Elsevier

Journal of Affective Disorders

Volume 168, 15 October 2014, Pages 167-183
Journal of Affective Disorders

Review
A systematic review of amyloid-β peptides as putative mediators of the association between affective disorders and Alzheimer׳s disease

https://doi.org/10.1016/j.jad.2014.06.050Get rights and content

Abstract

Background

Affective disorders are associated with an increased occurrence of cognitive deficits and have been linked to cognitive impairment and Alzheimer׳s disease. The putative molecular mechanisms involved in these associations are however not clear. The aim of this systematic review was to explore clinically founded evidence for amyloid-β peptides in cerebrospinal fluid and blood as putative biomarkers for affective disorders.

Method

Systematic searches in Embase and PubMed databases yielded 23 eligible, observational studies.

Results

Despite inconsistencies that were partly ascribed to the application of different assay formats, study results indicate a potentially altered amyloid-β metabolism in affective disorder.

Limitations

Since most studies used a cross-sectional design, causality is difficult to establish. Moreover, methodological rigor of included studies varied and several studies were limited by very low sample numbers. Finally, different assays for amyloid-β were utilized in the different studies, thus hampering comparisons.

Conclusion

To unravel possible risk relations and causalities between affective disorder and Alzheimer׳s disease and to determine how amyloid-β concentrations change over time and are associated with cognition as well as affective symptomatology, future research should include prospective, longitudinal studies, implemented in large study populations, where peripheral and central amyloid-β ratios are quantified concomitantly and continuously across various affective phases. Also, to enable inter-survey comparisons, the use of standardized pre-analytical/analytical procedures is crucial.

Introduction

Affective disorders are incapacitating, highly prevalent and substantial contributors to the global burden of disease with an estimated lifetime prevalence of approximately 12% (Kessler et al., 2009). Empirically and epidemiologically founded evidence indicate significant heritability (Shih et al., 2004), high rates of non-recovery and recurrence that progressively enhance with each successive episode (Solomon et al., 2000) increased morbidity and mortality (Lauersen et al., 2007) a lifelong elevated risk of suicide (Rihmer, 2007), compromised psychosocial functioning and greatly reduced productivity that incurs immense healthcare costs to the society (Simon, 2003). Furthermore affective disorders are frequently associated with medical comorbidity and long-term population-based studies indicate that patients suffering from these mental illnesses also display an increased risk of developing dementia (Cruess et al., 2003, Musselman et al., 1998, Ownby et al., 2006, Spiegel and Giese-Davis, 2003).

Mood disorders are accompanied by cognitive abnormalities during depressive/manic episodes, but cognitive impairment is also prevalent among patients in remission, specifically within executive function and verbal memory (Bora et al., 2013, Robinson et al., 2006). Cognitive deficits have been linked to the number, duration and intensity of affective episodes and to the presence of mania or psychotic symptoms (Gorwood et al., 2008, Martinez-Aran et al., 2008, Zubieta et al., 2001), so that mood disorders in general would be expected to aggravate and accelerate the process of aging-related cognitive deterioration (Gualtieri and Johnson, 2008) and perhaps via synergistically interconnected pathophysiological processes eventually facilitate conversion to dementia.

Alzheimer׳s disease (AD) is the leading cause of progressive dementia. The histopathological hallmarks of AD, which begin decades prior to its clinical expression (Villemagne et al., 2013) encompass characteristic amyloid-β (Aβ) plaques derived from the larger amyloid precursor protein (APP), neurofibrillary tangles comprising a hyperphosphorylated form of tau proteins and neurodegeneration affecting several neurotransmitter systems (Blennow and Hampel, 2003). APP, if processed by the amyloidogenic pathway involving β- and γ-secretases, generates Aβ40, which is primarily associated with cerebral amyloid angiopathy, and Aβ42 – representing the major component of amyloid deposits (Blennow and Hampel, 2003). Amyloid markers can be measured in both cerebrospinal fluid (CSF) and in blood. While no consensus has been reached regarding plasma profiles, decreased levels of Aβ42 peptide combined with elevated tau (t-tau) and phosphorylated tau (p-tau) in CSF are sensitive and specific markers for AD and incipient AD (Blennow and Hampel, 2003, Zetterberg, 2008). To characterize the nature of cognitive impairment in affective disorder, several studies have measured these AD-related markers, both peripherally as well as in CSF. While tau markers are generally unchanged, Aβ42 shows significant alterations in several studies. To evaluate the strength of evidence for the association of Aβ alterations this review critically and systematically assesses studies that have measured Aβ markers in plasma/serum or CSF in samples from patients with affective disorder.

