You are on page 1of 8

European Journal of Pharmacology 626 (2010) 64–71

Contents lists available at ScienceDirect

European Journal of Pharmacology


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / e j p h a r

Review

Depression and Alzheimer's disease: Neurobiological links and common


pharmacological targets
Filippo Caraci a, Agata Copani a,b, Ferdinando Nicoletti c,d, Filippo Drago e,⁎
a
Department of Pharmaceutical Sciences, University of Catania, 95125, Catania, Italy
b
I.B.B., CNR-Catania, 95125, Catania, Italy
c
I.N.M. Neuromed, Località Camerelle, 86077, Pozzilli, Italy
d
Department of Human Physiology and Pharmacology, University of Rome La Sapienza, 00185 Rome, Italy
e
Department of Experimental and Clinical Pharmacology, University of Catania, 95125, Catania, Italy

a r t i c l e i n f o a b s t r a c t

Article history: Depression is one of the most prevalent and life-threatening forms of mental illnesses, whereas Alzheimer's
Accepted 9 October 2009 disease is a neurodegenerative disorder that affects more than 37 million people worldwide. Recent evidence
Available online 18 October 2009 suggests a strong relationship between depression and Alzheimer's disease. A lifetime history of major
depression has been considered as a risk factor for later development of Alzheimer's disease. The presence of
Keywords: depressive symptoms can affect the conversion of mild cognitive impairment into Alzheimer's disease.
Major depression
Neuritic plaques and neurofibrillary tangles, the two major hallmarks of Alzheimer's disease brain, are more
Alzheimer's disease
Chronic inflammation
pronounced in the brains of Alzheimer's disease patients with comorbid depression as compared with
β-amyloid Alzheimer's disease patients without depression. On the other hand, neurodegenerative phenomena have
Transforming-growth-factor-β1 been observed in different brain regions of patients with a history of depression. Recent evidence suggests
Brain-derived neurotrophic factor that molecular mechanisms and cascades that underlie the pathogenesis of major depression, such as chronic
Neuroprotection inflammation and hyperactivation of hypothalamic–pituitary–adrenal (HPA) axis, are also involved in the
pathogenesis of Alzheimer's disease. In particular, a specific impairment in the signaling of some
neurotrophins such as transforming-growth-factor β1 (TGF-β1) and brain-derived neurotrophic factor
(BDNF) has been observed both in depression and Alzheimer's disease. In the present review we will
examine the evidence on the common molecular pathways between depression and Alzheimer's disease and
we will discuss these pathways as new pharmacological targets for the treatment of both major depression
and Alzheimer's disease.
© 2009 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
2. The connection between depression and dementia: epidemiological evidence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65
3. The neurobiological links between depression and Alzheimer's disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
3.1. Stress and dysfunction of the hypothalamic–pituitary–adrenal axis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
3.2. Chronic inflammation in depression and Alzheimer's disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 66
3.3. Impairment of neurotrophin signalling in depression and Alzheimer's disease: the role of BDNF and TGF-β1 . . . . . . . . . . . . . . 67
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69

1. Introduction

Depression is one of the most prevalent and life-threatening forms


of mental illnesses and a major cause of morbidity worldwide.
⁎ Corresponding author. Department of Experimental and Clinical Pharmacology,
University of Catania, Viale Andrea Doria 6, 95125, Catania, Italy. Tel.: +39 095
Estimates of prevalence range from 5 to 20% of the general population,
7384236; fax: +39 095 7384238. when people with mild depressive episodes are included (Fava and
E-mail address: fdrago1@netzero.net (F. Drago). Kendler, 2000; Kessler et al., 2005). Depression is associated with

0014-2999/$ – see front matter © 2009 Elsevier B.V. All rights reserved.
doi:10.1016/j.ejphar.2009.10.022
F. Caraci et al. / European Journal of Pharmacology 626 (2010) 64–71 65

