Cytotoxic ApoE Cannot Reach Nucleus from Cytosol

Hence No Effect on Gene Expression, or Effect on Microtubules



Ronald B. DeMattos (now at Eli Lilly in Indianapolis), Fayanne E. Thorngate (still at SUNY), and David L. Williams (deceased in early 2000s?) . . . Department of Pharmacological Sciences, University Medical Center, State University of New York at Stony Brook, Stony Brook, New York 11794


Abstract: Genetic evidence indicates that apolipoprotein E4 (apoE4) is a risk factor for the development of Alzheimer’s disease. A controversial hypothesis proposes that apoE, a typical secretory protein, accesses the neuronal cytosol in which apoE3, but not apoE4, protects tau from hyperphosphorylation. However, no conclusive evidence for the presence of apoE in the cytosolic compartment has been presented. We designed a novel assay to test whether apoE can access the cytosol via escape from the endocytic pathway by incorporating a nuclear localization signal (NLS) into apoE. Control experiments demonstrated that apoE plus NLS (apoE+NLS) is chaperoned to the nucleus if it reaches the cytosolic compartment. When exogenous apoE+NLS was endocytosed by neuronal cells, no nuclear apoE was detected, indicating that apoE remains within the endocytic pathway and does not escape into the cytosol. Furthermore, we show that direct cytosolic expression of apoE is cytotoxic. These data argue that effects of apoE on the neuronal cytoskeleton and on neurite outgrowth are not mediated via cytosolic interactions but rather by actions originating at the cell surface.


DeMattos et. al. is a reaction to In vitro research about the allegedly toxic nature of apoE4 proteins.

The paper then goes on to show that ApoE proteins are confined to lisosomes, and do not normally enter neuronal cytosol (fluid and material within the cell membrane), much less the nucleus of the cell.

DeMattos describes using Neuro-2a neuroblastoma cells transfected with plasmid coding for either secreted apoE (outside the cell) or intracellular apoE (inside the cell membrane in the endosomes, cytosol, and nucleus.

MoreLater


The second sentence points at some papers worth reading about ApoE production outside the liver

and in the brain

where it is secreted by astrocytes and microglial cells (as naked molecules or as part of lipid packages?)


What's happened since?

36 Citations on Google Scholar, 25 available


  • The role of apolipoprotein E in Alzheimer's disease

  • J Kim, JM Basak, DM Holtzman - Neuron, 2009 - Elsevier
    • The ɛ4 allele of apolipoprotein E (APOE) is the major genetic risk factor for Alzheimer's disease (AD). Although there have been numerous studies attempting to elucidate theunderlying mechanism for this increased risk, how apoE4 influences AD onset and progression has yet to be proven. However, prevailing evidence suggests that the differential effects of apoE isoforms on Aβ aggregation and clearance play the major role in AD pathogenesis. Other potential mechanisms, such as the differential modulation of neurotoxicity and tau phosphorylation by apoE isoforms as well as its role in synaptic plasticity and neuroinflammation, have not been ruled out. Inconsistent results among studies have made it difficult to define whether the APOE ε4 allele represents a gain of toxic function, a loss of neuroprotective function, or both. Therapeutic strategies based on apoE propose to reduce the toxic effects of apoE4 or to restore the physiological, protective functions of apoE.
  • 1-s2.0-S0896627309005492-main.pdf "DeMattos et al., 1999"