Section snippets

Search strategy and selection criteria

A systematic search was performed between July and August 2013 via the electronic databases PubMed and EMBASE, so as to identify and evaluate all available literature exploring the association between Aβ and affective disorders. Search profiles using various combinations of pertinent keywords: “Aβ”, “CSF”, “plasma”, “serum”, “depression”, “major depressive disorder”, “bipolar disorder”, “mood disorders”, “affective disorders” and “psychiatric disease”, were employed. Rigorous abstract

Population characteristics and study design

A total of 23 eligible, English-language articles (Baba et al., 2012, Blasko et al., 2006, Blasko et al., 2010, Bibl et al., 2007, Direk et al., 2013, Gudmundsson et al., 2007, Hertze et al., 2010, Hock et al., 1998, Jakobsson et al., 2013, Jensen et al., 1999, Kramberger et al., 2012, Metti et al., 2013, Moon et al., 2011, Piccinni et al., 2012, Pomara et al., 2006, Pomara et al., 2012, Portelius et al., 2010, Post et al., 2006, Reis et al., 2012, Schröder et al., 1997, Sjögren et al., 2000,

Longitudinal studies

One (Direk et al., 2013) of the four longitudinal studies included in this review, was considered of high research quality, whereas the remaining three (Blasko et al., 2010, Hertze et al., 2010, Metti et al., 2013) were of moderate quality. Methodological limitations/flaws included non-random sampling strategies (Hertze et al., 2010) missing Aβ40 measurements (Blasko et al., 2010) the use of non-diagnostic assessment tools for depression (Direk et al., 2013, Metti et al., 2013) and increased

Future research perspectives

Overall, results drawn from the 23 studies included in this review indicate that Aβ metabolism may be altered in patients with affective disorder but far from settles the issue. Thus, the exact correlation between AD and affective disorder remains elusive despite a clear clinico-epidemiological evidence of an increased incidence of cognitive impairment in patients suffering from affective disorders. Future research requires prospective, longitudinal studies, implemented in large study

Role of funding source

There were no funding sources in the writing of the manuscript or the decision to submit it for publication.

Conflict of interest

The authors have no conflicts of interest.

Acknowledgment

None.

References (150)

  • N. Fullwood et al.

    Plasma amyloid-beta concentrations in Alzheimer׳s disease: an alternative hypothesis

    Lancet Neurol.

    (2006)
  • J. Ghiso et al.

    Systemic catabolism of Alzheimer׳s Abeta40 and Abeta42

    J. Biol. Chem.

    (2004)
  • V. Giedraitis et al.

    The normal equilibrium between CSF and plasma amyloid beta levels is disrupted in Alzheimer׳s disease

    Neurosci. Lett.

    (2007)
  • P. Gudmundsson et al.

    The relationship between cerebrospinal fluid biomarkers and depression in elderly women

    Am. J. Geriatr. Psychiatry

    (2007)
  • A. Hofman et al.

    Atherosclerosis, apolipoprotein E, and prevalence of dementia an Alzheimer׳s disease in the Rotterdam study

    Lancet

    (1997)
  • N. Iwata et al.

    Metabolism of amyloid-β peptide and Alzheimer׳s disease

    Pharmacol. Ther.

    (2005)
  • F. Kapczinski et al.

    Peripheral biomarkers and illness activity in bipolar disorder

    J. Pscyhiatr. Res.

    (2011)
  • Y.M. Kuo et al.

    High levels of circulating Aβ42 are sequestered by plasma proteins in Alzheimer׳s disease

    Biochem. Biophys. Res. Commun.

    (1999)
  • Y.M. Kuo et al.