significant disability (Murray and Lopez, 1997) and with excess


mortality (Cuijpers and Smit, 2002) particularly from cardiovascular
disease (Rivelli and Jiang, 2007).
Current antidepressant drugs, which are directed against mono-
aminergic systems, provide a useful therapeutic tool, but approxi-
mately 30% of depressed patients failed to respond to these drugs
and antidepressants produce remission only in 30% of patients. This
can be explained by the fact that the pathophysiology of depression
has not been completely elucidated, and treatments have been
developed following the “monoaminergic hypothesis” of depression
without taking into account other mechanisms involved in the
pathophysiology of the disorder. The identification of these mechan-
isms represents a fundamental step for the identification of new
targets and the design of new antidepressant drugs. Recent studies
suggest that factors other than monoamine deficiency or imbalances
between various neurotransmitter systems must be considered when
describing the neurobiological basis of major depression, such as
alterations in mechanisms to resilience to stress, reduced synaptic
plasticity, impaired adult hippocampal neurogenesis and neurode-
generative phenomena in the hippocampus (Krishnan and Nestler, Fig. 1. From depression to Alzheimer's disease: neurobiological links and hypothetical
pathophysiological mechanisms. Dysfunction of hypothalamic–pituitary–adrenal (HPA)
2008).
axis, chronic inflammation and deficit of neurotrophin signaling exert a central role both in
Neurodegeneration is also the main histopathological feature of the pathophysiology of depression and Alzheimer's disease by increasing levels of
Alzheimer's disease, a devastating disorder affecting more than 37 glucocorticoids and pro-inflammatory cytokines and also reducing brain-derived
million people worldwide. It is mainly characterized by memory loss, neurotrophic factor (BDNF), and transforming-growth-factor-β1 (TGF-β1) levels. These
with disoriented behaviour and impairments in language, compre- alterations might lead to an increased vulnerability to β-amyloid toxicity and hippocampal
atrophy, thus favouring the onset of cognitive deficit and finally the progression from
hension, and spatial skills. Neuropsychiatric symptoms, such as
depression to Alzheimer's disease.
depression, psychosis and agitation are also frequent in people with
Alzheimer's disease, and are a common precipitant of institutional
care (Ballard et al., 2008). Current drugs for Alzheimer's disease are
only symptomatic, but do not interfere with the underlying molecular pathways as new pharmacological targets for the treatment
pathogenic mechanisms of the disease (Klafki et al., 2006). Therefore, of both depression and Alzheimer's disease.
recent efforts in Alzheimer's disease research are now directed to the
identification of new molecular targets for the development of 2. The connection between depression and dementia:
disease-modifying drugs able to counteract the degenerative pro- epidemiological evidence
cesses and the resulting memory loss in Alzheimer's disease patients.
Alzheimer's disease is characterized by the presence of ß-amyloid Different studies suggest that depressive disorder is associated
in the senile plaques, intracellular aggregates of tau protein in the with increased risk of developing cognitive dysfunction (Kessing,
neurofibrillary tangles, and progressive neuronal loss (Hardy and 1998; Castaneda et al., 2008) and eventually dementia, in particular
Selkoe, 2002). Genetic studies and evidence from animal models Alzheimer's disease (Geerlings et al., 2000; Ownby et al., 2006).
support a primary role for ß-amyloid in the cascade of events which Several groups have examined the temporal relationship between
leads to neuronal death in Alzheimer's disease brain (Hardy and depression and Alzheimer's disease to understand whether depression
Selkoe, 2002). Oligomeric species composed of aggregated ß-amyloid is simply a prodromal symptom of Alzheimer's disease which precedes
are believed to exert toxic effects on synaptic and cellular functions, the onset of cognitive deficits, or whether a history of depression might
finally leading to neurodegeneration (Cerpa et al., 2008). Different represent an independent risk factor for the development of
mechanisms have been proposed to explain ß-amyloid neuro- Alzheimer's disease. The systematic meta-analysis from Ownby et al.
toxicity, such as oxidative stress (Butterfield et al., 2007), amplifica- (2006) clearly shows that the interval between diagnoses of
tion of N-methyl-D-aspartate (NMDA) toxicity (Hynd et al., 2004), cell depression and Alzheimer's disease is positively related to an
cycle activation in differentiated neurons (Giovanni et al., 1999; increased risk of developing Alzheimer's disease, suggesting that
Copani et al., 1999; Herrup et al., 2004; Copani et al., 2007), and finally rather than a prodrome, depression can be considered as a risk factor
the sustained loss of the canonical Wnt pathway, a signalling pathway for Alzheimer's disease. Interestingly, depression occurring early in life
essential for maintaining neuronal homeostasis (Caricasole et al., (more than 25 years before the diagnosis of dementia) is associated
2004; Inestrosa et al., 2007). with a late development of Alzheimer's disease (Robert et al., 2003).
Recent evidence, from epidemiology to neurobiology, suggests a Kessing and Andersen (2004) have demonstrated that the risk to
strong relationship between depression and dementia. For different develop Alzheimer's disease increases with every new affective
years depression and dementia have been considered completely episode associated to mood disorders (Kessing, 1998; Kessing and
distinct clinical entities. The term “pseudodementia” is used to define a Andersen, 2004). In particular, the authors found that the rate of
clinical picture characterized by depression associated with cognitive dementia tended to increase by 13% with every episode leading to
impairment and responsiveness to antidepressant treatment, as hospital admission for patients with depressive disorder.
opposed to depression as an early symptom of dementia, which is There is also epidemiological and clinical evidence suggesting a
instead unresponsive to antidepressants (Gualtieri and Johnson, strong relationship between depression and dementia. Late-life
2008). Recent evidence suggests that depression can act as a risk depression is characterized by the onset of a depressive episode
factor for dementia, in particular Alzheimer's disease, and, most after age 50 (Gualtieri and Johnson, 2008). Late-life depression
importantly, common pathophysiological mechanisms between these patients frequently exhibit a significant cognitive impairment
two diseases have been identified, which might explain the progres- (Gallassi et al., 2006). In particular, deficits have been reported in
sion from depression to Alzheimer's disease (Fig. 1). In the present domains of attention, language, episodic recall, semantic recall,
review we will examine this evidence, focusing on the common visuospatial/visuoconstruction, working memory, and executive
66 F. Caraci et al. / European Journal of Pharmacology 626 (2010) 64–71