    • p294 /8: Along with amyloid plaque formation, hyperphosphorylation of the microtubule-binding protein tau and subsequent formation of neurofibrillary tangles are hallmarks of AD pathology. The relationship between apoE and tau has not been thoroughly investigated and the results are much less clear than the association between APOE ε4 allele dose and amyloid plaque burden. Although some studies reported a positive relationship between the neurofibrillary tangle density and APOE ε4 allele dosage (Ghebremedhin et al., 1998; Nagy et al., 1995; Ohm et al., 1995; Polvikoski et al., 1995), others found no clear correlation (Itoh and Yamada, 1996; Landen et al., 1996b; Morris et al., 1995; Olichney et al., 1996; Oyama et al., 1995). Unlike studies with human subjects, data from in vitro and animal models appear to be more consistent among studies. Under in vitro conditions, apoE3 binds tightly to tau and forms an SDS-stable complex through the interaction of the N-terminal domain of apoE3 and the microtubule-binding repeat regions of tau, whereas apoE4 does not interact significantly with tau (Strittmatter et al., 1994). The interaction between apoE3 and tau was prevented by the phosphorylation of tau, suggesting that apoE3 binds preferentially to nonphosphorylated tau. However, there is currently no conclusive evidence demonstrating localization of apoE to the neuronal cytosol, where the majority of tau exists under normal conditions (DeMattos et al., 1999). Therefore, the physiological relevance of the direct interaction between apoE and tau remains to be determined. Of note, more recent data suggests the possibility that a fragment of apoE4 (1–272 amino acids), but not full-length apoE4, the secretory pathway, translocates to the cytosolic compartment, and interacts with cytoskeletal components, including tau and neurofilament (Chang et al., 2005). Alternatively, the effects of apoE on tau phosphorylation could be explained by an intracellular signaling pathway induced by apoE (Harris et al., 2004; Hoe et al., 2005a), rather than the direct interaction between apoE and tau. Although in vitro studies have provided some insights, in vivo studies proving whether an apoE-isoform dependent effect on tau even exists will be critical.

      • Chang, S., ran Ma, T., Miranda, R.D., Balestra, M.E., Mahley, R.W., and Huang, Y. (2005). Lipid- and receptor-binding regions of apolipoprotein E4 fragments act in concert to cause mitochondrial dysfunction and neurotoxicity. free: Proc. Natl. Acad. Sci. USA 102, 18694–18699.

      • Harris, F.M., Brecht, W.J., Xu, Q., Mahley, R.W., and Huang, Y. (2004). Increased tau phosphorylation in apolipoprotein E4 transgenic mice is associated with activation of extracellular signal-regulated kinase: modulation by zinc. free: J. Biol. Chem. 279, 44795–44801.

      • Hoe, H.S., Harris, D.C., and Rebeck, G.W. (2005a). Multiple pathways of apolipoprotein E signaling in primary neurons.free: J. Neurochem. 93, 145–155.


  • Apolipoprotein E fragments present in Alzheimer's disease brains induce neurofibrillary tangle-like intracellular inclusions in neurons

  • Y Huang, XQ Liu, T Wyss-Coray, WJ Brecht, DA Sanan, and RW Mahley- Proceedings of the National Academy of Sciences, 2001
    • Human apolipoprotein (apo) E4, a major risk factor for Alzheimer's disease (AD), occurs in amyloid plaques and neurofibrillary tangles (NFTs) in AD brains; however, its role in the pathogenesis of these lesions is unclear. Here we demonstrate that carboxyl-terminal-truncated forms of apoE, which occur in AD brains and cultured neurons, induce intracellular NFT-like inclusions in neurons. These cytosolic inclusions were composed of phosphorylated tau, phosphorylated neurofilaments of high molecular weight, and truncated apoE. Truncated apoE4, especially apoE4(⌬272–299), induced inclusions in up to 75% of transfected neuronal cells, but not in transfected nonneuronal cells. ApoE4 was more susceptible to truncation than apoE3 and resulted in much greater intracellular inclusion formation. These results suggest that apoE4 preferentially undergoes intracellular processing, creating a bioactive fragment that interacts with cytoskeletal components and induces NFT-like inclusions containing phosphorylated tau and phosphorylated neurofilaments of high molecular weight in neurons.

