuang DM. Combined Treatment with the Mood Stabilizers Lithium and Valproate Produces Multiple Beneficial Effects in Transgenic Mouse Models of Huntington’s Disease. Neuropsychopharmacology. 2011 doi: 10.1038/npp.2011.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Valencia A, et al. Mutant huntingtin and glycogen synthase kinase 3-beta accumulate in neuronal lipid rafts of a presymptomatic knock-in mouse model of Huntington’s disease. J Neurosci Res. 2010;88:179–90. doi: 10.1002/jnr.22184. [DOI] [PubMed] [Google Scholar]
- 112.Qing H, et al. Valproic acid inhibits Abeta production, neuritic plaque formation, and behavioral deficits in Alzheimer’s disease mouse models. J Exp Med. 2008;205:2781–9. doi: 10.1084/jem.20081588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Gottlicher M. Valproic acid: an old drug newly discovered as inhibitor of histone deacetylases. Ann Hematol. 2004;83(Suppl 1):S91–2. doi: 10.1007/s00277-004-0850-2. [DOI] [PubMed] [Google Scholar]
- 114.Herrmann N, Lanctot KL, Rothenburg LS, Eryavec G. A placebo-controlled trial of valproate for agitation and aggression in Alzheimer’s disease. Dement Geriatr Cogn Disord. 2007;23:116–9. doi: 10.1159/000097757. [DOI] [PubMed] [Google Scholar]
- 115.Fardilha M, Esteves SL, Korrodi-Gregorio L, da Cruz e Silva OA, da Cruz e Silva FF. The physiological relevance of protein phosphatase 1 and its interacting proteins to health and disease. Curr Med Chem. 2010;17:3996–4017. doi: 10.2174/092986710793205363. [DOI] [PubMed] [Google Scholar]
- 116.Cheng H, et al. S-palmitoylation of gamma-secretase subunits nicastrin and APH-1. J Biol Chem. 2009;284:1373–84. doi: 10.1074/jbc.M806380200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Huang K, et al. Neuronal palmitoyl acyl transferases exhibit distinct substrate specificity. FASEB J. 2009;23:2605–15. doi: 10.1096/fj.08-127399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Yanai A, et al. Palmitoylation of huntingtin by HIP14 is essential for its trafficking and function. Nat Neurosci. 2006;9:824–31. doi: 10.1038/nn1702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Benjannet S, et al. Post-translational processing of beta-secretase (beta-amyloid-converting enzyme) and its ectodomain shedding. The pro- and transmembrane/cytosolic domains affect its cellular activity and amyloid-beta production. J Biol Chem. 2001;276:10879–87. doi: 10.1074/jbc.M009899200. [DOI] [PubMed] [Google Scholar]
- 120.Fukata Y, Fukata M. Protein palmitoylation in neuronal development and synaptic plasticity. Nat Rev Neurosci. 2010;11:161–75. doi: 10.1038/nrn2788. [DOI] [PubMed] [Google Scholar]
- 121.Huang K, et al. Wild-type HTT modulates the enzymatic activity of the neuronal palmitoyl transferase HIP14. Hum Mol Genet. 2011;20:3356–65. doi: 10.1093/hmg/ddr242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Huang K, et al. Wildtype HTT modulates the enzymatic activity of the neuronal palmitoyl transferase HIP14. Human Molecular Genetics. doi: 10.1093/hmg/ddr242. under review. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Singaraja RR, et al. Altered palmitoylation and neuropathological deficits in mice lacking HIP14. Hum Mol Genet. 2011 doi: 10.1093/hmg/ddr308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Robakis NK. An Alzheimer’s disease hypothesis based on transcriptional dysregulation. Amyloid. 2003;10:80–5. doi: 10.3109/13506120309041729. [DOI] [PubMed] [Google Scholar]