function (Elderkin-Thompson et al., 2003; Sheline et al., 2006; Köhler disease (Gurevich et al., 1990; Belanoff et al., 2001b). High basal
et al., 2009). Several evidences suggest that patients with late-life cortisol levels are associated with more rapid cognitive decline in
depression are at increased risk to develop dementia (Baldwin et al., Alzheimer's disease patients (Greenwald et al., 1986; Charles et al.,
2006; Thomas and O'Brien, 2008). A recent study has also demon- 1986; Davous, 1987; Carlson et al., 1999), and prednisone treatment
strated an acceleration in age-related cognitive decline in patients causes an impairment of cognition (Aisen et al., 2000).
with depression compared to age-matched normal controls (Gualtieri Excess glucocorticoids may have a central role in the pathophys-
and Johnson, 2008). On the other hand, neurodegenerative phenom- iology of both depression (Sheline et al., 2003; Krishnan and Nestler,
ena have been observed in different brain regions of depressed 2008) and Alzheimer's disease (Landfield et al., 2007; Sotiropoulos
patients, such as the hippocampus (Ballmaier et al., 2008), which is et al., 2008) via multiple mechanisms. High levels of glucocorticoids,
also one of the first brain regions to be affected in Alzheimer's disease through the activation of glucocorticoid receptors, can reduce
brain. Furthermore, the presence of depressive symptoms promote neurogenesis in the hippocampal dentate gyrus (Krishnan and
the conversion of mild cognitive impairment into Alzheimer's disease Nestler, 2008), induce the retraction of hippocampal apical dendrites
(Houde et al., 2008; Modrego and Ferrández, 2004). Houde et al. (Sapolsky, 2000; Magariños and McEwen, 1995; McLaughlin et al.,
(2008) have found that the persistence of depression over two to 2007) and cause neuronal death in hippocampal neurons (Sapolsky,
three years in mild cognitive impairment patients significantly 1986; Yu et al., 2008). In particular, glucocorticoids trigger apoptotic
predicts cognitive deterioration leading to Alzheimer's disease. death in hippocampal neurons via the activation of glucocorticoid
Modrego and Ferrández (2004) have demonstrated that patients receptors (Crochemore et al., 2005), and also can increase neuronal
with mild cognitive impairment and depression are at more than vulnerability to different toxins, such as radical oxygen species (Behl
twice the risk of developing Alzheimer's disease than those without et al., 1997), excitotoxins (Elliott and Sapolsky, 1992) and β-amyloid
depression and, more important, patients with a poor response to (Abraham et al., 2000; Goodman et al., 1996). All these events can
antidepressants are at an increased risk of developing Alzheimer's contribute to the reduction in hippocampal volume found both in
disease. Finally, depression may occur in 30–40% of the Alzheimer's depression and early Alzheimer's disease. Interestingly, in animal
disease patients (Assal and Cummings, 2002; Starkstein et al., 2005) models of Alzheimer's disease stress-level glucocorticoid administra-
and it affects clinical evolution of Alzheimer's disease (Shim and Yang, tion enhances β-amyloid formation by increasing steady-state levels
2006). Alzheimer's disease patients with major depression show a of amyloid precursor protein (APP) and the β-APP cleaving enzyme,
greater and faster cognitive impairment than non depressed patients and also promotes the development of neurofibrillary tangles, a major
(Lyketsos et al., 1997; Milwain and Nagy, 2005; Shim and Yang, 2006), histopathological hallmark of Alzheimer's disease, that strongly
and, interestingly, neuritic plaques and neurofibrillary tangles, the correlates with the clinical onset and severity of dementia (Green
two major hallmarks of Alzheimer's disease brain, are more et al., 2006).
pronounced in the hippocampus of Alzheimer's disease patients All these data demonstrate a central role of HPA dysfunction and
with comorbid depression as compared with Alzheimer's disease high levels of glucocorticoids both in depression and Alzheimer's
patients without depression (Rapp et al., 2008). disease pathogenesis and suggest the possibility that glucocorticoid
