Elsevier

Neurobiology of Aging

Volume 34, Issue 4, April 2013, Pages 1007-1017
Neurobiology of Aging

Review
APOE and neuroenergetics: an emerging paradigm in Alzheimer's disease

https://doi.org/10.1016/j.neurobiolaging.2012.10.011Get rights and content

Abstract

APOE is the major known genetic risk factor for late-onset Alzheimer's disease. Though relationships between APOE-encoded apolipoprotein E and β-amyloid are increasingly well described, mounting evidence supports wide-ranging effects of APOE on the brain. Specifically, APOE appears to affect brain network activity and closely related neuroenergetic functions that might be involved in vulnerability to neurodegenerative pathophysiology. These effects highlight the salience of further investigation into the diverse influences of APOE. Therefore, this article reviews the interplay between APOE and neuroenergetics and proposes areas for further investigation. This research might lead to the identification of novel therapeutic targets for the treatment and/or prevention of Alzheimer's disease.

Introduction

Despite decades of intense research, the causes of Alzheimer's disease (AD) remain poorly understood and truly effective therapies remain out of reach. AD is expected to become markedly more prevalent over the next half century (Ferri et al., 2005), which further intensifies the need to develop therapies as soon as possible. Since the initial reports linking APOE to AD in the early 1990s (Corder et al., 1993; Strittmatter et al., 1993), considerable research has focused on elucidating the mechanisms by which the gene contributes to risk for the disease. Current evidence supports APOE-encoded apolipoprotein E (apoE) isoforms differentially modulating β-amyloid aggregation and clearance (Bu, 2009; Holtzman et al., 2012; Kim et al., 2009). Genetically, APOE ε4 is associated with dramatically increased risk, APOE ε3 is associated with neutral risk, and APOE ε2 is associated with decreased risk (Bertram and Tanzi, 2008; Gomez-Isla et al., 1996). APOE-related risk is gene–dose dependent: in the United States, when compared with persons homozygous for risk-neutral APOE ε3, APOE ε4 homozygotes have up to 15 times and APOE ε4 heterozygotes up to 4 times the risk for developing AD (Ashford and Mortimer, 2002; Raber et al., 2004). Therefore, ameliorating APOE ε4’s powerful effects might be a viable strategy to decrease AD incidence—delaying the average age of onset by 5 years could reduce the number of cases by more than 50% and save nearly $300 billion in Medicare spending in coming years (Sperling et al., 2011).

Though findings regarding apoE and its interactions with β-amyloid are essential to the current understanding of AD pathophysiology, it is important to further develop knowledge of the potential for APOE to affect brain function in a manner that might precede or be independent of β-amyloid pathology. Notably, apoE has known effects on cholesterol transport, inflammation, neurodevelopment, and synaptic plasticity, and study in these contexts clearly represents vital avenues of research. Mitochondrial energy metabolism and cellular bioenergetics in the brain (i.e., neuroenergetics) have also begun to be linked to the genetic risk conferred by APOE. Therefore, the intent of this article is to review the brain imaging background and potential cellular and molecular mechanisms for this emerging avenue of approach, with the hope that this rapidly evolving knowledge might stimulate innovative research approaches and the identification of tractable therapeutic targets for treatment and/or prevention of disease.

Section snippets

Metabolic brain imaging in Alzheimer's disease

Long-standing efforts have focused on the relevance of neuroenergetics in AD both as a mediator of β-amyloid-induced changes and as an independent driver (Reddy and Beal, 2008; Smith et al., 2002; Swerdlow et al., 2010; Yao et al., 2011). The energetic needs of the human brain are remarkable; despite comprising only 2% of gross body mass, the brain accounts for 20% of the body's glucose and oxygen consumption (Jolivet et al., 2009). The provision of energy to the synapse is vital for the

Metabolic brain imaging and APOE in older populations

The use of brain imaging to investigate APOE's effects is rooted in the idea of using APOE-related changes in CMRgl as an endophenotype—a quantitative, genetically-based biomarker associated with disease risk (Reiman, 2007). Thus, we have proposed CMRgl as an end point in the evaluation of AD treatments and/or preventive therapies, with the underlying assertion being that region-specific CMRgl alterations correlate with disease risk (Reiman et al., 2001), perhaps as a measure of cognitive

Brain imaging and APOE in young adults

In addition to studies of middle- and late-middle age individuals, we have used FDG PET to study even earlier effects of APOE ε4 on brain functional measures. In a study of cognitively normal subjects 20–39 years old, APOE ε4 carriers exhibited significantly decreased CMRgl in the PCC and other cortical regions associated with metabolic defects in older APOE ε4 carriers and AD patients, in this case several decades before the potential onset of dementia, and also several decades before any

Limitations and future directions of metabolic brain imaging

Importantly, both FDG PET and cytochrome oxidase studies have an inherent limitation in not being able to identify with certainty whether neurons or glial cells (particularly astrocytes) are the cellular source of the metabolic signal. Though the brain contains different cell types with different bioenergetic profiles, the compartmentalization of these bioenergetic processes has often been ignored, in part because of the limited resolution of brain imaging. For example, the astrocyte-neuron

Cellular functions of apoE in the brain

Though brain imaging can provide a great deal of insight to the effects of APOE, an understanding of cellular function and mechanisms conferring risk is essential for any effective therapeutic development. The primary function of apoE in the brain is to traffic cholesterol and other lipids. Though it is expressed in several peripheral tissues, apoE is most highly expressed in the liver and brain (Elshourbagy et al., 1985). In the brain, apoE is the primary apolipoprotein that associates with

ApoE structure and its relevance to disease

Aspects of apoE structure are thought to be a driving force in its role in AD risk. ApoE is a 34 kDa, 299 amino acid glycoprotein with 2 major functional domains: the N-terminal domain exists as a 4 helix bundle and contains the apoE receptor binding region at residues 136–150; the C-terminal domain is highly α-helical and contains the major lipid binding region at residues 244–272 (Aggerbeck et al., 1988; Dong et al., 1994; Wetterau et al., 1988; Wilson et al., 1991). When unbound by lipid,

Mechanisms of apoE cleavage and processing

As previously emphasized, the cellular source of apoE is highly regulated, and neuronal production of apoE appears to be driven by astrocytic signaling mechanisms (Harris et al., 2004b), particularly as a result of neural injury. It has been demonstrated in postmortem human samples that apoE4 undergoes neuron-specific proteolysis (Huang et al., 2001); this dramatic increase in intracellular cleavage of apoE4 compared with apoE3 and apoE2 is thought to be because of apoE4's much greater tendency

Effects of apoE on cytoskeletal components and intracellular trafficking

ApoE cleavage fragments have been shown to have a number of effects on the cytoskeleton and related intracellular trafficking functions (Fig. 3). In Neuro-2a cells, expressed apoE4 (Δ272–299) has been found to interact with cytoskeletal proteins to form neurofibrillary tangle-like structures containing phosphorylated tau (Huang et al., 2001). Mice expressing high levels of apoE4 (Δ272–299) in neurons display AD-like neurofibrillary tangles and die at 2–4 months. With lower levels of expression

ApoE and mitochondrial function

In addition to its deleterious effects on intracellular transport, apoE4 has been shown to have direct effects on mitochondria. In an early study, apoE4 was shown to bind the α and β subunits of the F1 portion of ATP synthase in liver (Mahley et al., 1989). Though direct effects on enzyme function were not assessed in that study, neuronal apoE4 fragments have subsequently been shown to perturb mitochondrial function. In Neuro-2a cultures expressing apoE4 (Δ272–299), apoE4 fragments that escape

ApoE effects on astrocytes

Beyond the effects shown in neurons, apoE4 also appears to alter function in astrocytes. ApoE4 induces endoplasmic reticulum stress in astrocytes (Zhong et al., 2009) that does not occur in neurons (Brodbeck et al., 2011). However, again pointing to the cell type-specific importance of apoE expression, mouse primary astrocyte cultures expressing apoE4 do not show significant changes in ETC gene expression (Chen et al., 2011). Additionally, transgenic mice that express apoE4 under the control of

Potential roles for TOMM40

APOE is located in a region of linkage disequilibrium on chromosome 19 that also encompasses TOMM40 and APOC1. TOMM40, which encodes Tom40, the pore-forming subunit of the translocase of the outer mitochondrial membrane, has now been proposed as a potential genetic risk factor for AD (Roses et al., 2010). However, the exact nature of the relationship, at least partly, but perhaps not entirely attributable, to linkage disequilibrium, is not yet understood as the initial findings have not been

Conclusions

The strong association between APOE and AD has been known for nearly 2 decades, during which time significant advances have been made in understanding how APOE might contribute to disease risk. Though considerably more research is needed to establish the mechanistic effects of APOE on disease processes, mounting evidence linking APOE to alterations in neuroenergetics has illuminated exciting new areas for research. Because of the apparently early nature of these functional changes (i.e., often

Disclosure statement

All authors report no conflicts of interest.

