Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Feb 3.
Published in final edited form as: Exp Neurol. 2012 Jun 4;237(1):90–95. doi: 10.1016/j.expneurol.2012.05.018

TDP-43: A New Player on the AD Field?

Katherine L Youmans 1, Benjamin Wolozin 1,2,3,*
PMCID: PMC3910368  NIHMSID: NIHMS546624  PMID: 22691390

Introduction

Alzheimer’s disease accounts for an estimated 60–80% of dementia cases, and is the 5th leading cause of death for people aged 65 and older (Heron, 2008). The cognitive decline and amnestic dementia characteristic of the disease result from brain region-specific neuronal dysfunction and death, leading to memory loss, confusion, and changes in behavior, mood and personality. Ultimately, AD patients require help with daily tasks such as eating, bathing and communicating, primarily from unpaid caregivers including family members and friends. In 2009, the contribution of these unpaid caregivers to our nation was valued at $144 billion. As a result of longer life expectancies and the aging baby boomer population, it is expected that by 2050 the number of people aged 65 and older with AD will be between 11–16 million (Hebert, et al., 2003). It is thus imperative that clinical trials toward potential therapeutic targets, which take many years and millions of dollars to reach completion, are initiated now.

The regulation of gene expression in the presence and absence of cellular stress is increasingly recognized to be dependent on post-transcriptional mRNA regulation (Liu-Yesucevitz, et al., 2011). In the absence of stress, mature mRNA is transported from the nucleus to the cytosol where it is translated into protein. However, under stress conditions mRNA may be reversibly bound by RNA binding proteins (RBPs) and converted to quiescent cytoplasmic RNA/protein complexes called stress granules (SGs). TAR DNA-binding protein 43 (TDP-43) is a nuclear protein expressed ubiquitously from the Tardbp gene on chromosome 1 (Liu-Yesucevitz, et al., 2010). It contains two highly conserved DNA/RNA recognition motifs, followed by a C-terminal glycine-rich domain and nuclear localization and export signals, which allow TDP-43 to be continuously shuttled between the nucleus and the cytoplasm. Under normal conditions, the predominant function for TDP-43 identified to date is regulation of nuclear transcription, splicing and stability of RNA transcripts (Kumar-Singh, 2011). However, during stress TDP-43 is sequestered in the cytoplasm as a component of insoluble protein aggregates. TDP-43 may also become cleaved by activated caspases, and the phosphorylated C-terminus has become a hallmark of TDP-43 proteinopathy (Liu-Yesucevitz, et al., 2010, Toh, et al., 2011). Although initially recognized for its role in repressing the human immunodeficiency viruse type 1 (HIV-1) gene (Ou, et al., 1995), recent advances in molecular genetics have identified over 30 mutations in TDP-43 that are linked to amyotrophic lateral sclerosis (ALS) and fronto-temporal lobar dementia (FTLD) (Lagier-Tourenne and Cleveland, 2009), suggesting a causative role for the protein in disease development. Indeed, TDP-43 inclusions occur primarily in degenerating motor neurons of the brain and spinal cord. Thus, RBPs have emerged as an important player in neurodegenerative processes, and are a potential targets for therapeutic development.

Pathology of ALS, FTLD and AD

AD is characterized by region-specific brain atrophy primarily in the frontal cortex and hippocampus; regions associated with learning, memory and behavior. Intraneuronal tangles of hyperphosphorylated tau protein and extracellular plaques composed of amyloid-β (Aβ peptides are the two pathological hallmarks of AD and accumulate systematically throughout the brain. Interestingly, TDP-43 aggregates also appear in the AD brain, which suggests linkage between the pathophysiology of Aβ, tau and TDP-43. However, the nature of the relationship between TDP-43 and Aβ or tau, and indeed, the true physiologic functions of APP and Aβ are unknown, and the cellular processes underlying these pathological events remain poorly understood.

ALS is considered the most common adult-onset motor neuron disease (MND), and FTLD is the second most common form of young-onset cortical dementia (Seelaar, et al., 2011). ALS causes selective degeneration of both upper and lower motor neurons of the brain and spinal cord and culminates in muscle atrophy and death due to paralysis of the respiratory muscles (Ferraiuolo, et al., 2011, Janssens, et al., 2011), and most patients with FTLD also develop symptoms of MND such as ubiquitinated intraneuronal aggregates of TDP-43. In addition, 15–20% of ALS patients meet the criteria for FTLD (Strong and Yang, 2011), underscoring the significant clinical and pathological overlap between these two nervous system diseases.

Although distinct in many aspects, similarities between ALS, FTLD and AD are increasingly observed. These diseases primarily lead to neuronal loss and are classically diagnosed by the intraneuronal accumulation of misfolded or misaggregated proteins (ie: ubiquitin, TDP-43, Aβ and tau). Hyperactivation of kinases is another common factor, resulting in hyperphosphorylated tau filaments (in AD) or TDP-43 (in ALS and FTLD), possibly via common molecular mechanisms upstream in the disease process. TDP-43-positive inclusions are increasingly observed in combination with classic AD pathology, and are estimated to be present in up to 75% of brains from AD patients (Amador-Ortiz, et al., 2007, Wilson, et al., 2011). As we learn more about the convergence of these diseases, aligned treatment strategies may be developed to target the causative neuropathological insults prior to dementia, which appears only after pathology is pervasive.

TDP-43: Friend or Foe?

Homozygous disruption of Tardbp is embryonically lethal in mice, while disrupting only one copy of the gene leads to more subtle motor disturbances and muscle weakness (Kraemer, et al., 2010, Sephton, et al., 2010, Wu, et al., 2010). TDP-43 may therefore be linked to disease via loss-of-function mutations in which pathological TDP-43 fails to exercise its critical role in maintaining cellular homeostasis. This theory is supported by the 96% homology shared between the murine (m) and human (h) forms of TDP-43 (Feiguin, et al., 2009), perhaps resulting from an evolutionary conservation of gene function. Surprisingly, mice over-expressing wild type (WT) hTDP-43 develop ubiquitin-positive TDP-43 inclusions and ALS/FTLD-like neurodegeneration (Igaz, et al., 2011, Kumar-Singh, 2009, Swarup, et al., 2011, Xu, et al., 2010) yet mice over-expressing mTDP-43 have a very similar phenotype (Wils, et al., 2010), indicating that too much TDP-43 of either species may be just as detrimental as mutant TDP-43. Other groups have used cultured cells, Drosophila, zebrafish embryos and mouse models to confirm these results, with cumulative data demonstrating that over-expression of either mutant or WT hTDP-43 is neurotoxic, concomitant with cytoplasmic aggregation of TDP-43 and a motor phenotype (ie: shorter motor neuron axons, premature and excessive neuronal branching, and movement deficits). Despite strong evidence indicating a toxic gain-of-function for TDP-43 in vivo, the presence of these pathologies even in the absence of any TDP-43 (Barmada and Finkbeiner, 2010, Feiguin, et al., 2009, Lu, et al., 2009, Tatom, et al., 2009) raises three alternate possibilities: 1) forced over-expression of TDP-43 in animal and cellular models might be deleterious and induce degeneration through a mechanism unrelated to ALS, 2) TDP-43 might cause neurodegeneration through a mechanism that does not require formation of insoluble protein aggregates, and 3) mutant TDP-43 may cause disease via a loss-of-positive-function phenotype (Kabashi, et al., 2010). While additional studies are certainly necessary to tease out the specialized role of TDP-43 in cellular viability, ultimately there may be truth to both.