receptors might be a suitable target both for antidepressant and
3. The neurobiological links between depression and antidementia drugs.
Alzheimer's disease Clinical data in support of this view have been obtained with
mifepristone, a glucocorticoid receptor antagonist, which seems to be
3.1. Stress and dysfunction of the hypothalamic–pituitary–adrenal axis efficacious in the treatment of psychotic depression, a subtype of
depression characterized by hypercortisolemia (Belanoff et al., 2001b;
Epidemiological evidence supports a role for stress as a risk factor Schatzberg and Lindley, 2008; Blasey et al., 2009). Mifepristone at
both for depression (Pittenger and Duman, 2008) and Alzheimer's doses ranging from 600 to 1200 mg/day causes, in patients with
disease (Bao et al., 2008; Simard et al., 2009). Several studies clearly psychotic depression, a rapid improvement in depressive and
show that chronic exposure to stress and stressful life events may lead psychotic symptoms (Belanoff et al., 2001a; Belanoff et al., 2002).
to the development of major depression (Czéh and Lucassen, 2007; Preliminary evidence suggests a potential role of mifepristone also in
Kendler et al., 1999; van Praag, 2004; Charney and Manji, 2004; Alzheimer's disease (Pomara et al., 2002; DeBattista and Belanoff,
Pariante, 2003), and that elderly individuals prone to psychological 2005; Dhikav and Anand, 2007). Mifepristone at the dose of 200 mg/
distress are more likely to develop dementia than age-matched non- day showed a positive effect compared to placebo on global cognitive
stressed individuals (Wilson et al., 2006; Simard et al., 2009). function after 6 weeks, as assessed by Alzheimer's Disease Assessment
Stress can be considered as a physiological response to a threat, Scale (DeBattista and Belanoff, 2005). Unfortunately no studies have
which usually activates a specific neuroendocrine pathway known as been conducted yet in Alzheimer's disease patients to assess the
the hypothalamic–pituitary–adrenal (HPA) axis (de Kloet et al., 2005). clinical efficacy and the safety of mifepristone after a long-term
Stressful stimuli activate the hypothalamus to release the peptide treatment (e.g. 6 month), alone or in combination with cholinesterase
corticotropin-releasing factor (CRF), which then promotes the release inhibitors. Large and long-term clinical studies will be necessary to
in the pituitary gland of the adrenocorticotropic hormone (ACTH) into further validate this attractive putative drug target both in depression
the circulation. Finally ACTH stimulates the release of glucocorticoids and Alzheimer's disease.
from the adrenal cortex. Glucocorticoids are steroid hormones that
readily cross the blood–brain barrier and bind to low-affinity 3.2. Chronic inflammation in depression and Alzheimer's disease
glucocorticoid receptors and high-affinity mineralocorticoid recep-
tors, exerting a negative feedback on HPA axis in the hippocampus and Over the last two decades several studies have demonstrated that
regulating normal cellular metabolic activity finally influencing many chronic inflammation has a central role in the pathophysiology of both
CNS functions, such as learning and memory (de Kloet et al., 2005). depression and dementia (Maes et al., 2009; Adler et al., 2006;
Stress and HPA axis abnormalities have been demonstrated both in Leonard, 2007; Rojo et al., 2008). An altered activation of the immune
depression and Alzheimer's disease, with an impaired negative feed- system and the ensuing state of chronic inflammation seem to be
back of glucocorticoids on the activity of the HPA axis, which results in strictly correlated to the HPA dysfunction observed in depressed
elevated cortisol levels (Krishnan and Nestler, 2008; de Kloet et al., patients (Maes, 1999). It has been hypothesized that both high cortisol
2005; Davis et al., 1986; Masugi et al., 1989; Swanwick et al., 1998). levels and the activation of immune system can be explained with the
Interestingly, the dysfunction of HPA axis occurs early in Alzheimer's development of glucocorticoid receptor resistance found in depressed
F. Caraci et al. / European Journal of Pharmacology 626 (2010) 64–71 67