- 125.Hathorn T, Snyder-Keller A, Messer A. Nicotinamide improves motor deficits and upregulates PGC-1alpha and BDNF gene expression in a mouse model of Huntington’s disease. Neurobiol Dis. 2011;41:43–50. doi: 10.1016/j.nbd.2010.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hockly E, et al. Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington’s disease. Proc Natl Acad Sci U S A. 2003;100:2041–6. doi: 10.1073/pnas.0437870100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ricobaraza A, et al. Phenylbutyrate ameliorates cognitive deficit and reduces tau pathology in an Alzheimer’s disease mouse model. Neuropsychopharmacology. 2009;34:1721–32. doi: 10.1038/npp.2008.229. [DOI] [PubMed] [Google Scholar]
- 128.Ross CA, Poirier MA. Protein aggregation and neurodegenerative disease. Nat Med. 2004;10(Suppl):S10–7. doi: 10.1038/nm1066. [DOI] [PubMed] [Google Scholar]
- 129.Greenwald J, Riek R. Biology of amyloid: structure, function, and regulation. Structure. 2010;18:1244–60. doi: 10.1016/j.str.2010.08.009. [DOI] [PubMed] [Google Scholar]
- 130.Perrin V, et al. Neuroprotection by Hsp104 and Hsp27 in lentiviral-based rat models of Huntington’s disease. Mol Ther. 2007;15:903–11. doi: 10.1038/mt.sj.6300141. [DOI] [PubMed] [Google Scholar]
- 131.Dedeoglu A, Ferrante RJ, Andreassen OA, Dillmann WH, Beal MF. Mice overexpressing 70-kDa heat shock protein show increased resistance to malonate and 3- nitropropionic acid. Exp Neurol. 2002;176:262–5. doi: 10.1006/exnr.2002.7933. [DOI] [PubMed] [Google Scholar]
- 132.Heiser V, et al. Inhibition of huntingtin fibrillogenesis by specific antibodies and small molecules: implications for Huntington’s disease therapy. Proc Natl Acad Sci U S A. 2000;97:6739–44. doi: 10.1073/pnas.110138997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Zhang X, et al. A potent small molecule inhibits polyglutamine aggregation in Huntington’s disease neurons and suppresses neurodegeneration in vivo. Proc Natl Acad Sci U S A. 2005;102:892–7. doi: 10.1073/pnas.0408936102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Ehrnhoefer DE, et al. EGCG redirects amyloidogenic polypeptides into unstructured, off- pathway oligomers. Nat Struct Mol Biol. 2008;15:558–66. doi: 10.1038/nsmb.1437. [DOI] [PubMed] [Google Scholar]
- 135.Ehrnhoefer DE, et al. Green tea (-)-epigallocatechin-gallate modulates early events in huntingtin misfolding and reduces toxicity in Huntington’s disease models. Hum Mol Genet. 2006;15:2743–51. doi: 10.1093/hmg/ddl210. [DOI] [PubMed] [Google Scholar]
- 136.Scherzer-Attali R, et al. Complete phenotypic recovery of an Alzheimer’s disease model by a quinone-tryptophan hybrid aggregation inhibitor. PLoS One. 2010;5:e11101. doi: 10.1371/journal.pone.0011101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hamaguchi T, Ono K, Yamada M. Anti-amyloidogenic therapies: strategies for prevention and treatment of Alzheimer’s disease. Cell Mol Life Sci. 2006;63:1538–52. doi: 10.1007/s00018-005-5599-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Pickhardt M, et al. N-phenylamine derivatives as aggregation inhibitors in cell models of tauopathy. Curr Alzheimer Res. 2007;4:397–402. doi: 10.2174/156720507781788765. [DOI] [PubMed] [Google Scholar]
- 139.Mereles D, Buss SJ, Hardt SE, Hunstein W, Katus HA. Effects of the main green tea polyphenol epigallocatechin-3-gallate on cardiac involvement in patients with AL amyloidosis. Clin Res Cardiol. 2010;99:483–90. doi: 10.1007/s00392-010-0142-x. [DOI] [PubMed] [Google Scholar]
- 140.Rezai-Zadeh K, et al. Green tea epigallocatechin-3-gallate (EGCG) reduces beta-amyloid mediated cognitive impairment and modulates tau pathology in Alzheimer transgenic mice. Brain Res. 2008;1214:177–87. doi: 10.1016/j.brainres.2008.02.107. [DOI] [PubMed] [Google Scholar]