Acknowledgements

This work was supported by the Arizona Alzheimer's Consortium, the State of Arizona, and the Arizona Alzheimer's Disease Core Center (P30 AG19610).

References (157)

  • J.R. Cirrito et al.

    Endocytosis is required for synaptic activity-dependent release of amyloid-beta in vivo

    Neuron

    (2008)
  • J.R. Cirrito et al.

    Synaptic activity regulates interstitial fluid amyloid-beta levels in vivo

    Neuron

    (2005)
  • N.A. Dennis et al.

    Temporal lobe functional activity and connectivity in young adult APOE4 carriers

    Alzheimers Dement.

    (2010)
  • L.M. Dong et al.

    Human apolipoprotein E4 domain interaction. Arginine 61 and glutamic acid 255 interact to direct the preference for very low density lipoproteins

    J. Biol. Chem.

    (1996)
  • L.M. Dong et al.

    Human apolipoprotein E. role of arginine 61 in mediating the lipoprotein preferences of the E3 and E4 isoforms

    J. Biol. Chem.

    (1994)
  • C.P. Ferri et al.

    Global prevalence of dementia: a Delphi consensus study

    Lancet

    (2005)
  • R.P. Friedland et al.

    Pathological and positron emission tomographic correlations in Alzheimer's disease

    Lancet

    (1985)
  • F.M. Harris et al.

    Increased tau phosphorylation in apolipoprotein E4 transgenic mice is associated with activation of extracellular signal-regulated kinase: modulation by zinc

    J. Biol. Chem.

    (2004)
  • F.M. Harris et al.

    Astroglial regulation of apolipoprotein E expression in neuronal cells. Implications for Alzheimer's disease

    J. Biol. Chem.

    (2004)
  • R.E. Hartman et al.

    Behavioral phenotyping of GFAP-apoE3 and -apoE4 transgenic mice: apoE4 mice show profound working memory impairments in the absence of Alzheimer's-like neuropathology

    Exp. Neurol.

    (2001)
  • D.M. Hatters et al.

    Apolipoprotein E structure: insights into function

    Trends Biochem. Sci.

    (2006)
  • D.M. Hatters et al.

    Amino-terminal domain stability mediates apolipoprotein E aggregation into neurotoxic fibrils

    J. Mol. Biol.

    (2006)
  • Y. Huang

    Abeta-independent roles of apolipoprotein E4 in the pathogenesis of Alzheimer's disease

    Trends Mol. Med.

    (2010)
  • Y. Ji et al.

    Apolipoprotein E isoform-specific regulation of dendritic spine morphology in apolipoprotein E transgenic mice and Alzheimer's disease patients

    Neuroscience

    (2003)
  • S.C. Johnson et al.

    The effect of TOMM40 poly-T length on gray matter volume and cognition in middle-aged persons with APOEε3/ε3 genotype

    Alzheimer's Dement.

    (2011)
  • J. Kim et al.

    The role of apolipoprotein E in Alzheimer's disease

    Neuron

    (2009)
  • R.C. Klein et al.

    Progressive loss of synaptic integrity in human apolipoprotein E4 targeted replacement mice and attenuation by apolipoprotein E2

    Neuroscience

    (2010)
  • G. Li et al.

    GABAergic interneuron dysfunction impairs hippocampal neurogenesis in adult apolipoprotein E4 knockin mice

    Cell Stem Cell

    (2009)
  • Z. Li et al.

    The importance of dendritic mitochondria in the morphogenesis and plasticity of spines and synapses

    Cell

    (2004)
  • W.S. Liang et al.

    Association between GAB2 haplotype and higher glucose metabolism in Alzheimer's disease-affected brain regions in cognitively normal APOEε4 carriers

    Neuroimage

    (2011)
  • A.F. MacAskill et al.

    Control of mitochondrial transport and localization in neurons

    Trends Cell. Biol.

    (2010)
  • S. Minoshima et al.

    Posterior cingulate cortex in Alzheimer's disease

    Lancet

    (1994)
  • J.A. Morrow et al.

    Apolipoprotein E4 forms a molten globule. a potential basis for its association with disease

    J. Biol. Chem.

    (2002)
  • B.P. Nathan et al.

    The inhibitory effect of apolipoprotein E4 on neurite outgrowth is associated with microtubule depolymerization

    J. Biol.Chem.

    (1995)
  • B.P. Nathan et al.

    Apolipoprotein E4 inhibits, and apolipoprotein E3 promotes neurite outgrowth in cultured adult mouse cortical neurons through the low-density lipoprotein receptor-related protein

    Brain Res.

    (2002)
  • R.E. Pitas et al.

    Astrocytes synthesize apolipoprotein E and metabolize apolipoprotein E-containing lipoproteins

    Biochim. Biophys. Acta

    (1987)
  • P. Alexopoulos et al.

    Hippocampal volume differences between healthy young apolipoprotein E ε2 and ε4 carriers

    J. Alzheimer's Dis.

    (2011)
  • I. Allaman et al.

    Amyloid-beta aggregates cause alterations of astrocytic metabolic phenotype: impact on neuronal viability

    J. Neurosci.

    (2010)
  • Y. Andrews-Zwilling et al.

    Apolipoprotein E4 causes age- and tau-dependent impairment of GABAergic interneurons, leading to learning and memory deficits in mice

    J. Neurosci.

    (2010)
  • K. Aoki et al.

    Increased expression of neuronal apolipoprotein E in human brain with cerebral infarction

    Stroke

    (2003)
  • J.W. Ashford et al.

    Non-familial Alzheimer's disease is mainly due to genetic factors

    J. Alzheimer's Dis.

    (2002)
  • L.M. Bekris et al.

    APOE mRNA and protein expression in postmortem brain are modulated by an extended haplotype structure

    Am. J. Med. Genet. B Neuropsychiatr. Genet.

    (2010)
  • R.D. Bell et al.

    Apolipoprotein E controls cerebrovascular integrity via cyclophilin A

    Nature

    (2012)
  • A.W. Bero et al.

    Neuronal activity regulates the regional vulnerability to amyloid-β deposition

    Nat. Neurosci.

    (2011)
  • L. Bertram et al.

    Thirty years of Alzheimer's disease genetics: the implications of systematic meta-analyses

    Nat. Rev. Neurosci.

    (2008)
  • N. Bolender et al.

    Multiple pathways for sorting mitochondrial precursor proteins

    EMBO Rep.

    (2008)
  • P.A. Boyle et al.

    Mild cognitive impairment: risk of Alzheimer disease and rate of cognitive decline

    Neurology

    (2006)
  • J.K. Boyles et al.

    Apolipoprotein E associated with astrocytic glia of the central nervous system and with nonmyelinating glia of the peripheral nervous system

    J. Clin. Invest.

    (1985)
  • W.J. Brecht et al.

    Neuron-specific apolipoprotein e4 proteolysis is associated with increased tau phosphorylation in brains of transgenic mice

    J. Neurosci.

    (2004)
  • G. Bu

    Apolipoprotein E and its receptors in Alzheimer's disease: pathways, pathogenesis and therapy

    Nat. Rev. Neurosci.

    (2009)
  • Cited by (58)

    • APOE and metabolic dysfunction in Alzheimer's disease

      2020, International Review of Neurobiology
      Citation Excerpt :

      Furthermore, recent evidence suggests that these alterations in glucose metabolism occur very early in the neurodegenerative process (de Leon et al., 2001; Mosconi et al., 2008; Reiman et al., 1996; Small et al., 1995). Perhaps the most consistent metabolic feature associated with E4 carriage is a pattern of brain glucose hypometabolism similar to that seen in AD (Brandon et al., 2018; Wolf et al., 2013). This E4-associated reduction in glucose uptake via FDG-PET, first described several decades ago, is typically observed in the posterior cingulate, anterior cingulate cortex, and several other parietal, temporal, and prefrontal regions, including many regions within the default mode network (Altmann et al., 2015; Reiman et al., 1996).

    • Dissecting the structural and functional impact of SNPs located in the spinal muscular atrophy associated gene SMN1 using in silico analysis

      2019, Gene Reports
      Citation Excerpt :

      SNPs that result in an alteration in the corresponding protein product are known as non-synonymous or missense SNPs. Missense SNPs are of particular interest because they might have damaging effect on protein structure, function, solubility and stability, which may result in diseased phenotypes (Hamosh et al., 1992; Wolf et al., 2013). Nearly half of the genetic variations related to human inherited diseases can be attributed to missense SNPs (Krawczak et al., 2000).

    View all citing articles on Scopus
    View full text