    • PNAS-2001-Huang-8838-43-1.pdf ref 23 needs evaluation

  • p8840/3 Carboxyl-Terminal-Truncated ApoE Induces Intracellular NFT-Like Inclusions. To determine whether carboxyl-terminal-truncated apoE induces intracellular NFT-like inclusions, we expressed apoE3 or apoE4 constructs possessing carboxyl-terminal truncations in Neuro-2a cells. Expression of apoE4 lacking the first 28 amino acids of the carboxyl terminus [apoE4(Δ272–299)] resulted in intracellular NFT-like inclusions in 78 ± 8% of transfected Neuro-2a cells (Fig. 2e). The inclusions were recognized by anti-p-tau (Fig. 2f ), which colocalized with apoE (Fig. 2g), and anti-p-NF-H (data not shown). Expression of apoE3(Δ272–299) also induced intracellular inclusions, but they were smaller (Fig. 2h) and occurred in significantly fewer cells (32 ± 5% versus 78 ± 8%, P < 0.001). Importantly, exogenous apoE4 (Δ272–299), which had been complexed with β-very low density lipoproteins as a lipid transport vehicle and incubated with Neuro-2a cells, also induced intracellular NFT-like inclusions (Fig. 2d). Thus, both endogenously expressed and exogenously added apoE with the carboxyl-terminal truncation induced NFT-like inclusions in Neuro-2a cells. The truncated apoE probably escapes the secretory or the endosomal-lysosomal internalization pathway, enters the cytosol, and interacts with p-tau and p-NF-H. There is evidence that apoE can appear in the cytosol of various cells (20–22), although another study failed to show this (23).


  • Expression of human apolipoprotein E4 in neurons causes hyperphosphorylation of protein tau in the brains of transgenic mice

  • I Tesseur, J Van Dorpe, K Spittaels, C van den Haute, D Moechars, and F van Leuven, The American journal of Pathology, 2000
    • Epidemiological studies have established that the epsilon 4 allele of the ApoE gene (ApoE4) constitutes an important risk factor for Alzheimer's disease and might influence the outcome of central nervous system injury. The mechanism by which ApoE4 contributes to thedevelopment of neurodegeneration remains unknown. To test one hypothesis or mode of action of ApoE, we generated transgenic mice that overexpressed human ApoE4 in different cell types in the brain, using four distinct gene promoter constructs. Many transgenic mice expressing ApoE4 in neurons developed motor problems accompanied by muscle wasting, loss of body weight, and premature death. Overexpression of human ApoE4 in neurons resulted in hyperphosphorylation of the microtubule-associated protein tau. In three independent transgenic lines from two different promoter constructs, increased phosphorylation of protein tau was correlated with ApoE4 expression levels. Hyperphosphorylation of protein tau increased with age. In the hippocampus, astrogliosis and ubiquitin-positive inclusions were demonstrated. These findings demonstrate that expression of ApoE in neurons results in hyperphosphorylation of protein tau and suggests a role for ApoE in neuronal cytoskeletal stability and metabolism.

  • 1-s2.0-S0002944010649632-main.pdf Ref 74
    • p 961 /11 To explain the genetic association of ApoE4 to AD, two types of mechanisms have been proposed. First, ApoE could function as a “pathological chaperone,” affecting clearance of β-amyloid and causing amyloid deposition. (70–72) We obtained no evidence for the presence of amyloid plaques in any of our ApoE4 transgenic mice, which of course might be because only endogenous mouse APP is present, which is less amyloidogenic. (39) The second hypothesis states that ApoE interacts with the microtubule-associated protein tau, thereby altering its phosphorylation state, and hence is involved in stabilizing the neuronal cytoskeleton. (2,73) The results presented here support this hypothesis. However, the route by which ApoE gains access to the neuronal cytoplasm has been the major criticism against this hypothesis and subject of much speculation. Most recently, it was shown that direct expression of ApoE in the cytosol of Neuro-2a cells is toxic. (74) The ApoE4 transgenic mice are, however, not expected to express ApoE directly in the cytosol, and we also did not observe obvious signs of neurotoxicity. In addition, the hyperphosphorylation of protein tau was not restricted to brain regions expressing human ApoE as shown by immunohistochemistry. Therefore, we favor a mechanism involving an indirect interaction between ApoE and protein tau. In addition, this implies that hyper-phosphorylation of protein tau is not simply a nonspecific downstream event marking degeneration.