Pathological inclusions of ubiquitin and TDP-43 have been observed in degenerating motor neurons of the brain and spinal cord of ALS and FTLD patients for decades. However, in the past 5 years a wave of attention has been paid to TDP-43 not only as it relates to ALS and FTLD, but also for its potential role in AD. Indeed, a consensus panel from the United States and Europe was recently convened to update the guidelines by which AD stages should be assessed (Hyman, et al., 2012). These guidelines were last updated in 1997, and have been revised not only to account for plaque and tangle staging criteria but also for nonconventional pathologies such as ubiquitin and TDP-43 accumulation (Montine, et al., 2012). A common factor among ALS, FTLD and AD neuropathology is the predominance of pathological misfolded protein aggregates that appear well before cognitive decline, and the nature of TDP-43 as a regulator of mRNA translation and SG formation suggests that it may effect much early aspects of disease than previously considered.

The recent work, by two groups based at Georgetown University who have made significant contributions to the understanding of AD and PD, confirms TDP-43 pathology present in brain tissue from AD patients. Previously, Herman et al. used gene-manipulation techniques for the targeted accumulation of intracellular Aβ42 and found that it induced pathological changes in endogenous TDP-43, including increased expression, cleavage, and cytosolic aggregation (Herman, et al., 2011). In the current manuscript, the same group has used these techniques to express WT hTDP-43 specifically in the motor cortex of rats in the presence and absence of intracellular Aβ42 and found that, in addition to its role in RNA regulation, TDP-43 may modify the expression and activity of the BACE1 enzyme; a necessary initiating enzyme in the AD amyloid cascade.

The ABC’s of APP Processing

Aβ is a 39–43 amino acid, 4 kDa peptide resulting from the sequential proteolysis of APP, and autosomal dominant mutations in APP that increase Aβ are causal factors for early-onset familial AD (FAD) (Selkoe and Podlisny, 2002). APP is a type I transmembrane protein expressed by neurons in the CNS, with a large extracellular N-terminal domain and a smaller intracellular C-terminal domain (Figure 1). During maturation, APP becomes N- and O-glycosylated, giving rise to the mature form of APP that undergoes cleavage by β-, α- and γ-secretases (Buoso, et al., 2010). For the release of Aβ, APP must first be cleaved by β-secretase, a transmembrane aspartic protease commonly known as BACE1 (Lin, et al., 2000, Vassar, et al., 1999, Yan, et al., 1999), followed by the intramembrane γ-secretase complex consisting of nicastrin, anterior pharynx defective 1, presenilin enhancer 2 (PEN2) and presenilin 1 (PSEN1)(Edbauer, et al., 2003). Amyloidogenic cleavage of APP thus results in the release of soluble N-terminal APP (sAPPβ) or Aβ peptide from the plasma membrane, and the remaining membrane-bound C-terminal APP fragment (CTF-β). Non-amyloidogenic processing of APP is initiated primarily by A Disintegrin and Metalloproteinase Domain-Containing Protein 10 (ADAM10; α-secretase) cleavage within the Aβ domain, resulting in sAPPα. The C-terminal cytoplasmic tail of APP is further processed into the APP intracellular domain (AICD), which binds intracellular adapter proteins for complex roles in transcription and modulation of cytoskeletal dynamics (Chang and Suh, 2010). Because AD, ALS and FTLD share similar pathologies, Herman et al. ambitiously tried to find a common underlying molecular mechanism. They found that over-expressing hTDP-43 increased the levels of endogenous CTF-β via increased BACE1 activation while sparing the non-amyloidogenic pathway. This result is intriguing as recent therapeutic strategies for AD have centered around selectively modulating or inhibiting BACE1 activity (for review, see (Evin, et al., 2011, Mancini, et al., 2011)). A primary challenge in this respect has been creating a small molecule inhibitor with appropriate bioavailability in the brain (i.e.: the ability to cross the blood brain barrier in an active form). Additional challenges have been the efficacy with which these compounds actually enhance cognitive function (or prevent cognitive decline) rather than merely decreasing the Aβ burden. Unfortunately, the BACE1-inhibiting compounds created to-date have not withstood the rigors of clinical trials, either due to lack of efficacious or the pervasiveness of unwanted side effects. However, gene-based strategies rather than pharmacological intervention may be a new method to specifically target BACE1 expression without compromising other sensitive signaling cascades within the CNS, and selective genetic modification prior to the onset of clinical symptoms are crucial because memory and behavioral changes appear at the end-stages of disease progression.

Figure 1. Simplified diagram of APP structure and processing.

Figure 1

APP undergoes sequential proteolysis by b-secretase (b), a-secretase (a) and g-secretase (g) for the release of Ab from the neuronal plasma membrane. TDP-43 has been shown to increase intraneuronal Ab accumulation via increased b-secretase activation.

Rather than plaques, recent research has focused on soluble oligomeric assemblies of Aβ as the proximate cause of neuronal injury, synaptic deterioration and the eventual dementia characteristic of AD (Haass and Steiner, 2001, Klein, et al., 2001, Terry, 2001, Youmans, et al., 2012). Indeed, stable Aβ42 oligomers isolated from brain, plasma and CSF correlate with the severity of neurodegeneration in AD (Lue, et al., 1999, McLean, et al., 1999, Roher, et al., 2000, Wang, et al., 1999). Previous research has demonstrated that Aβ42 oligomers are neurotoxic in vitro, directly disrupt cognitive function and inhibit neuroplasticity that may be necessary for the formation and maintenance of memory (Cleary, et al., 2005, Dahlgren, et al., 2002, Gotz, et al., 2001, Hartley, et al., 1999, Walsh, et al., 1999, Walsh, et al., 2002, Wang, et al., 2002, Wang, et al., 1999, Westerman, et al., 2002). Evidence also indicates that soluble Aβ42 is taken up by axons, and that it may accumulate in axons or synapses leading to impaired axonal transport processes (Pigino, et al., 2009, Takahashi, et al., 2002). Interestingly, increased soluble Aβ correlates with cognitive decline in both AD patients and Aβ-transgenic mouse models (Aoki, et al., 2008, Christensen, et al., 2010, Thal, et al., 2006, Tomiyama, et al., 2010, Yu, et al., 2010). In the 3xTg mouse model of AD, increased pathological TDP-43 correlates with increased soluble Aβ oligomers, and removal of oligomeric Aβ restores TDP-43 to normal levels (Caccamo, et al., 2010). In addition, intraneuronal Aβ42 accumulates prior to extracellular amyloid deposits in humans and mice (Cataldo, et al., 2004, Christensen, et al., 2010, Gouras, et al., 2010, Langui, et al., 2004, Oakley, et al., 2006, Oddo, et al., 2006). These intraneuronal accumulations increase with age, particularly in disease-relevant brain regions, and may correlate with cognitive and behavioral deficits (Billings, et al., 2005, Busciglio, et al., 2002, Oddo, et al., 2003, Oddo, et al., 2006). To determine whether intraneuronal Aβ42 might also play a role in TDP-43 pathology, Herman et al. produced a virus that targeted Aβ42 to the endoplasmic reticulum such that Aβ42 produced by cells is not secreted but accumulates intracellulary. Thus, their model recapitulates several physiologically-relevant aspects of disease. Intraneuronal Aβ aggregates may seed further Aβ assembly, as has been proposed (Kayed, et al., 2010, Wu, et al., 2010), creating an important route for preventative intervention. Targeted delivery of pH-activated compounds to the H+ rich lysosomes or late endosomes may be a method for manipulating TDP-43-positive intraneuronal Aβ inclusions, and may provide a novel means of toxic Aβ removal.