- 141.Smith A, et al. Nanolipidic particles improve the bioavailability and alpha-secretase inducing ability of epigallocatechin-3-gallate (EGCG) for the treatment of Alzheimer’s disease. Int J Pharm. 2010;389:207–12. doi: 10.1016/j.ijpharm.2010.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Arrasate M, Mitra S, Schweitzer ES, Segal MR, Finkbeiner S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature. 2004;431:805–10. doi: 10.1038/nature02998. [DOI] [PubMed] [Google Scholar]
- 143.Umeda T, et al. Intraneuronal amyloid beta oligomers cause cell death via endoplasmic reticulum stress, endosomal/lysosomal leakage, and mitochondrial dysfunction in vivo. J Neurosci Res. 2011 doi: 10.1002/jnr.22640. [DOI] [PubMed] [Google Scholar]
- 144.Davies JE, Sarkar S, Rubinsztein DC. The ubiquitin proteasome system in Huntington’s disease and the spinocerebellar ataxias. BMC Biochem. 2007;8(Suppl 1):S2. doi: 10.1186/1471-2091-8-S1-S2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Oddo S. The ubiquitin-proteasome system in Alzheimer’s disease. J Cell Mol Med. 2008;12:363–73. doi: 10.1111/j.1582-4934.2008.00276.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Mitra S, Tsvetkov AS, Finkbeiner S. Single neuron ubiquitin-proteasome dynamics accompanying inclusion body formation in huntington disease. J Biol Chem. 2009;284:4398–403. doi: 10.1074/jbc.M806269200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Bingol B, Sheng M. Deconstruction for reconstruction: the role of proteolysis in neural plasticity and disease. Neuron. 2011;69:22–32. doi: 10.1016/j.neuron.2010.11.006. [DOI] [PubMed] [Google Scholar]
- 148.Wang J, et al. Impaired ubiquitin-proteasome system activity in the synapses of Huntington’s disease mice. J Cell Biol. 2008;180:1177–89. doi: 10.1083/jcb.200709080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Bedford L, Lowe J, Dick LR, Mayer RJ, Brownell JE. Ubiquitin-like protein conjugation and the ubiquitin-proteasome system as drug targets. Nat Rev Drug Discov. 2011;10:29–46. doi: 10.1038/nrd3321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Lee BH, et al. Enhancement of proteasome activity by a small-molecule inhibitor of USP14. Nature. 2010;467:179–84. doi: 10.1038/nature09299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Yu WH, et al. Macroautophagy--a novel Beta-amyloid peptide-generating pathway activated in Alzheimer’s disease. J Cell Biol. 2005;171:87–98. doi: 10.1083/jcb.200505082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Lee JH, et al. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell. 2010;141:1146–58. doi: 10.1016/j.cell.2010.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Yang DS, et al. Reversal of autophagy dysfunction in the TgCRND8 mouse model of Alzheimer’s disease ameliorates amyloid pathologies and memory deficits. Brain. 2011;134:258–77. doi: 10.1093/brain/awq341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.del Toro D, et al. Mutant huntingtin impairs post-Golgi trafficking to lysosomes by delocalizing optineurin/Rab8 complex from the Golgi apparatus. Mol Biol Cell. 2009;20:1478–92. doi: 10.1091/mbc.E08-07-0726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Jaeger PA, Wyss-Coray T. Beclin 1 complex in autophagy and Alzheimer disease. Arch Neurol. 2010;67:1181–4. doi: 10.1001/archneurol.2010.258. [DOI] [PubMed] [Google Scholar]
- 156.Shibata M, et al. Regulation of intracellular accumulation of mutant Huntingtin by Beclin 1. J Biol Chem. 2006;281:14474–85. doi: 10.1074/jbc.M600364200. [DOI] [PubMed] [Google Scholar]
- 157.Lipinski MM, et al. Genome-wide analysis reveals mechanisms modulating autophagy in normal brain aging and in Alzheimer’s disease. Proc Natl Acad Sci U S A. 2010;107:14164–9. doi: 10.1073/pnas.1009485107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Yamamoto A, Cremona ML, Rothman JE. Autophagy-mediated clearance of huntingtin aggregates triggered by the insulin-signaling pathway. J Cell Biol. 2006;172:719–31. doi: 10.1083/jcb.200510065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Qin ZH, et al. Autophagy regulates the processing of amino terminal huntingtin fragments. Hum Mol Genet. 2003;12:3231–44. doi: 10.1093/hmg/ddg346. [DOI] [PubMed] [Google Scholar]
- 160.Spencer B, et al. Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in alpha-synuclein models of Parkinson’s and Lewy body diseases. J Neurosci. 2009;29:13578–88. doi: 10.1523/JNEUROSCI.4390-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Wong E, Cuervo AM. Autophagy gone awry in neurodegenerative diseases. Nat Neurosci. 2010;13:805–11. doi: 10.1038/nn.2575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Ravikumar B, et al. Inhibition of mTOR induces autophagy and reduces toxicity of polyglutamine expansions in fly and mouse models of Huntington disease. Nat Genet. 2004;36:585–95. doi: 10.1038/ng1362. [DOI] [PubMed] [Google Scholar]
- 163.Spilman P, et al. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer’s disease. PLoS One. 2010;5:e9979. doi: 10.1371/journal.pone.0009979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Sofroniadou S, Goldsmith D. Mammalian target of rapamycin (mTOR) inhibitors: potential uses and a review of haematological adverse effects. Drug Saf. 2011;34:97–115. doi: 10.2165/11585040-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 165.Williams A, et al. Novel targets for Huntington’s disease in an mTOR-independent autophagy pathway. Nat Chem Biol. 2008;4:295–305. doi: 10.1038/nchembio.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Hampel H, et al. Lithium trial in Alzheimer’s disease: a randomized, single-blind, placebo- controlled, multicenter 10-week study. J Clin Psychiatry. 2009;70:922–31. [PubMed] [Google Scholar]
- 167.Tsvetkov AS, et al. A small-molecule scaffold induces autophagy in primary neurons and protects against toxicity in a Huntington disease model. Proc Natl Acad Sci U S A. 2010;107:16982–7. doi: 10.1073/pnas.1004498107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Floto RA, et al. Small molecule enhancers of rapamycin-induced TOR inhibition promote autophagy, reduce toxicity in Huntington’s disease models and enhance killing of mycobacteria by macrophages. Autophagy. 2007;3:620–2. doi: 10.4161/auto.4898. [DOI] [PubMed] [Google Scholar]
- 169.Petersen RC. Early diagnosis of Alzheimer’s disease: is MCI too late? Curr Alzheimer Res. 2009;6:324–30. doi: 10.2174/156720509788929237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Hansson O, et al. Association between CSF biomarkers and incipient Alzheimer’s disease in patients with mild cognitive impairment: a follow-up study. Lancet Neurol. 2006;5:228–34. doi: 10.1016/S1474-4422(06)70355-6. [DOI] [PubMed] [Google Scholar]
- 171.Ballard C, et al. Alzheimer’s disease. Lancet. 2011;377:1019–31. doi: 10.1016/S0140-6736(10)61349-9. [DOI] [PubMed] [Google Scholar]
- 172.Calissano P, Matrone C, Amadoro G. Nerve growth factor as a paradigm of neurotrophins related to Alzheimer’s disease. Dev Neurobiol. 2010;70:372–83. doi: 10.1002/dneu.20759. [DOI] [PubMed] [Google Scholar]
- 173.Murer MG, et al. An immunohistochemical study of the distribution of brain-derived neurotrophic factor in the adult human brain, with particular reference to Alzheimer’s disease. Neuroscience. 1999;88:1015–32. doi: 10.1016/s0306-4522(98)00219-x. [DOI] [PubMed] [Google Scholar]
- 174.Colucci-D’Amato L, Perrone-Capano C, di Porzio U. Chronic activation of ERK and neurodegenerative diseases. Bioessays. 2003;25:1085–95. doi: 10.1002/bies.10355. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.