  • The role of apolipoprotein E in Alzheimer's disease, acute brain injury and cerebrovascular disease: evidence of common mechanisms and utility of animal models

  • K Horsburgh, MO McCarron, F White, JAR Nicoll - Neurobiology of aging, 2000

    • The ϵ4 allele of apolipoprotein E (APOE denotes gene; apoE denotes protein) is a major risk factor for Alzheimer's disease (AD). More recent evidence indicates an association with a poor outcome after acute brain injury including that due to head trauma and intracerebral hemorrhage. APOE gene polymorphism also influences the risk of hemorrhage in cerebral amyloid angiopathy. These diverse brain disorders seem to have some mechanisms in common. The multiplicity of the roles of apoE within the central nervous system is currently being unraveled. For example, apoE can interact with amyloid β-protein and tau, proteins central to the pathogenesis of AD. In addition to these effects, it is proposed that one of the major functions of apoE is to mediate neuronal protection, repair and remodeling. In all of the different roles proposed, there are marked apoE-isoform specific differences. Although it remains to be clarified which is the most important mechanism(s) in each disorder in which apoE is involved, these isoform specific differences seem to underly a genetically determined susceptibility to outcome from acute brain injury and to AD with APOE ε4 conferring relative vulnerability. This review focuses on apoE research, from clinical studies to animal models, in AD, acute brain injury and cerebrovascular disease and explores the common mechanisms that may explain some of the complex underlying neurobiology.
  • 1-s2.0-S019745800000097X-main.pdf ref 20
    • p251/7 In vitro evidence is consistent with an isoform-dependent neurotrophic role of apoE. Rabbit dorsal root ganglion neurons and neuroblastoma cells incubated with lipoproteins alone have enhanced neurite outgrowth that is further enhanced in the presence of apoE E3 lipoproteins and inhibited in the presence of apoE E4 lipoproteins. [33,86]. Similar results are obtained when cells are stably transfected to secrete apoE E3 or apoE E4; in the presence of lipoproteins, cells expressing APOE ε3 have extensive neurite outgrowth whereas neurite extension is suppressed in cells expressing APOE ε4 [7]. One of the cellular events associated with neurite outgrowth is an apoE-isoform specific effect on the cytoskeleton. ApoE E3 stabilizes the formation of microtubules in vitro in contrast to apoE E4 that is associated with a destabilization of the microtubule assembly [23]. ApoE E3 can interact with tau, a microtubule associated protein, whereas apoE E4 is unable to bind tau. One hypothesis is that apoE E3 by binding to tau protects it from hyperphosphorylation and the generation of intracellular neurofibrillary tangles in AD brain. ApoE is present at high concentrations in neurons after injury in rodent and human brain [30,41– 44,56] and may be expressed at low levels normally in human brain [134] providing functional relevance between the interactions of apoE with intracellular cytoskeletal proteins. In vitro studies have also indicated that there may be a preferential accumulation of apoE E3 and E2 in neurons as compared to apoE E4 [51]. Some groups have proposed that apoE E3 escapes lysosomal degradation and enters the cytosol where it is able to interact with cytosolic proteins such as tau and microtubule associated proteins. This mechanism may account for the apoE isoform specific effect of apoE E3 in promoting neurite outgrowth and microtubule stability and the lack of such an effect with apoE E4. However, it has been shown that apoE remains within the endocytic pathway and does not escape into the cytosol and argues that the effects of apoE are mediated by actions on the cell surface and not via cytosolic interactions [20]. The intracellular fate of the apoE isoforms remains a critical question.










T Hosono and M Michikawa - Molecular Neurodegeneration, 2009