patients (Maes et al., 2009). Depressed patients have higher levels of developing Alzheimer's disease and delayed the onset of Alzheimer's
pro-inflammatory cytokines, such as interleukin-1 (IL-1), interleukin- disease (Pasinetti, 2002; Imbimbo, 2009). The inducible cyclooxy-
2 (IL-2), interleukin-6 (IL-6), interleukin-8 (IL-8), interleukin-12 (IL- genase-2 (COX-2) promotes amyloidogenic processing (Xiang et al.,
12), interferon-γ (IFNγ) and tumor necrosis factor-α (TNFα) 2002) and COX-2 has been found to be elevated in Alzheimer's disease
(Schiepers et al., 2005), as well as increased acute phase proteins, brain (Pasinetti, 2002). Interestingly in vitro and in vivo studies have
chemokines and cellular adhesion molecules (Maes, 1999). On the also shown that some NSAIDs, such as ibuprofen and indomethacin
other hand, reduced levels of anti-inflammatory cytokines, such as and sulindac sulfide, decrease both β-amyloid 1-42 production and
interleukin-4 (IL-4), interleukin-10 (IL-10) and Transforming-Growth- aggregation by direct modulation of γ-secretase activity (Weggen
Factor-β1 (TGF-β1), have been found in the plasma of depressed et al., 2003) independently from COX inhibition (Weggen et al., 2001;
patients (Maes, 1999; Maes et al., 2009; Myint et al., 2005). Different Rojo et al., 2008). Unfortunately long-term, placebo-controlled
studies have demonstrated a positive correlation between the severity clinical trials with both non-selective and COX-2 selective NSAIDs in
of the symptoms of depression and the increase in the inflammatory Alzheimer's disease patients have produced negative results. New
status (Maes, 1999; Owen, 2001; Maes et al., 2009). Pro-inflammatory studies should be conducted in high-risk populations such as
cytokines interfere with many of the pathophysiological mechanisms amnestic mild cognitive impairment patients carrying one or more
that characterize the pathogenesis of depression, altering serotonin epsilon4 allele of the apolipoprotein E, with or without depression, to
metabolism, and reducing both synaptic plasticity and hippocampal assess the potential neuroprotective efficacy of these drugs. Alterna-
neurogenesis (Maes et al., 2009). Furthermore, pro-inflammatory tively NSAIDs might be used in Alzheimer's disease drug discovery as
cytokines might also promote neurodegenerative processes both in precursors for the development of new β-amyloid-lowering agents .
depression and Alzheimer's disease through the induction of
tryptophan 2,3 dioxygenase (TDO) and indoleamine 2,3 dioxygenase 3.3. Impairment of neurotrophin signalling in depression and Alzheimer's
(IDO) (Leonard, 2007). Activation of these two enzymes leads to an disease: the role of BDNF and TGF-β1
increase in the synthesis of the neurotoxins 3-hydroxykynurenine (3-
OHK) and quinolinic acid (QA), which can induce neuronal apoptosis Neurotrophins are essential for the maintenance of neuronal
activating the NMDA receptor (Myint and Kim, 2003; Leonard, 2007). homeostasis and modulation of synaptic plasticity (Fumagalli et al.,
Chronic inflammation play a central role also in the cascade 2008). Changes in the levels and activities of neurotrophic factors,
of molecular events underlying Alzheimer's disease pathogenesis (Rojo such as brain-derived neurotrophic factor (BDNF), and TGF-β1 have
et al., 2008). Increased cerebrospinal fluid (CSF) levels of interleukin-1β been described both in depression and Alzheimer's disease (Karege
(IL-1β) (Cacabelos et al., 1991) and augmented plasma levels of IL-6 et al., 2005; Murer et al., 2001; Cotman, 2005; Myint et al., 2005; Lee
(Huberman et al., 1995; Singh and Guthikonda, 1997) have been found and Kim, 2006; Tesseur et al., 2006; Mocali et al., 2004).
in Alzheimer's disease patients. In vitro and in vivo studies have Significant decreases in BDNF have been detected in stress-induced
demonstrated that ß-amyloid can activate the microglia to release pro- animal models of depression (reviewed by Krishnan and Nestler,
inflammatory cytokines such as IL-1β, IL-6 and TNF-α (Maccioni et al., 2008) as well as in depressed patients (Angelucci et al., 2005; Karege
2009). Inflammatory responses elicited by elevated Aβ peptides play an et al., 2005). Stress reduces BDNF-mediated signalling in the hippo-
important role in the progression of Alzheimer's disease, and microglia campus, whereas chronic treatment with antidepressants increases