Inflammation

Chronic inflammatory processes within the central nervous system (CNS) involve prolonged astrocytic and microglial activation and increased cytokine expression, and have been highlighted as a key contributor to AD pathogenesis. Activated microglia are initially beneficial by actively clearing Aβ from the brain (Lee and Landreth, 2010, Mandrekar, et al., 2009), yet chronic stimulation leads to release of pro-inflammatory cytokines such as TNFα, IL-6 or IL-1β (Hickman, et al., 2008, Holmes, et al., 2009, White, et al., 2005), initiating a self-propagating feedback mechanism that eventually decreases Aβ phagocytosis and increases neuronal damage (White, et al., 2005). In AD, amyloid plaques are often surrounded by microglia, likely due to initial attempts at Aβ clearance followed by an ultimate inability to overcome substantial Aβ burden. A concomitant breakdown of the blood-brain barrier (BBB) in advanced stages of AD also results in the infiltration of peripheral macrophages to the CNS to help decrease amyloid, yet removing plaques at such a late stage does little to benefit dementia. Therefore, inflammation due to neurodegeneration, as well as autoimmune disorders such as multiple sclerosis (MS), is an expanding area for clinical intervention.

In addition to inflammation, mitochondria are a primary source of reactive oxygen species (ROS) that can oxidize DNA, protein or lipids (Nunomura, et al., 2007, Orrenius, et al., 2007, Sultana, et al., 2009). Healthy mitochondria also express enzymes to detoxify ROS, such as superoxide dismutase (SOD), to remove oxidative stressors that might otherwise induce cellular apoptosis. The importance of mitochondria in the removal of O2 cannot be overstated, particularly in motor neuron degeneration. Mutations in the SOD1 gene targets mutant SOD1 protein to mitochondria, making them dysfunctional (Shi, et al., 2010), and for reasons that are not entirely understood SOD1 mutations cause familial ALS by selectively affecting motor neurons (Brotherton, et al., 2012, Kawamata and Manfredi, 2010). Oxidative stress also increases BACE1 expression (Tamagno, et al., 2005) which facilitates Aβ production, and Aβ itself generates free radicals during oligomerization (Tabner, et al., 2005). Thus, another feedback loop of mitochondrial impairment is created. Intraneuronal Aβ accumulates early in disease in humans and AD mouse models (Youmans, et al., 2012), and is consequently stationed perfectly (both spatially and temporally) to wreak havoc on already-overburdened mitochondria.

Non-steroidal anti-inflammatory drugs (NSAIDs) have been administered for years as symptomatic drugs that inhibit cyclooxygenase (COX) enzymes, thereby decreasing the conversion from arachodonic acid to prostaglandins. COX2 is increased in AD brain and it has been shown to increase Aβ pathology in vitro and in vivo, possibly via modulation of the γ-secretase complex (Qin, et al., 2006, Xiang, et al., 2002). Unfortunately, no COX2 inhibitors that have been brought to AD clinical trials (such as ibuprofen, tarenflurbil, naproxen, or clecoxib) have succeeded in slowing AD progression (for review, see (Dumont and Beal, 2011)). The Herman et al. group looked at inflammation using both immunohistochemical and biochemical methods (Figure 2 of their manuscript) and determined that hTDP-43 enhanced inflammatory processes even in the absence of exogenous Aβ42, suggesting that TDP-43 itself may regulate aspects of inflammation in the CNS. Interestingly, their data suggests that TDP-43 and TNFα may have overlapping pathways while Aβ42 increase IL-6 above that of TDP-43, indicative of two separate methods for IL-6 stimulation. If the overlapping mechanism for TNFα induction can be determined, innovative intervention strategies may be revealed.

As an alternative to traditional pharmacological modulation, some research groups have focused on natural compounds to reduce inflammation or free radical production. Curcumen appears to be at the forefront of this movement. Curcumen is a polyphenol derived from the spice turmeric that acts as a free radical scavenger. It is capable of crossing the BBB from the bloodstream, and can reduce Aβ fibrillogenesis and plaque deposition in vitro and in vivo, respectively (Frautschy, et al., 2001, Garcia-Alloza, et al., 2007, Lim, et al., 2001, Yang, et al., 2005). Unfortunately, curcumen has very little bioactivity when ingested (Anand, et al., 2007, Yang, et al., 2008), limiting its practical at-home use by AD patients or caregivers. However, alterations to its chemical structure may generate analogous compounds that are not broken down during digestion and maintain the ability to inhibit Aβ aggregation. Development of such compounds seems imminent and would make a huge impact on diseases of amyloidosis, making this a very exciting time in the field of drug development.

What Remains to be Studied

Some research suggests that Aβ40 peptide alone is insufficient to form dense-core Aβ deposits in the absence of Aβ42, and that the longer more hydrophobic Aβ42 peptide is necessary for plaque formation in vivo (McGowan, et al., 2005). In the current manuscript, Herman et al. reveal that over-expression of hTDP-43 alone increases murine Aβ40 (Figure 4 of their manuscript). This would be expected based on its general BACE1 up-regulation. However, it is interesting that hTDP-43 increases Aβ40 in the formic acid (FA)-soluble fraction. FA is commonly used as a terminal extraction buffer during serial protein extractions, as it effectively breaks up dense-cored amyloid (Youmans, et al., 2011). The current data suggests that Aβ40 is an essential component of plaques, as viral expression of Aβ42 in the absence of hTDP43 does not induce presumable plaque deposition (ie: Aβ42 alone does not increase FA-soluble Aβ40). However, viral Aβ42 expression combined with hTDP-43 seemingly flips the ratio of soluble:insoluble Aβ40 compared to hTDP-43 alone, which suggests that altering the Aβ40:Aβ42 ratio may determine the aggregation properties of murine Aβ40 (Tai, et al., 2011). Nixon and colleagues have provided overwhelming evidence for lysosomal Aβ accumulation that is supported by recent work from the LaDu lab (Youmans, et al., 2012), which they predict leads to lysosomal membrane destabilization followed by neuronal death (Cataldo, et al., 1997, Nixon, 2007, Nixon and Cataldo, 2006). It has been proposed that death of Aβ-containing neurons and the subsequent release of lysosomal content may seed dense-cored neuritic plaques. Cellular TDP-43 has not been reported to accumulate in lysosomal compartments, but it remains a possibility that intracellular Aβ may merge with cytosolic TDP-43 aggregates and that neuronal death releases these “seeds” upon which Aβ40 peptides can deposit. Although hTDP-43 alone does not increase Aβ42 levels in this study (Figure 1 of their work), APP is endogenous from rat in this model and therefore does not harbor the familial AD mutations that favor γ-secretase-based Aβ42 production in humans. The authors also overexposed Western blots of rat brain tissue to depict high molecular weight Aβ aggregates (see their 4th figure), providing further evidence for TDP-43 modulation of Aβ assembly state. Several critical next steps toward understanding the link between TDP-43 and Aβ include the use of Thioflavin-S to monitor fibrillogenesis, and oligomer-specific antibodies to reveal toxic oligomer production specifically in transgenic mice expressing mutant forms of human APP.

The apoE gene is the sole genetic risk factor modulating both FAD and sporadic AD. The majority of apoE in the CNS is produced locally by glial cells (DeMattos, et al., 2001), and a major role for apoE in the CNS is neuronal uptake of cholesterol for growth, repair and synaptogenesis (Hayashi, et al., 2004, Mahley, 1988, Mauch, et al., 2001). Indeed, following nervous system injury, glial apoE synthesis increases by up to 150-fold, and post-mortem apolipoprotein E (apoE) immunoreactivity is localized to extracellular amyloid plaques (Huang, et al., 2001, Zhang, et al., 2001). Additionally, intraneuronal Aβ forms cytoplasmic complexes with apoE in AD brains, and these complexes localize to neurons undergoing cell death (LaFerla, et al., 1997). In vitro, apoE enhances the uptake of soluble Aβ at nerve terminals (Gylys, et al., 2003), indicating that internalization may be a prerequisite for intraneuronal Aβ accumulation. This uptake is blocked by the addition of apoE receptor associated protein (RAP; an antagonist for apoE receptors), implicating a role for apoE in the uptake of soluble Aβ species (Gylys, et al., 2003). Further, the presence of RAP abolishes Aβ42-induced neurotoxicity in vitro (Manelli, et al., 2007). It is therefore surprising that TDP-43 pathology does not appear to correlate with apoE genotype (Nelson, et al., 2011). Single nucleotide polymorphisms (SNPs) in the Tardbp gene do not significantly correlate with apoE status (Shibata, et al., 2009), although a comprehensive evaluation of apoE as it relates to TDP-43 has not been reported. Accordingly, apoE genotyping of brains containing TDP-43 inclusions or the presence/absence of apoE receptors may provide essential missing information that would allow us to integrate our understanding of AD pathology with that of other neurodegenerative diseases of aging.