    Amyloid-β peptides interact with plasma proteins and erythrocytes: implications for their quantification in plasma

    Biochem. Biophys. Res. Commun.

    (2000)
  • Y. Liu et al.

    Interleukin(IL)-6, tumor necrosis factor alpha (TNF-α) and soluble interleukin-2 receptors (sIL-2R) are elevated in patients with major depressive disorder: a meta-analysis and metaregression

    J. Affect. Disord.

    (2012)
  • K. Madsen et al.

    Lack of association between prior depressive episodes and cerebral (11C)PiB binding

    Neurobiol. Aging

    (2012)
  • M. Maes et al.

    A review on the oxidative and nitrosative stress (O&NS) pathways in major depression and their possible contribution to the (neuro)degenerative processes in that illness

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2011)
  • F. Mangialasche et al.

    Biomarkers of oxidative and nitrosative damage in Alzheimer׳s disease and mild cognitive impairment

    Ageing Res. Rev.

    (2009)
  • S. Marco et al.

    Amyloid β-peptide1–42 alters tight junction protein distribution and expression in brain microvessel endothelial cells

    Neurosci. Lett.

    (2006)
  • Y.S. Moon et al.

    The correlation of plasma Aβ42 levels, depressive symptoms, and cognitive function in the Korean elderly

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2011)
  • R.E. Mrak et al.

    Glial cytokines in Alzheimer׳s disease: review and pathogenic implications

    Hum. Pathol.

    (1995)
  • Y. Namekawa et al.

    Heterogeneity of elderly depression: Increased risk of Alzheimer׳s disease and Aβ protein metabolism

    Prog. Neuro-Psychopharmacol. Biol. Psychiatry

    (2013)
  • R.L. Nelson et al.

    Prophylactic treatment with paroxetine ameliorates behavioural deficits and retards the development of amyloid and tau pathologies in 3xTgAD mice

    Exp. Neurol.

    (2007)
  • T.W.W. Pace et al.

    Cytokine-effects on glucocorticoid receptor function: relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression

    Brain Behav. Immun.

    (2007)
  • M. Pakaski et al.

    Imipramine and citalopram facilitate amyloid precursor protein secretion in vitro

    Neurochem. Int.

    (2005)
  • A. Piccinni et al.

    Plasma β-amyloid peptide levels: a pilot study in bipolar depressed patients

    J Affect. Disord.

    (2012)
  • N. Pomara et al.

    Does increased platelet release of Abeta peptide contribute to brain abnormalities in individuals with depression?

    Med. Hypotheses

    (2003)
  • N. Adab et al.

    Common antiepileptic drugs in pregnancy in women with epilepsy

    Cochrane Database Syst. Rev.

    (2004)
  • H. Baba et al.

    Metabolism of amyloid-β protein may be affected in depression

    J. Clin. Psychiatry

    (2012)
  • C. Bell et al.

    Tryptophan depletion and its implications for psychiatry

    Br. J. Psychiatry

    (2001)
  • G.E. Berrios et al.

    Depressive illness in multiple sclerosis: clinical and theoretical aspects of the association

    Br. J. Psychiatry

    (1990)
  • J.L. Beyer et al.

    Hyperintense MRI lesions in bipolar disorder: a meta-analysis and review

    Int. Rev. Psychiatry

    (2009)
  • M. Bibl et al.

    Cerebrospinal fluid amyloid β peptide patterns in Alzheimer׳s disease patients and non-demented controls depend on sample pretreatment: indication of carrier-mediated epitope masking of amyloid beta peptides

    Electrophoresis

    (2004)
  • M. Bibl et al.

    Blood-based neurochemical diagnosis of vascular dementia: a pilot study

    J. Neurochem.

    (2007)
  • M. Bibl et al.

    Stability of amyloid-β peptides in plasma and serum

    Electrophoresis

    (2012)
  • J.G. Biessels et al.

    Risk of dementia in diabetes mellitus: a systematic review

    Lancet Neurol.

    (2006)
  • I. Blasko et al.

    Measurement of thirteen biological markers in CSF of patients with Alzheimer׳s disease and other dementias

    Dement. Geriatr. Cognit. Disord.