activation is an early event in Alzheimer's disease pathogenesis and BDNF-mediated signaling (Duman and Monteggia, 2006). However,
can be already detected in patients with mild cognitive impairment recent studies suggest that chronic stress can also increase the amount
(Okello et al., 2009). A direct correlation has been established between of BDNF in the nucleus accumbens (Berton et al., 2006), suggesting
ß-amyloid-induced neurodegeneration and cytokine production (Rojo the existence of region-specific alterations of BDNF signaling after
et al., 2008). Pro-inflammatory cytokines, such as IL-1β and IL-6, chronic stress. BDNF is also involved in the pathogenesis of treatment-
contribute to reach the signal to trigger neuronal death in Alzheimer's resistant depression, because an association has been found between
disease brain (Rojo et al., 2008). Pro-inflammatory cytokines can also a genotype of BDNF that significantly impairs the intracellular traf-
activate the cyclin-dependent kinase 5 (CDK5), one of the main kinases ficking and activity-dependent release of BDNF (Egan et al., 2003),
involved in tau hyperphosphorylation, thus finally promoting neurofi- and an increased risk to develop treatment-resistant depression
brillary tangles formation (Rojo et al., 2008; Quintanilla et al., 2004). (Anttila et al., 2007).
These data demonstrate that chronic inflammation is a common An impairment of BDNF signalling has been demonstrated also in
pathophysiological feature both in depression and Alzheimer's Alzheimer's disease (Murer et al., 2001; Cotman, 2005). The levels of
disease and might explain why a history of depression, with the BDNF and its receptor, tropomyosin receptor kinase B (TrkB), are
following chronic inflammatory changes, can promote the develop- reduced in Alzheimer's disease brain, and a deficiency of BDNF
ment of Alzheimer's disease (see Fig. 1). Furthermore, this new signalling has been related to synaptic dysfunction, neurodegenera-
evidence suggests that anti-inflammatory drugs might be considered tion and finally cognitive deficits (Murer et al., 2001; Cotman, 2005).
as a new tool for treatment of depression and Alzheimer's disease. Pro-inflammatory cytokines, such as IL-1β, render neurons vulnerable
Antidepressant drugs exert immune-regulatory effects, reducing the to degeneration by interfering with BDNF-induced neuroprotection
production of pro-inflammatory cytokines and stimulating the (Tong et al., 2008). In addition, β-amyloid oligomers reduce BDNF
synthesis of anti-inflammatory cytokines such as IL-10 and TGF-ß1 signaling by impairing the axonal transport of BDNF in neurons of
(Maes et al., 2009). Unfortunately these drugs produce remission only Alzheimer's disease transgenic mice (Tg2576) (Poon et al., in press).
in 30% of patients. Existing anti-inflammatory drugs such as non- Interestingly, genetic variations of BDNF play a central role in
steroidal anti-inflammatory drugs (NSAIDs) could be selected in trials Alzheimer's disease-related depression (Borroni et al., 2009). For
aimed to augment the clinical efficacy of antidepressants. Interest- example, the presence of the functional single-nucleotide polymor-
ingly it has been demonstrated that acetylsalicylic acid (ASA) phism (G196A; Val 66 → Met 66), which impairs BDNF signaling,
accelerates the antidepressant effect of fluoxetine in animal model significantly increases the risk to develop depression in Alzheimer's
of depression (Brunello et al., 2006), and also shortens the onset of disease patients (Borroni et al., 2009).
action of selective reuptake inhibitors (SSRI) in major depressed non- BDNF can also interact in the CNS with other neurotrophins such as
responder patients (Mendlewicz et al., 2006). TGF-β1. TGF-β1 is an anti-inflammatory cytokine which regulates the
The potential efficacy of anti-inflammatory drugs has also been balance between T helper-1 and T helper-2 cytokines, but it can also
studied in Alzheimer's disease models, starting from the epidemio- act as a neurotrophic factor in the CNS protecting neurons against a
logical evidence that long-term use of NSAIDs reduces the risk of diverse number of insults, including excitotoxicity, hypoxia, ischemia,
68 F. Caraci et al. / European Journal of Pharmacology 626 (2010) 64–71

and most importantly β-amyloid (Vivien and Ali, 2006; Caraci et al., Table 1
2008a). TGF-β1 can also increase synaptic plasticity by enhancing the Potential pharmacological approaches to rescue neurotrophin signaling in depression
and AD.
expression of BDNF and TrkB (Sometani et al., 2001).
Plasma TGF-ß1 levels are reduced in major depressed patients and Drug Mechanism Authors
show a significant negative correlation with the Hamilton Depression Antidepressants Increased TGF-ß1 and BDNF Groves (2007)
Rating Scale (Myint et al., 2005; Lee and Kim, 2006; Sutcigil et al., 2007). plasma levels in vivo and Sutcigil et al., 2007;
Interestingly, TGF-ß1 levels significantly increase after antidepressant augmented neurogenesis Myint et al., 2005;
Vollmar et al., 2008
treatment (Myint et al., 2005), and SSRI drugs such as sertraline might
Aspirin Increased TGF-ß1 Redondo et al. (2007)
exert immunomodulatory effects in vivo through a decrease in the pro- plasma levels in vivo
inflammatory cytokine IL-12 and an increase in the anti-inflammatory Glatiramer Induction of TGF-ß1 Aharoni et al., 2005;
cytokines such as IL-4 and TGF-β1 (Sutcigil et al., 2007). Similarly, and BDNF synthesis Tsai, 2007
therapeutic concentrations of venlafaxine prevent microglial activation, Lithium Increased synthesis Leyhe et al., 2009;
of BDNF and TGF-ß1 Caraci et al., 2008b
reduce pro-inflammatory cytokine secretion, and finally increases the
mGlu2/3 receptor agonists Increased synthesis Bruno et al. (1998)
release of TGF-ß1 in an astroglia–microglia co-culture model (Vollmar and secretion of
et al., 2008). TGF-ß1 from glial cells
Recently an impairment of TGF-β1 signaling has been demon- Valproate Increased synthesis of BDNF Walz et al. (2007)
strated in Alzheimer's disease brain. Tesseur et al. (2006) have found
that the expression of TGF-β type II receptor in neurons is reduced
very early in the course of Alzheimer's disease, and this alteration is heterozygous for a BDNF null mutation are resistant to the effects of
specific for Alzheimer's disease. An impairment of TGF-β1 signaling antidepressants in the forced swim test (Saarelainen et al., 2003).
axis might exert a pathogenetic role in Alzheimer's disease, depriving Activation of the transcription regulator cAMP response element
cortical neurons of trophic support, increasing β-amyloid accumula- binding protein (CREB) is essential for the induction of BDNF by
tion and finally promoting β-amyloid-induced neurodegeneration, as antidepressants (Krishnan and Nestler, 2008; Malberg and Blendy,
observed in different in vitro and in vivo models of Alzheimer's 2005). Antidepressant drugs, as above discussed, induce in vitro ed in
disease (Wyss-Coray, 2006; Caraci et al. 2008a). A loss of function vivo the synthesis of TGF-ß1. A recent study by Kessing et al. (2009)
within the TGF-ß1 signaling pathway may also contribute to tau shows that continued long-term antidepressants treatment is asso-
pathology and neurofibrillary tangle formation in Alzheimer's disease ciated with a reduction in the rate of Alzheimer's disease, suggesting
brain (Chalmers and Love, 2007). Finally, TGF-β1 has a constitutive that it might be worth to assess whether TGF-β1 signaling is a
role in the suppression of inflammation, and several studies have common target for both depression and Alzheimer's disease, and
demonstrated that TGF-β1 reduces microglia activation and promotes whether antidepressant drugs exert their neuroprotective effects
the degradation of β-amyloid by microglial cells (Wyss-Coray, 2006). against Alzheimer's disease via the rescue of TGF-β1 signaling.
Therefore a failure of TGF-β1 signaling in Alzheimer's disease brain Other psychotropic drugs such as lithium and valproate increase
may contribute to enhance both chronic inflammation and neuronal BDNF contents in rat hippocampus and frontal cortex (Walz et al.,
vulnerability to ß-amyloid, thus accelerating the progression of 2007). Lithium increases BDNF serum levels in Alzheimer's disease
Alzheimer's disease (Fig. 1). patients, and additionally determined a significant decrease of
Accordingly a reduction of both the active (25 kDa) and inactive Alzheimer's disease AS-Cog sum scores in comparison to placebo-
(50 kDa) forms of TGF-β1 has been reported in plasma of Alzheimer's treated patients (Leyhe et al., 2009). Prevalence of Alzheimer's disease
disease patients (Mocali et al., 2004). A single nucleotide polymorph- is lower in patients treated with lithium (Yeh and Tsai, 2008), and
isms (SNPs) at codon + 10 (T(C) and +25 (G/C), that reduces the lithium reduces the risk of developing Alzheimer's disease in elderly