Conclusions

Over the past 5 years TDP-43 has been increasingly linked to neurodegenerative pathology, with an emphasis on TDP-43-positive inclusions in AD brains. In 2008, V. Lee’s group wrote a comprehensive review describing TDP-43 proteinopathies as insoluble cytoplasmic aggregates in which TDP-43 moves out of the nucleus and into the cytoplasm, and how these inclusions may be linked to AD (Kwong, et al., 2008). Although the nature of scientific research often necessitates focusing on one or two disease-related proteins, converging evidence suggests that multiple pathways may overlap in ALS, FTLD and AD, including signaling cascades, axonal transport and feedback loops for regulation of cell cycling, growth or regulated cell death. Although the specifics of RBP-based regulation are beyond the scope of this review, RBPs are emerging as crucial chaperones for many such pathways, and their dysregulation apparently moves beyond cellular death to overall systemic failure. The next 5 years will undoubtedly reveal distinct fundamental roles for RBPs in neurodegenerative disease progression. Results from Herman et al. have established the groundwork for uncovering the synergistic effects of TDP-43 and intraneuronal Ab42, which hopefully will reveal upstream molecular targets that can be modulated toward the ultimate goal of disease prevention.

Acknowledgments

We would like to acknowledge financial support to BW (NINDS, NIEHS) and KY (NIA T32-AG06697).