    (2006)
  • E. Bora et al.

    Cognitive impairment in euthymic major depressive disorder: a meta-analysis

    Psychol. Med.

    (2013)
  • K. Bordji et al.

    Synapses, NMDA receptor activity and neuronal Aβ production in Alzheimer׳s disease

    Rev. Neurosci.

    (2011)
  • M.A. Butters et al.

    Imaging Alzheimer pathology in late-life depression with PET and Pittsburgh Compound-B

    Alzheimer Dis. Assoc. Disord.

    (2008)
  • D. Carnevale et al.

    Hypertension induces brain β-amyloid accumulation, cognitive impairment, and memory deterioration through activation of receptor for advanced glycation end products in brain vasculature

    Hypertension

    (2012)
  • N. Catteruccia et al.

    Ultrastructural localization of the putative precursors of the A4 amyloid protein associated with Alzheimer׳s disease

    Am. J. Pathol.

    (1990)
  • L. Chami et al.

    BACE1 is at the crossroad of a toxic vicous cycle involving cellular stress and β-amyloid production in Alzheimer׳s disease

    Mol. Neurodegener.

    (2012)
  • S. Champaneri et al.

    Biological basis of depression in adults with diabetes

    Curr. Diabetes Rep.

    (2010)
  • S.A. Cosentino et al.

    Plasma β-amyloid and cognitive decline

    Arch. Neurol.

    (2010)
  • Cited by (13)

    • Alzheimer's disease related biomarkers in bipolar disorder – A longitudinal one-year case-control study

      2022, Journal of Affective Disorders
      Citation Excerpt :

      The pathological process has not yet been fully clarified, although dysfunctional clearance of Aβ from the brain appears to be integral to disease development (Versele et al., 2020). As a possible link between AD and affective disorders pathophysiology, several studies have investigated CSF-Aβ42 and plasma-Aβ42 levels in patients suffering from major depression, as recently reviewed (Abbasowa and Heegaard, 2014). Despite inconsistencies that are partly ascribed to the application of different assay formats (Mattsson et al., 2013), study results indicate a potentially altered Aβ metabolism in affective disorder.

    • Rethinking IRPs/IRE system in neurodegenerative disorders: Looking beyond iron metabolism

      2022, Ageing Research Reviews
      Citation Excerpt :

      However, whether the above two aspects exist independently or in series with each other has not been well explained (Piñero, 2000). APP is a widely distributed single-pass transmembrane protein, which can produce toxic β-amyloid protein and cause neurotoxicity after protease cleavage (Abbasowa and Heegaard, 2014; Kayed et al., 2011). IRE was also found in the 5’UTR of the APP.

    • Overlapping mechanisms linking insulin resistance with cognition and neuroprogression in bipolar disorder

      2020, Neuroscience and Biobehavioral Reviews
      Citation Excerpt :

      A smaller study by Piccini et al., on the other hand, found the opposite trend with lower Aβ42/ Aβ40 ratios in plasma (Piccinni et al., 2012). Possible confounding factors including study design and instrument use, as well as a review of Aβ in BD and depression, are explained in Abbasowa et al. (Abbasowa and Heegaard, 2014). Post-mortem autopsy studies by Shioya et al. found a variety of neuropathological markers (amyloid, Lewy bodies, tau) in six out of eleven brains from BD patients using immunohistochemistry (Shioya et al., 2015).

    • Depression related cerebral pathology and its relationship with cognitive functioning: A systematic review

      2019, Journal of Affective Disorders
      Citation Excerpt :

      Alternatively, LOD is characterised by greater overall levels of cognitive impairment, as well as greater subcortical atrophy, vascular and amyloid burden (Herrmann et al., 2008; Choi et al., 2017). To date, current systematic reviews have only focused on singular aspects of either cognition or cerebral pathology, or relied on clinical endpoints and not cognitive assessment, thereby overlooking the complex and interrelated association between depression and these constructs (Abbasowa and Heegaard, 2014; Herbert and Lucassen, 2016). Therefore, this review will synthesise the available literature regarding depression related cerebral pathologies, and the associated neurocognitive impairment.

    View all citing articles on Scopus
    View full text