levels of expression of TGF-β1, has also been associated with an patients with bipolar disorder (Nunes et al., 2007). Different molecular
increased conversion of mild cognitive impairment in Alzheimer's mechanisms have been suggested to explain the neuroprotective
disease (Arosio et al., 2007). effects of lithium. Recently, we have found that lithium can strongly
Overall these data demonstrate that a deficit of neurotrophins such induce the release of TGF-ß1 from glial cells (Caraci et al., 2008b).
as BDNF and TGF-β1 might be considered a common pathophysio- Among anti-inflammatory drugs aspirin can promote the synthesis
logical event both in depression and Alzheimer's disease, suggesting and release TGF-ß1 in vivo (Redondo et al., 2007) and, as above
that the rescue of neurotrophin signaling pathways could be a new discussed, it increases the response to antidepressant drugs (Brunello
strategy for the treatment of these two diseases. et al., 2006). Interestingly, aspirin is known to reduce the risk of
Neurotrophic factor therapy represents a difficult challenge for developing Alzheimer's disease (Broe et al., 2000). Further studies will
CNS drug discovery, because protein growth factors do not cross the be necessary to assess whether this drug can exert its clinical and
blood–brain barrier and require intracerebral administration to be neuroprotective efficacy by increasing TGF-ß1 signaling.
effective. Central administration of BDNF into the ventricles or the Glatiramer, a synthetic amino acid copolymer currently approved
hippocampus induce neurogenesis and exerts antidepressant-like for the treatment of multiple sclerosis (MS), induces in mice specific
effects in different models of depression (Malberg and Blendy, 2005). suppressor cells of the T helper-2 type that migrate to the brain where
Recently, BDNF gene delivery has also been found to exert substantial they express anti-inflammatory cytokines such as IL-10 and TGF-ß1 in
protective effects in Alzheimer's disease models and has been addition to BDNF (Aharoni et al., 2005). Several studies have shown
proposed as a new potential therapy for Alzheimer's disease that peripheral administration of glatiramer increases neurogenesis in
(Nagahara et al., 2009). Nevertheless central delivery of neurotrophic rodent brain (Tsai, 2007). Glatiramer might exert antidepressant
factors cannot be considered a suitable approach for general medical effects by increasing central BDNF, stimulating neurogenesis or
practice. Alternatively new efforts might be directed to identify alternatively through its anti-inflammatory action. Unfortunately no
centrally available drugs able to increase the local production of BDNF studies have been yet conducted in depressed or Alzheimer's disease
and TGF-ß1. patients to assess the potential neuroprotective effects of glatiramer.
Different drugs are known to induce the synthesis and the release Finally, the production of TGF-ß1 can be enhanced by agonists of
of BDNF and TGF-ß1 in vitro and in vivo (Table 1). Antidepressants group II metabotropic glutamate receptors both in cultured astrocytes
stimulate BDNF synthesis with a time course in line with the onset of (Bruno et al., 1998) and in the mouse brain (D'Onofrio et al., 2001).
clinical efficacy (Duman, 2004; Groves, 2007). Induction of BDNF has a Interestingly agonists of group II metabotropic glutamate receptors
central role in mediating the effects of antidepressant drugs. Mice shorten the latency of antidepressants drugs in animal models of
F. Caraci et al. / European Journal of Pharmacology 626 (2010) 64–71 69

depression (Matrisciano et al., 2007). In addition, these drugs protect Borroni, B., Archetti, S., Costanzi, C., Grassi, M., Ferrari, M., Radeghieri, A., Caimi, L.,
Caltagirone, C., Di Luca, M., Padovani, A., ITINAlzheimer's disease Working Group,
cortical neurons against ß-amyloid toxicity (Copani et al., 1995). It is 2009. Role of BDNF Val66Met functional polymorphism in Alzheimer's disease-
significant that highly potent and centrally available group II related depression. Neurobiol. Aging 30, 1406–1412.
metabotropic glutamate receptor agonists, such as LY404039, are Broe, G.A., Grayson, D.A., Creasey, H.M., Waite, L.M., Casey, B.J., Bennett, H.P., Brooks, W.S.,
Halliday, G.M., 2000. Anti-inflammatory drugs protect against Alzheimer disease at
under clinical development for the treatment of schizophrenia (Patil low doses. Arch. Neurol. 57, 1586–1591.
et al., 2007). We suggest that these drugs are potential candidates as Brunello, N., Alboni, S., Capone, G., Benatti, C., Blom, J.M., