Review article
Therapies for Alzheimer’s disease: a metabolic perspective

https://doi.org/10.1016/j.ymgme.2021.01.011Get rights and content

Highlights

  • In Alzheimer’s disease, cerebral glucose transporters (mainly GLUT1 and GLUT3) seem to be downregulated.

  • Glucose hypometabolism destabilizes the normal flow of the hexosamine biosynthesis pathway, reducing O-GlcNAcylation rates.

  • An unbalanced relation between glycation and O-GlcNAcylation can promote Alzheimer’s disease pathological features.

  • Impaired insulin signalling is often observed in AD patients and associated with its more prominent features.

  • Metabolic therapies have shown to be promising therapeutic approaches for AD.

Abstract

Alzheimer’s disease (AD) is one of the most common forms of dementia in the elderly. Currently, there are over 50 million cases of dementia worldwide and it is expected that it will reach 136 million by 2050. AD is described as a neurodegenerative disease that gradually compromises memory and learning capacity. Patients often exhibit brain glucose hypometabolism and are more susceptible to develop type 2 diabetes or insulin resistance in comparison with age-matched controls. This suggests that there is a link between both pathologies. Glucose metabolism and the tricarboxylic acid cycle are tightly related to mitochondrial performance and energy production. Impairment of both these pathways can evoke oxidative damage on mitochondria and key proteins linked to several hallmarks of AD. Glycation is also another type of post-translational modification often reported in AD, which might impair the function of proteins that participate in metabolic pathways thought to be involved in this illness. Despite needing further research, therapies based on insulin treatment, usage of anti-diabetes drugs or some form of dietary intervention, have shown to be promising therapeutic approaches for AD in its early stages of progression and will be unveiled in this paper.

Introduction

Alzheimer’s disease (AD) is a progressive neurological condition that gradually deteriorates memory and learning capabilities through neuronal degeneration. It is the most common form of dementia, currently affecting 20 to 30 million people worldwide [1]. By 2050, it is estimated that around 136 million people will be afflicted by some form of dementia, making it a global health and economical challenge [2].

Major hallmarks of this illness are formation and accumulation of senile plaques, mostly made of amyloid-β (Aβ) and dystrophic neurites, and neurofibrillary tangles (composed primarily by hyperphosphorylated tau protein). Aβ plaques are primarily comprised of Aβ with 40 or 42 (Aβ42) amino acids, which are two by-products of amyloid precursor protein (APP) metabolism [3]. The Aβ42 peptide is the most abundant isoform due to its higher fibrillization rate and insolubility [4]. An imbalance between Aβ production and its clearance is a trigger for AD’s pathological process. Tau is a major microtubule-associated protein (MAP), and it interacts with tubulin to promote microtubules assembly and network stabilization. In AD, hyperphosphorylated tau loses the ability to bind tubulin and thus disrupts microtubule assembly. This toxic property involves the sequestration of normal tau protein and other MAPs, which impairs their normal function [5].

One of the major genetic risk factors associated with sporadic AD is the allele ε4 pertaining to the apolipoprotein E (APOE) gene. Its major roles are related to lipid transport and cholesterol homeostasis [6]. In addition, APOE protein also mediates other processes like synapse formation, modulation of neurite outgrowth, synaptic plasticity, destabilization of microtubules, and Aβ clearance [7]. APOE has three isoforms: APOE2, APOE3 and APOE4. Carriers of APOE4 are shown to be at higher risk of developing AD. Among the three isoforms, APOE4 is primarily responsible for increasing cholesterol levels in the brain. This isoform was shown to be hypolipidated and less effective than APOE3 in inducing cholesterol efflux, thus implying that the pathological effects of APOE4 could be related to lipid metabolism. APOE2 carriers on the other hand tend to exhibit lower cholesterol levels due to the reduced binding affinity for low-density lipoprotein [6].

Another important feature of this illness is neuroinflammation. It entails the activation of different types of brain immune cells like microglia and astrocytes [8,9]. It is largely mediated by redox events both at molecular (e.g., nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), inflammasome) and cellular level (e.g., inflammatory signals transmitted to neurons by astrocytes, via oxidants like H2O2) [10]. It is assumed that the inflammatory responses are driven by a positive feedback-loop relationship between astrocytes and microglia. Both cells types can amplify each other’s inflammatory responses, worsening the neurotoxic environment and promoting neurodegeneration [10]. Aβ is able to activate both microglia and astrocytes, inducing the production of H2O2, nitric oxide, prostaglandins, pro-inflammatory cytokines and chemokines, which can lead to neuronal death [10,11]. However, despite this, it is important to emphasize that a pro-inflammatory environment is required for the activation of microglial cells that are able to clear Aβ and even support neurogenesis after neuronal damage [12]. During an inflammatory process, active microglia exhibit a shift in their cellular metabolism from oxidative phosphorylation to aerobic glycolysis. This is known as the Warburg effect [11]. Despite being less effective mechanism to generate adenosine triphosphate (ATP) than mitochondrial respiration, glycolysis is much faster than oxidative phosphorylation (OXPHOS), allowing high energy processes like proliferation, migration, cytokine secretion and phagocytosis to occur. A study conducted by Baik et al. [13] showed that Aβ induced microglial activation by shifting their metabolism toward aerobic glycolysis. This activation was dependent on glycolysis and the mammalian target of rapamycin (mTOR)- hypoxia-inducible factor (HIF)-1α pathway. However, after prolonged exposure to Aβ, microglia became tolerant and exhibited defects in their cellular metabolism, involving both glycolysis and OXPHOS, as well as diminished inflammatory responses. They also reported that treatment with interferon-γ was able to restore phagocytic activity to tolerant microglia, as well as reduce Aβ plaque accumulation and neuronal loss. This alludes to the conclusion that metabolic modulation of microglia phenotypes could reduce the extent of neuronal damage.

Mitochondrial dysfunction, endoplasmic reticulum (ER) stress and abnormal calcium influxes are also very prominent events observed in this pathology and they are very closely related [14]. Oxidative damage to DNA can interfere with gene transcription and proper promoter function, hindering the transcription of essential genes and generating mutations. Mitochondrial DNA can also be affected, which contributes to energy production impairment and mitochondrial dysfunction [15]. In turn, this leads to a decrease in ATP production and dysfunctional ion gradient maintenance, hindering neurotransmission [16]. Loss of ion gradients also leads to higher intracellular calcium levels, which reduces the buffering ability of the ER and mitochondria. This causes stress to both organelles and further aggravates synaptic dysfunction, causing neuronal death. High levels of intraneuronal free calcium also lead to loss of fidelity of microtubule assembly, consequently, reducing anterograde and retrograde transport of mitochondria and neurotransmitter vesicles [17].

Currently, AD treatment is based on symptom management using cholinesterase inhibitors, N-methyl-D-aspartate antagonists and combination therapy. However, these drugs exert weak beneficial effects on cognitive function, only providing some relief of the behavioural and psychological symptoms of dementia [18]. Several biomarkers are used for AD diagnosis, namely Aβ42, normal and phosphorylated tau in cerebrospinal fluid, APOE genotype and brain imaging techniques [19,20]. It is known that individuals who will eventually develop AD, exhibit pathological signs almost two decades before the onset of clinical symptoms. It follows that current therapy may begin too late for the reversal of AD’s hallmarks. Further research on how to achieve an earlier diagnosis is required to develop new approaches in order to slow, or preferably halt, AD progression [16]. In this paper we sought to review the potential therapeutic effect of metabolic strategies.

Section snippets

Metabolism and energy production in AD

With age, neuronal glucose uptake and proper mitochondrial function decline, reducing the electron transport chain’s activity and promoting an increase in oxidants production [10]. It is stated that these events occur decades before the onset of clinical symptoms of AD, as it was seen in a study performed on young adults at high risk of developing AD [21].

The brain’s main source of energy is glucose. It is used to produce ATP via OXPHOS (i.e., glycolysis, the tricarboxylic acid (TCA) cycle and

Metabolic therapies for AD

The research done in order to find a suitable therapy for AD has been extensive and covering multiple aspects of its pathological features. However, the current treatment for AD simply ameliorates symptoms and modestly slows down cognitive and functional decline. Research has been centred on tackling Aβ accumulation and tau hyperphosphorylation and has yielded few promising results. The growing interest in the link between T2D and AD has promoted investigations on the effect of anti-diabetes

Conclusion

Neurodegenerative diseases like AD are becoming much more prevalent as life expectancy worldwide increases. Despite all the intensive research done on the subject, so many aspects of its pathophysiology, genetic factors and biochemical mechanisms that trigger and promote its development remain unknown. As more research is conducted, we are beginning to realize that AD shares several pathological features with some metabolic disorders. This would imply that the underlying mechanism might be a

Funding

This work was financed by the European Regional Development Fund (ERDF), through the Centro 2020 Regional Operational Programme under project CENTRO-01-0145-FEDER-000012 (HealthyAging2020) and through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalisation and Portuguese national funds via FCT – Fundação para a Ciência e a Tecnologia, under project[s] POCI-01-0145-FEDER-029369 and UID/NEU/04539/2019.

References (147)

  • C. Reitz et al.

    Epidemiology of Alzheimer disease

    Nat. Rev. Neurol.

    (2011)
  • J.B. Toledo et al.

    Metabolic network failures in Alzheimer’s disease: A biochemical road map

    Alzheimers Dement.

    (2017)
  • C.A. Lane et al.

    Alzheimer’s disease

    Eur. J. Neurol.

    (2018)
  • J.T. Jarrett et al.

    The carboxy terminus of the .beta. amyloid protein is critical for the seeding of amyloid formation: Implications for the pathogenesis of Alzheimer’s disease

    Biochemistry

    (1993)
  • K. Iqbal et al.

    Tau in Alzheimer Disease and Related Tauopathies

    CAR.

    (2010)
  • M. Safieh et al.

    ApoE4: an emerging therapeutic target for Alzheimer’s disease

    BMC Med.

    (2019)
  • H.C. Hunsberger et al.

    The role of APOE4 in Alzheimer’s disease: strategies for future therapeutic interventions

    Neuronal Signaling.

    (2019)
  • E. Colombo et al.

    Astrocytes: Key Regulators of Neuroinflammation

    Trends Immunol.

    (2016)
  • A. Fernandes et al.

    Microglia and inflammation: conspiracy, controversy or control?

    Cell. Mol. Life Sci.

    (2014)
  • F. Yin et al.

    Energy metabolism and inflammation in brain aging and Alzheimer’s disease

    Free Radic. Biol. Med.

    (2016)
  • C. Lauro et al.

    Metabolic Reprograming of Microglia in the Regulation of the Innate Inflammatory Response

    Front. Immunol.

    (2020)
  • A.Y. Lai et al.

    Clearance of amyloid-β peptides by microglia and macrophages: the issue of what, when and where

    Future Neurol.

    (2012)
  • S.H. Baik et al.

    A Breakdown in Metabolic Reprogramming Causes Microglia Dysfunction in Alzheimer’s Disease

    Cell Metab.

    (2019)
  • E. Area-Gomez et al.

    Upregulated function of mitochondria-associated ER membranes in Alzheimer disease: Upregulated function of MAM in AD

    EMBO J.

    (2012)
  • R.X. Santos et al.

    Nuclear and mitochondrial DNA oxidation in Alzheimer’s disease

    Free Radic. Res.

    (2012)
  • D.A. Butterfield et al.

    Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease

    Nat. Rev. Neurosci.

    (2019)
  • I. Bezprozvanny et al.

    Neuronal calcium mishandling and the pathogenesis of Alzheimer’s disease

    Trends Neurosci.

    (2008)
  • K.G. Yiannopoulou, S.G. Papageorgiou, Current and future treatments for Alzheimer’s disease, Ther. Adv. Neurol. Disord....
  • S.C. Thomas et al.

    Brain metabolism and Alzheimer’s disease: The prospect of a metabolite-based therapy

    J. Nutr. Health Aging

    (2015)
  • L. Hertz et al.

    Effects of ketone bodies in Alzheimer’s disease in relation to neural hypometabolism, β-amyloid toxicity, and astrocyte function

    J. Neurochem.

    (2015)
  • E.M. Reiman et al.

    Functional brain abnormalities in young adults at genetic risk for late-onset Alzheimer’s dementia

    Proc. Natl. Acad. Sci.

    (2004)
  • S. Cunnane et al.

    Brain fuel metabolism, aging, and Alzheimer’s disease

    Nutrition.

    (2011)
  • Y. Peng et al.

    Central and Peripheral Metabolic Defects Contribute to the Pathogenesis of Alzheimer’s Disease: Targeting Mitochondria for Diagnosis and Prevention

    Antioxid. Redox Signal.

    (2020)
  • R.J. Bateman et al.

    Clinical and Biomarker Changes in Dominantly Inherited Alzheimer’s Disease

    N. Engl. J. Med.

    (2012)
  • M. Grabacka et al.

    Regulation of Ketone Body Metabolism and the Role of PPARα

    IJMS.

    (2016)
  • M. Ogawa et al.

    Altered energy metabolism in Alzheimer’s disease

    J. Neurol. Sci.

    (1996)
  • U. Lying-Tunell et al.

    Cerebral blood flow and metabolic rate of oxygen, glucose, lactate, pyruvate, ketone bodies and amino acids: II. PRESENILE DEMENTIA AND NORMAL-PRESSURE HYDROCEPHALUS

    Acta Neurol. Scand.

    (2009)
  • L. Szablewski

    Glucose Transporters in Brain: In Health and in Alzheimer’s Disease

    JAD.

    (2016)
  • L.F. Barros et al.

    Near-critical GLUT1 and Neurodegeneration: Glucose Transport and Neurodegeneration

    J. Neurosci. Res.

    (2017)
  • Y. Benomar et al.

    Insulin and Leptin Induce Glut4 Plasma Membrane Translocation and Glucose Uptake in a Human Neuronal Cell Line by a Phosphatidylinositol 3-Kinase- Dependent Mechanism

    Endocrinology.

    (2006)
  • J. Apelt, G. Mehlhorn, R. Schliebs, Insulinsensitive GLUT4 glucose transporters are colocalized with GLUT3expressing...
  • C.A. Grillo et al.

    Insulin-stimulated translocation of GLUT4 to the plasma membrane in rat hippocampus is PI3-kinase dependent

    Brain Res.

    (2009)
  • G. Ashrafi et al.

    GLUT4 Mobilization Supports Energetic Demands of Active Synapses

    Neuron

    (2017)
  • M. Piert et al.

    Diminished glucose transport and phosphorylation in Alzheimer’s disease determined by dynamic FDG-PET

    J. Nucl. Med.

    (1996)
  • E.A. Winkler et al.

    GLUT1 reductions exacerbate Alzheimer’s disease vasculo-neuronal dysfunction and degeneration

    Nat. Neurosci.

    (2015)
  • T. Niccoli et al.

    Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila

    Curr. Biol.

    (2016)
  • I.A. Simpson et al.

    Decreased concentrations of GLUT1 and GLUT3 glucose transporters in the brains of patients with Alzheimer’s disease

    Ann. Neurol.

    (1994)
  • Y. Liu et al.

    Decreased glucose transporters correlate to abnormal hyperphosphorylation of tau in Alzheimer disease

    FEBS Lett.

    (2008)
  • Y. Huang et al.

    Normal glucose uptake in the brain and heart requires an endothelial cell-specific HIF-1 -dependent function

    Proc. Natl. Acad. Sci.

    (2012)
  • J. Pearson-Leary et al.

    Intrahippocampal Administration of Amyloid-β1-42 Oligomers Acutely Impairs Spatial Working Memory, Insulin Signaling, and Hippocampal Metabolism

    JAD.

    (2012)
  • T.S. Pinho et al.

    Diminished O-GlcNAcylation in Alzheimer’s disease is strongly correlated with mitochondrial anomalies

    Biochim. Biophys. Acta (BBA) - Mol. Basis Dis.

    (2019)
  • S. Wang et al.

    Quantitative proteomics identifies altered O-GlcNAcylation of structural, synaptic and memory-associated proteins in Alzheimer’s disease: Brain protein O-GlcNAcylation in Alzheimer’s disease

    J. Pathol.

    (2017)
  • A.C. Wang et al.

    Loss of O -GlcNAc glycosylation in forebrain excitatory neurons induces neurodegeneration

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

    (2016)
  • F. Liu et al.

    Involvement of aberrant glycosylation in phosphorylation of tau by cdk5 and GSK-3β

    FEBS Lett.

    (2002)
  • Y.S. Chun et al.

    O-GlcNAcylation Promotes Non-Amyloidogenic Processing of Amyloid-β Protein Precursor via Inhibition of Endocytosis from the Plasma Membrane

    JAD.

    (2015)
  • Y.S. Chun et al.

    O-GlcNAcylation of amyloid-β precursor protein at threonine 576 residue regulates trafficking and processing

    Biochem. Biophys. Res. Commun.

    (2017)
  • C. Kim et al.

    O-linked β-N-acetylglucosaminidase inhibitor attenuates β-amyloid plaque and rescues memory impairment

    Neurobiol. Aging

    (2013)
  • I.-H. Ryu et al.

    Aβ-affected pathogenic induction of S-nitrosylation of OGT and identification of Cys-NO linkage triplet

    Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics

    (2016)
  • P. Gkogkolou et al.

    Advanced glycation end products: Key players in skin aging?

    Dermato-Endocrinology.

    (2012)
  • V.V. Shuvaev et al.

    Increased protein glycation in cerebrospinal fluid of Alzheimer’s disease

    Neurobiol. Aging

    (2001)
  • Cited by (0)

    View full text