References

  1. Amador-Ortiz C, Lin WL, Ahmed Z, Personett D, Davies P, Duara R, Graff-Radford NR, Hutton ML, Dickson DW. TDP-43 immunoreactivity in hippocampal sclerosis and Alzheimer’s disease. Ann Neurol. 2007;61:435–445. doi: 10.1002/ana.21154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Anand P, Kunnumakkara AB, Newman RA, Aggarwal BB. Bioavailability of curcumin: problems and promises. Mol Pharm. 2007;4:807–818. doi: 10.1021/mp700113r. [DOI] [PubMed] [Google Scholar]
  3. Aoki M, Volkmann I, Tjernberg LO, Winblad B, Bogdanovic N. Amyloid beta-peptide levels in laser capture microdissected cornu ammonis 1 pyramidal neurons of Alzheimer’s brain. Neuroreport. 2008;19:1085–1089. doi: 10.1097/WNR.0b013e328302c858. [DOI] [PubMed] [Google Scholar]
  4. Barmada SJ, Finkbeiner S. Pathogenic TARDBP mutations in amyotrophic lateral sclerosis and frontotemporal dementia: disease-associated pathways. Rev Neurosci. 2010;21:251–272. doi: 10.1515/revneuro.2010.21.4.251. [DOI] [PubMed] [Google Scholar]
  5. Billings LM, Oddo S, Green KN, McGaugh JL, Laferla FM. Intraneuronal Abeta causes the onset of early Alzheimer’s disease-related cognitive deficits in transgenic mice. Neuron. 2005;45:675–688. doi: 10.1016/j.neuron.2005.01.040. [DOI] [PubMed] [Google Scholar]
  6. Brotherton TE, Li Y, Cooper D, Gearing M, Julien JP, Rothstein JD, Boylan K, Glass JD. Localization of a toxic form of superoxide dismutase 1 protein to pathologically affected tissues in familial ALS. Proc Natl Acad Sci U S A. 2012;109:5505–5510. doi: 10.1073/pnas.1115009109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Buoso E, Lanni C, Schettini G, Govoni S, Racchi M. beta-Amyloid precursor protein metabolism: focus on the functions and degradation of its intracellular domain. Pharmacol Res. 2010;62:308–317. doi: 10.1016/j.phrs.2010.05.002. [DOI] [PubMed] [Google Scholar]
  8. Busciglio J, Pelsman A, Wong C, Pigino G, Yuan M, Mori H, Yankner BA. Altered metabolism of the amyloid beta precursor protein is associated with mitochondrial dysfunction in Down’s syndrome. Neuron. 2002;33:677–688. doi: 10.1016/s0896-6273(02)00604-9. [DOI] [PubMed] [Google Scholar]
  9. Caccamo A, Magri A, Oddo S. Age-dependent changes in TDP-43 levels in a mouse model of Alzheimer disease are linked to Abeta oligomers accumulation. Mol Neurodegener. 2010;5:51. doi: 10.1186/1750-1326-5-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Cataldo AM, Barnett JL, Pieroni C, Nixon RA. Increased neuronal endocytosis and protease delivery to early endosomes in sporadic Alzheimer’s disease: neuropathologic evidence for a mechanism of increased beta-amyloidogenesis. Journal of Neuroscience. 1997;17:6142–6151. doi: 10.1523/JNEUROSCI.17-16-06142.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Cataldo AM, Petanceska S, Terio NB, Peterhoff CM, Durham R, Mercken M, Mehta PD, Buxbaum J, Haroutunian V, Nixon RA. Abeta localization in abnormal endosomes: association with earliest Abeta elevations in AD and Down syndrome. Neurobiol Aging. 2004;25:1263–1272. doi: 10.1016/j.neurobiolaging.2004.02.027. [DOI] [PubMed] [Google Scholar]
  12. Chang KA, Suh YH. Possible roles of amyloid intracellular domain of amyloid precursor protein. BMB Rep. 2010;43:656–663. doi: 10.5483/BMBRep.2010.43.10.656. [DOI] [PubMed] [Google Scholar]
  13. Christensen DZ, Schneider-Axmann T, Lucassen PJ, Bayer TA, Wirths O. Accumulation of intraneuronal Abeta correlates with ApoE4 genotype. Acta Neuropathol. 2010;119:555–566. doi: 10.1007/s00401-010-0666-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cleary JP, Walsh DM, Hofmeister JJ, Shankar GM, Kuskowski MA, Selkoe DJ, Ashe KH. Natural oligomers of the amyloid-beta protein specifically disrupt cognitive function. Nat Neurosci. 2005;8:79–84. doi: 10.1038/nn1372. [DOI] [PubMed] [Google Scholar]
  15. Dahlgren KN, Manelli AM, Stine WB, Jr, Baker LK, Krafft GA, LaDu MJ. Oligomeric and fibrillar species of amyloid-beta peptides differentially affect neuronal viability. J Biol Chem. 2002;277:32046–32053. doi: 10.1074/jbc.M201750200. [DOI] [PubMed] [Google Scholar]
  16. DeMattos RB, Brendza RP, Heuser JE, Kierson M, Cirrito JR, Fryer J, Sullivan PM, Fagan AM, Han X, Holtzman DM. Purification and characterization of astrocyte-secreted apolipoprotein E and J-containing lipoproteins from wild-type and human apoE transgenic mice. Neurochem Int. 2001;39:415–425. doi: 10.1016/s0197-0186(01)00049-3. [DOI] [PubMed] [Google Scholar]
  17. Dumont M, Beal MF. Neuroprotective strategies involving ROS in Alzheimer disease. Free Radic Biol Med. 2011;51:1014–1026. doi: 10.1016/j.freeradbiomed.2010.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Edbauer D, Winkler E, Regula JT, Pesold B, Steiner H, Haass C. Reconstitution of gamma-secretase activity. Nat Cell Biol. 2003;5:486–488. doi: 10.1038/ncb960. [DOI] [PubMed] [Google Scholar]
  19. Evin G, Lessene G, Wilkins S. BACE inhibitors as potential drugs for the treatment of Alzheimer’s disease: focus on bioactivity. Recent Pat CNS Drug Discov. 2011;6:91–106. doi: 10.2174/157488911795933938. [DOI] [PubMed] [Google Scholar]
  20. Feiguin F, Godena VK, Romano G, D’Ambrogio A, Klima R, Baralle FE. Depletion of TDP-43 affects Drosophila motoneurons terminal synapsis and locomotive behavior. FEBS Lett. 2009;583:1586–1592. doi: 10.1016/j.febslet.2009.04.019. [DOI] [PubMed] [Google Scholar]
  21. Ferraiuolo L, Kirby J, Grierson AJ, Sendtner M, Shaw PJ. Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis. Nat Rev Neurol. 2011;7:616–630. doi: 10.1038/nrneurol.2011.152. [DOI] [PubMed] [Google Scholar]
  22. Frautschy SA, Hu W, Kim P, Miller SA, Chu T, Harris-White ME, Cole GM. Phenolic anti-inflammatory antioxidant reversal of Abeta-induced cognitive deficits and neuropathology. Neurobiol Aging. 2001;22:993–1005. doi: 10.1016/s0197-4580(01)00300-1. [DOI] [PubMed] [Google Scholar]
  23. Garcia-Alloza M, Borrelli LA, Rozkalne A, Hyman BT, Bacskai BJ. Curcumin labels amyloid pathology in vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model. J Neurochem. 2007;102:1095–1104. doi: 10.1111/j.1471-4159.2007.04613.x. [DOI] [PubMed] [Google Scholar]
  24. Gotz J, Chen F, van Dorpe J, Nitsch RM. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science. 2001;293:1491–1495. doi: 10.1126/science.1062097. [DOI] [PubMed] [Google Scholar]
  25. Gouras GK, Tampellini D, Takahashi RH, Capetillo-Zarate E. Intraneuronal beta-amyloid accumulation and synapse pathology in Alzheimer’s disease. Acta neuropathologica. 2010;119:523–541. doi: 10.1007/s00401-010-0679-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Gylys KH, Fein JA, Tan AM, Cole GM. Apolipoprotein E enhances uptake of soluble but not aggregated amyloid-beta protein into synaptic terminals. J Neurochem. 2003;84:1442–1451. doi: 10.1046/j.1471-4159.2003.01643.x. [DOI] [PubMed] [Google Scholar]
  27. Haass C, Steiner H. Protofibrils, the unifying toxic molecule of neurodegenerative disorders? Nat Neurosci. 2001;4:859–860. doi: 10.1038/nn0901-859. [DOI] [PubMed] [Google Scholar]
  28. Hartley DM, Walsh DM, Ye CP, Diehl T, Vasquez S, Vassilev PM, Teplow DB, Selkoe DJ. Protofibrillar intermediates of amyloid beta-protein induce acute electrophysiological changes and progressive neurotoxicity in cortical neurons. J Neurosci. 1999;19:8876–8884. doi: 10.1523/JNEUROSCI.19-20-08876.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Hayashi H, Campenot RB, Vance DE, Vance JE. Glial lipoproteins stimulate axon growth of central nervous system neurons in compartmented cultures. J Biol Chem. 2004;279:14009–14015. doi: 10.1074/jbc.M313828200. [DOI] [PubMed] [Google Scholar]
  30. Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch Neurol. 2003;60:1119–1122. doi: 10.1001/archneur.60.8.1119. [DOI] [PubMed] [Google Scholar]
  31. Herman AM, Khandelwal PJ, Stanczyk BB, Rebeck GW, Moussa CE. beta-amyloid triggers ALS-associated TDP-43 pathology in AD models. Brain Res. 2011;1386:191–199. doi: 10.1016/j.brainres.2011.02.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Heron MH, DL, Xu J, Scot C, Jejada-Vera B. Deaths: Preliminary data for 2006. Hyattsville: 2008. [Google Scholar]
  33. Hickman SE, Allison EK, El Khoury J. Microglial dysfunction and defective beta-amyloid clearance pathways in aging Alzheimer’s disease mice. The Journal of neuroscience. 2008;28:8354–8360. doi: 10.1523/JNEUROSCI.0616-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Holmes C, Cunningham C, Zotova E, Woolford J, Dean C, Kerr S, Culliford D, Perry VH. Systemic inflammation and disease progression in Alzheimer disease. Neurology. 2009;73:768–774. doi: 10.1212/WNL.0b013e3181b6bb95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Huang Y, Liu XQ, Wyss-Coray T, Brecht WJ, Sanan DA, Mahley RW. Apolipoprotein E fragments present in Alzheimer’s disease brains induce neurofibrillary tangle-like intracellular inclusions in neurons. Proc Natl Acad Sci U S A. 2001;98:8838–8843. doi: 10.1073/pnas.151254698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Hyman BT, Phelps CH, Beach TG, Bigio EH, Cairns NJ, Carrillo MC, Dickson DW, Duyckaerts C, Frosch MP, Masliah E, Mirra SS, Nelson PT, Schneider JA, Thal DR, Thies B, Trojanowski JQ, Vinters HV, Montine TJ. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease. Alzheimers Dement. 2012;8:1–13. doi: 10.1016/j.jalz.2011.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Igaz LM, Kwong LK, Lee EB, Chen-Plotkin A, Swanson E, Unger T, Malunda J, Xu Y, Winton MJ, Trojanowski JQ, Lee VM. Dysregulation of the ALS-associated gene TDP-43 leads to neuronal death and degeneration in mice. J Clin Invest. 2011;121:726–738. doi: 10.1172/JCI44867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Janssens J, Kleinberger G, Wils H, Van Broeckhoven C. The role of mutant TAR DNA-binding protein 43 in amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Biochem Soc Trans. 2011;39:954–959. doi: 10.1042/BST0390954. [DOI] [PubMed] [Google Scholar]
  39. Kabashi E, Lin L, Tradewell ML, Dion PA, Bercier V, Bourgouin P, Rochefort D, Bel Hadj S, Durham HD, Vande Velde C, Rouleau GA, Drapeau P. Gain and loss of function of ALS-related mutations of TARDBP (TDP-43) cause motor deficits in vivo. Hum Mol Genet. 2010;19:671–683. doi: 10.1093/hmg/ddp534. [DOI] [PubMed] [Google Scholar]
  40. Kawamata H, Manfredi G. Import, maturation, and function of SOD1 and its copper chaperone CCS in the mitochondrial intermembrane space. Antioxid Redox Signal. 2010;13:1375–1384. doi: 10.1089/ars.2010.3212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Kayed R, Canto I, Breydo L, Rasool S, Lukacsovich T, Wu J, Albay R, 3rd, Pensalfini A, Yeung S, Head E, Marsh JL, Glabe C. Conformation dependent monoclonal antibodies distinguish different replicating strains or conformers of prefibrillar abeta oligomers. Molecular neurodegeneration. 2010;5:57. doi: 10.1186/1750-1326-5-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Klein WL, Krafft GA, Finch CE. Targeting small Abeta oligomers: the solution to an Alzheimer’s disease conundrum? Trends Neurosci. 2001;24:219–224. doi: 10.1016/s0166-2236(00)01749-5. [DOI] [PubMed] [Google Scholar]
  43. Kraemer BC, Schuck T, Wheeler JM, Robinson LC, Trojanowski JQ, Lee VM, Schellenberg GD. Loss of murine TDP-43 disrupts motor function and plays an essential role in embryogenesis. Acta Neuropathol. 2010;119:409–419. doi: 10.1007/s00401-010-0659-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Kumar-Singh S. Hereditary and sporadic forms of abeta-cerebrovascular amyloidosis and relevant transgenic mouse models. Int J Mol Sci. 2009;10:1872–1895. doi: 10.3390/ijms10041872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Kumar-Singh S. Progranulin and TDP-43: mechanistic links and future directions. J Mol Neurosci. 2011;45:561–573. doi: 10.1007/s12031-011-9625-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Kwong LK, Uryu K, Trojanowski JQ, Lee VM. TDP-43 proteinopathies: neurodegenerative protein misfolding diseases without amyloidosis. Neurosignals. 2008;16:41–51. doi: 10.1159/000109758. [DOI] [PubMed] [Google Scholar]
  47. LaFerla FM, Troncoso JC, Strickland DK, Kawas CH, Jay G. Neuronal cell death in Alzheimer’s disease correlates with apoE uptake and intracellular Abeta stabilization. J Clin Invest. 1997;100:310–320. doi: 10.1172/JCI119536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Lagier-Tourenne C, Cleveland DW. Rethinking ALS: the FUS about TDP-43. Cell. 2009;136:1001–1004. doi: 10.1016/j.cell.2009.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Langui D, Girardot N, El Hachimi KH, Allinquant B, Blanchard V, Pradier L, Duyckaerts C. Subcellular topography of neuronal Abeta peptide in APPxPS1 transgenic mice. Am J Pathol. 2004;165:1465–1477. doi: 10.1016/s0002-9440(10)63405-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Lee CY, Landreth GE. The role of microglia in amyloid clearance from the AD brain. Journal of neural transmission (Vienna, Austria. 2010;117:949–960. doi: 10.1007/s00702-010-0433-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM. The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J Neurosci. 2001;21:8370–8377. doi: 10.1523/JNEUROSCI.21-21-08370.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Lin X, Koelsch G, Wu S, Downs D, Dashti A, Tang J. Human aspartic protease memapsin 2 cleaves the beta-secretase site of beta-amyloid precursor protein. Proc Natl Acad Sci U S A. 2000;97:1456–1460. doi: 10.1073/pnas.97.4.1456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Liu-Yesucevitz L, Bassell GJ, Gitler AD, Hart AC, Klann E, Richter JD, Warren ST, Wolozin B. Local RNA translation at the synapse and in disease. J Neurosci. 2011;31:16086–16093. doi: 10.1523/JNEUROSCI.4105-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Liu-Yesucevitz L, Bilgutay A, Zhang YJ, Vanderweyde T, Citro A, Mehta T, Zaarur N, McKee A, Bowser R, Sherman M, Petrucelli L, Wolozin B. Tar DNA binding protein-43 (TDP-43) associates with stress granules: analysis of cultured cells and pathological brain tissue. PLoS One. 2010;5:e13250. doi: 10.1371/journal.pone.0013250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Lu Y, Ferris J, Gao FB. Frontotemporal dementia and amyotrophic lateral sclerosis-associated disease protein TDP-43 promotes dendritic branching. Mol Brain. 2009;2:30. doi: 10.1186/1756-6606-2-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Lue LF, Kuo YM, Roher AE, Brachova L, Shen Y, Sue L, Beach T, Kurth JH, Rydel RE, Rogers J. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer’s disease. American Journal Of Pathology. 1999;155:853–862. doi: 10.1016/s0002-9440(10)65184-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Mahley RW. Apolipoprotein E: Cholesterol transport protein with expanding role in cell biology. Science. 1988;240:622–630. doi: 10.1126/science.3283935. [DOI] [PubMed] [Google Scholar]
  58. Mancini F, De Simone A, Andrisano V. Beta-secretase as a target for Alzheimer’s disease drug discovery: an overview of in vitro methods for characterization of inhibitors. Anal Bioanal Chem. 2011;400:1979–1996. doi: 10.1007/s00216-011-4963-x. [DOI] [PubMed] [Google Scholar]
  59. Mandrekar S, Jiang Q, Lee CY, Koenigsknecht-Talboo J, Holtzman DM, Landreth GE. Microglia mediate the clearance of soluble Abeta through fluid phase macropinocytosis. J Neurosci. 2009;29:4252–4262. doi: 10.1523/JNEUROSCI.5572-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Manelli AM, Bulfinch LC, Sullivan PM, LaDu MJ. Abeta42 neurotoxicity in primary co-cultures: effect of apoE isoform and Abeta conformation. Neurobiol Aging. 2007;28:1139–1147. doi: 10.1016/j.neurobiolaging.2006.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Mauch DH, Nagler K, Schumacher S, Goritz C, Muller EC, Otto A, Pfrieger FW. CNS synaptogenesis promoted by glia-derived cholesterol. Science. 2001;294:1354–1357. doi: 10.1126/science.294.5545.1354. [DOI] [PubMed] [Google Scholar]
  62. McGowan E, Pickford F, Kim J, Onstead L, Eriksen J, Yu C, Skipper L, Murphy MP, Beard J, Das P, Jansen K, Delucia M, Lin WL, Dolios G, Wang R, Eckman CB, Dickson DW, Hutton M, Hardy J, Golde T. Abeta42 is essential for parenchymal and vascular amyloid deposition in mice. Neuron. 2005;47:191–199. doi: 10.1016/j.neuron.2005.06.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. McLean CA, Cherny RA, Fraser FW, Fuller SJ, Smith MJ, Beyreuther K, Bush AI, Masters CL. Soluble pool of Abeta amyloid as a determinant of severity of neurodegeneration in Alzheimer’s disease. Ann Neurol. 1999;46:860–866. doi: 10.1002/1531-8249(199912)46:6<860::aid-ana8>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
  64. Montine TJ, Phelps CH, Beach TG, Bigio EH, Cairns NJ, Dickson DW, Duyckaerts C, Frosch MP, Masliah E, Mirra SS, Nelson PT, Schneider JA, Thal DR, Trojanowski JQ, Vinters HV, Hyman BT. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease: a practical approach. Acta Neuropathol. 2012;123:1–11. doi: 10.1007/s00401-011-0910-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Nelson PT, Schmitt FA, Lin Y, Abner EL, Jicha GA, Patel E, Thomason PC, Neltner JH, Smith CD, Santacruz KS, Sonnen JA, Poon LW, Gearing M, Green RC, Woodard JL, Van Eldik LJ, Kryscio RJ. Hippocampal sclerosis in advanced age: clinical and pathological features. Brain. 2011;134:1506–1518. doi: 10.1093/brain/awr053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Nixon RA. Autophagy, amyloidogenesis and Alzheimer disease. J Cell Sci. 2007;120:4081–4091. doi: 10.1242/jcs.019265. [DOI] [PubMed] [Google Scholar]
  67. Nixon RA, Cataldo AM. Lysosomal system pathways: genes to neurodegeneration in Alzheimer’s disease. J Alzheimers Dis. 2006;9:277–289. doi: 10.3233/jad-2006-9s331. [DOI] [PubMed] [Google Scholar]
  68. Nunomura A, Moreira PI, Lee HG, Zhu X, Castellani RJ, Smith MA, Perry G. Neuronal death and survival under oxidative stress in Alzheimer and Parkinson diseases. CNS Neurol Disord Drug Targets. 2007;6:411–423. doi: 10.2174/187152707783399201. [DOI] [PubMed] [Google Scholar]
  69. Oakley H, Cole SL, Logan S, Maus E, Shao P, Craft J, Guillozet-Bongaarts A, Ohno M, Disterhoft J, Van Eldik L, Berry R, Vassar R. Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: potential factors in amyloid plaque formation. J Neurosci. 2006;26:10129–10140. doi: 10.1523/JNEUROSCI.1202-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R, Metherate R, Mattson MP, Akbari Y, LaFerla FM. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron. 2003;39:409–421. doi: 10.1016/s0896-6273(03)00434-3. [DOI] [PubMed] [Google Scholar]
  71. Oddo S, Caccamo A, Smith IF, Green KN, LaFerla FM. A dynamic relationship between intracellular and extracellular pools of Abeta. Am J Pathol. 2006;168:184–194. doi: 10.2353/ajpath.2006.050593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Orrenius S, Gogvadze V, Zhivotovsky B. Mitochondrial oxidative stress: implications for cell death. Annu Rev Pharmacol Toxicol. 2007;47:143–183. doi: 10.1146/annurev.pharmtox.47.120505.105122. [DOI] [PubMed] [Google Scholar]
  73. Ou SH, Wu F, Harrich D, Garcia-Martinez LF, Gaynor RB. Cloning and characterization of a novel cellular protein, TDP-43, that binds to human immunodeficiency virus type 1 TAR DNA sequence motifs. J Virol. 1995;69:3584–3596. doi: 10.1128/jvi.69.6.3584-3596.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Pigino G, Morfini G, Atagi Y, Deshpande A, Yu C, Jungbauer L, LaDu M, Busciglio J, Brady S. Disruption of fast axonal transport is a pathogenic mechanism for intraneuronal amyloid beta. Proc Natl Acad Sci U S A. 2009;106:5907–5912. doi: 10.1073/pnas.0901229106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Qin W, Peng Y, Ksiezak-Reding H, Ho L, Stetka B, Lovati E, Pasinetti GM. Inhibition of cyclooxygenase as potential novel therapeutic strategy in N141I presenilin-2 familial Alzheimer’s disease. Mol Psychiatry. 2006;11:172–181. doi: 10.1038/sj.mp.4001773. [DOI] [PubMed] [Google Scholar]
  76. Roher AE, Baudry J, Chaney MO, Kuo YM, Stine WB, Emmerling MR. Oligomerization and fibril assembly of the amyloid-beta protein. Biochim Biophys Acta. 2000;1502:31–43. doi: 10.1016/s0925-4439(00)00030-2. [DOI] [PubMed] [Google Scholar]
  77. Seelaar H, Rohrer JD, Pijnenburg YA, Fox NC, van Swieten JC. Clinical, genetic and pathological heterogeneity of frontotemporal dementia: a review. J Neurol Neurosurg Psychiatry. 2011;82:476–486. doi: 10.1136/jnnp.2010.212225. [DOI] [PubMed] [Google Scholar]
  78. Selkoe DJ, Podlisny MB. Deciphering the Genetic Basis of Alzheimer’s Disease. Annu Rev Genomics Hum Genet. 2002;6:6. doi: 10.1146/annurev.genom.3.022502.103022. [DOI] [PubMed] [Google Scholar]
  79. Sephton CF, Good SK, Atkin S, Dewey CM, Mayer P, 3rd, Herz J, Yu G. TDP-43 is a developmentally regulated protein essential for early embryonic development. J Biol Chem. 2010;285:6826–6834. doi: 10.1074/jbc.M109.061846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Shi P, Wei Y, Zhang J, Gal J, Zhu H. Mitochondrial dysfunction is a converging point of multiple pathological pathways in amyotrophic lateral sclerosis. J Alzheimers Dis. 2010;20(Suppl 2):S311–324. doi: 10.3233/JAD-2010-100366. [DOI] [PubMed] [Google Scholar]
  81. Shibata N, Ohnuma T, Baba H, Arai H. Genetic association analysis between TDP-43 polymorphisms and Alzheimer’s disease in a Japanese population. Dement Geriatr Cogn Disord. 2009;28:325–329. doi: 10.1159/000251194. [DOI] [PubMed] [Google Scholar]
  82. Strong MJ, Yang W. The frontotemporal syndromes of ALS. Clinicopathological correlates. J Mol Neurosci. 2011;45:648–655. doi: 10.1007/s12031-011-9609-0. [DOI] [PubMed] [Google Scholar]
  83. Sultana R, Perluigi M, Butterfield DA. Oxidatively modified proteins in Alzheimer’s disease (AD), mild cognitive impairment and animal models of AD: role of Abeta in pathogenesis. Acta Neuropathol. 2009;118:131–150. doi: 10.1007/s00401-009-0517-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Swarup V, Phaneuf D, Bareil C, Robertson J, Rouleau GA, Kriz J, Julien JP. Pathological hallmarks of amyotrophic lateral sclerosis/frontotemporal lobar degeneration in transgenic mice produced with TDP-43 genomic fragments. Brain. 2011;134:2610–2626. doi: 10.1093/brain/awr159. [DOI] [PubMed] [Google Scholar]
  85. Tabner BJ, El-Agnaf OM, Turnbull S, German MJ, Paleologou KE, Hayashi Y, Cooper LJ, Fullwood NJ, Allsop D. Hydrogen peroxide is generated during the very early stages of aggregation of the amyloid peptides implicated in Alzheimer disease and familial British dementia. J Biol Chem. 2005;280:35789–35792. doi: 10.1074/jbc.C500238200. [DOI] [PubMed] [Google Scholar]
  86. Tai LM, Youmans KL, Jungbauer L, Yu C, Ladu MJ. Introducing Human APOE into Abeta Transgenic Mouse Models. Int J Alzheimers Dis. 2011;2011:810981. doi: 10.4061/2011/810981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Takahashi RH, Milner TA, Li F, Nam EE, Edgar MA, Yamaguchi H, Beal MF, Xu H, Greengard P, Gouras GK. Intraneuronal Alzheimer abeta42 accumulates in multivesicular bodies and is associated with synaptic pathology. Am J Pathol. 2002;161:1869–1879. doi: 10.1016/s0002-9440(10)64463-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Tamagno E, Parola M, Bardini P, Piccini A, Borghi R, Guglielmotto M, Santoro G, Davit A, Danni O, Smith MA, Perry G, Tabaton M. Beta-site APP cleaving enzyme up-regulation induced by 4-hydroxynonenal is mediated by stress-activated protein kinases pathways. J Neurochem. 2005;92:628–636. doi: 10.1111/j.1471-4159.2004.02895.x. [DOI] [PubMed] [Google Scholar]
  89. Tatom JB, Wang DB, Dayton RD, Skalli O, Hutton ML, Dickson DW, Klein RL. Mimicking aspects of frontotemporal lobar degeneration and Lou Gehrig’s disease in rats via TDP-43 overexpression. Mol Ther. 2009;17:607–613. doi: 10.1038/mt.2009.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Terry RD. An honorable compromise regarding amyloid in Alzheimer disease. Ann Neurol. 2001;49:684. [PubMed] [Google Scholar]
  91. Thal DR, Capetillo-Zarate E, Del Tredici K, Braak H. The development of amyloid beta protein deposits in the aged brain. Science of aging knowledge environment. 2006;2006:re1. doi: 10.1126/sageke.2006.6.re1. [DOI] [PubMed] [Google Scholar]
  92. Toh H, Chitramuthu BP, Bennett HP, Bateman A. Structure, function, and mechanism of progranulin; the brain and beyond. J Mol Neurosci. 2011;45:538–548. doi: 10.1007/s12031-011-9569-4. [DOI] [PubMed] [Google Scholar]
  93. Tomiyama T, Matsuyama S, Iso H, Umeda T, Takuma H, Ohnishi K, Ishibashi K, Teraoka R, Sakama N, Yamashita T, Nishitsuji K, Ito K, Shimada H, Lambert MP, Klein WL, Mori H. A mouse model of amyloid beta oligomers: their contribution to synaptic alteration, abnormal tau phosphorylation, glial activation, and neuronal loss in vivo. The Journal of neuroscience. 2010;30:4845–4856. doi: 10.1523/JNEUROSCI.5825-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, Teplow DB, Ross S, Amarante P, Loeloff R, Luo Y, Fisher S, Fuller J, Edenson S, Lile J, Jarosinski MA, Biere AL, Curran E, Burgess T, Louis JC, Collins F, Treanor J, Rogers G, Citron M. Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science. 1999;286:735–741. doi: 10.1126/science.286.5440.735. [DOI] [PubMed] [Google Scholar]
  95. Walsh DM, Hartley DM, Kusumoto Y, Fezoui Y, Condron MM, Lomakin A, Benedek GB, Selkoe DJ, Teplow DB. Amyloid beta-protein fibrillogenesis. Structure and biological activity of protofibrillar intermediates. J Biol Chem. 1999;274:25945–25952. doi: 10.1074/jbc.274.36.25945. [DOI] [PubMed] [Google Scholar]
  96. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–539. doi: 10.1038/416535a. [DOI] [PubMed] [Google Scholar]
  97. Wang HW, Pasternak JF, Kuo H, Ristic H, Lambert MP, Chromy B, Viola KL, Klein WL, Stine WB, Krafft GA, Trommer BL. Soluble oligomers of beta amyloid (1–42) inhibit long-term potentiation but not long-term depression in rat dentate gyrus. Brain Res. 2002;924:133–140. doi: 10.1016/s0006-8993(01)03058-x. [DOI] [PubMed] [Google Scholar]
  98. Wang J, Dickson DW, Trojanowski JQ, Lee VM. The levels of soluble versus insoluble brain Abeta distinguish Alzheimer’s disease from normal and pathologic aging. Exp Neurol. 1999;158:328–337. doi: 10.1006/exnr.1999.7085. [DOI] [PubMed] [Google Scholar]
  99. Westerman MA, Cooper-Blacketer D, Mariash A, Kotilinek L, Kawarabayashi T, Younkin LH, Carlson GA, Younkin SG, Ashe KH. The relationship between Abeta and memory in the Tg2576 mouse model of Alzheimer’s disease. J Neurosci. 2002;22:1858–1867. doi: 10.1523/JNEUROSCI.22-05-01858.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. White JA, Manelli AM, Holmberg KH, Van Eldik LJ, LaDu MJ. Differential effects of oligomeric and fibrillar amyloid-beta1–42 on astrocyte-mediated inflammation. Neurobiol Dis. 2005;18:459–465. doi: 10.1016/j.nbd.2004.12.013. [DOI] [PubMed] [Google Scholar]
  101. Wils H, Kleinberger G, Janssens J, Pereson S, Joris G, Cuijt I, Smits V, Ceuterick-de Groote C, Van Broeckhoven C, Kumar-Singh S. TDP-43 transgenic mice develop spastic paralysis and neuronal inclusions characteristic of ALS and frontotemporal lobar degeneration. Proc Natl Acad Sci U S A. 2010;107:3858–3863. doi: 10.1073/pnas.0912417107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Wilson AC, Dugger BN, Dickson DW, Wang DS. TDP-43 in aging and Alzheimer’s disease - a review. Int J Clin Exp Pathol. 2011;4:147–155. [PMC free article] [PubMed] [Google Scholar]
  103. Wu JW, Breydo L, Isas JM, Lee J, Kuznetsov YG, Langen R, Glabe C. Fibrillar oligomers nucleate the oligomerization of monomeric amyloid beta but do not seed fibril formation. J Biol Chem. 2010;285:6071–6079. doi: 10.1074/jbc.M109.069542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Wu SB, Ma YS, Wu YT, Chen YC, Wei YH. Mitochondrial DNA mutation-elicited oxidative stress, oxidative damage, and altered gene expression in cultured cells of patients with MERRF syndrome. Mol Neurobiol. 2010;41:256–266. doi: 10.1007/s12035-010-8123-7. [DOI] [PubMed] [Google Scholar]
  105. Xiang Z, Ho L, Yemul S, Zhao Z, Qing W, Pompl P, Kelley K, Dang A, Teplow D, Pasinetti GM. Cyclooxygenase-2 promotes amyloid plaque deposition in a mouse model of Alzheimer’s disease neuropathology. Gene Expr. 2002;10:271–278. doi: 10.3727/000000002783992352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Xu YF, Gendron TF, Zhang YJ, Lin WL, D’Alton S, Sheng H, Casey MC, Tong J, Knight J, Yu X, Rademakers R, Boylan K, Hutton M, McGowan E, Dickson DW, Lewis J, Petrucelli L. Wild-type human TDP-43 expression causes TDP-43 phosphorylation, mitochondrial aggregation, motor deficits, and early mortality in transgenic mice. J Neurosci. 2010;30:10851–10859. doi: 10.1523/JNEUROSCI.1630-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Yan R, Bienkowski MJ, Shuck ME, Miao H, Tory MC, Pauley AM, Brashier JR, Stratman NC, Mathews WR, Buhl AE, Carter DB, Tomasselli AG, Parodi LA, Heinrikson RL, Gurney ME. Membrane-anchored aspartyl protease with Alzheimer’s disease beta-secretase activity. Nature. 1999;402:533–537. doi: 10.1038/990107. [DOI] [PubMed] [Google Scholar]
  108. Yang CS, Sang S, Lambert JD, Lee MJ. Bioavailability issues in studying the health effects of plant polyphenolic compounds. Mol Nutr Food Res. 2008;52(Suppl 1):S139–151. doi: 10.1002/mnfr.200700234. [DOI] [PubMed] [Google Scholar]
  109. Yang F, Lim GP, Begum AN, Ubeda OJ, Simmons MR, Ambegaokar SS, Chen PP, Kayed R, Glabe CG, Frautschy SA, Cole GM. Curcumin Inhibits Formation of Amyloid {beta} Oligomers and Fibrils, Binds Plaques, and Reduces Amyloid in Vivo. J Biol Chem. 2005;280:5892–5901. doi: 10.1074/jbc.M404751200. [DOI] [PubMed] [Google Scholar]
  110. Youmans KL, Leung S, Zhang J, Maus E, Baysac K, Bu G, Vassar R, Yu C, Ladu MJ. Amyloid-beta42 alters apolipoprotein E solubility in brains of mice with five familial AD mutations. J Neurosci Methods. 2011;196:51–59. doi: 10.1016/j.jneumeth.2010.12.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Youmans KL, Tai LM, Kanekiyo T, Stine WB, Jr, Michon SC, Nwabuisi-Heath E, Manelli A, Fu Y, Riordan S, Eimer WA, Binder L, Bu G, Yu C, Hartley DM, Ladu MJ. Intraneuronal Abeta detection in 5xFAD mice by a new Abeta-specific antibody. Mol Neurodegener. 2012;7:8. doi: 10.1186/1750-1326-7-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Yu C, Nwabuisi-Heath E, Laxton K, Ladu MJ. Endocytic pathways mediating oligomeric Abeta42 neurotoxicity. Mol Neurodegener. 2010;5:19. doi: 10.1186/1750-1326-5-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Zhang D, McQuade JA, Shockley K, Levin L, Marques M, Crutcher K. Proteolysis of apolipoprotein E and Alzheimer’s disease pathology. Alzheimer’s Reports. 2001;4:67–80. [Google Scholar]

RESOURCES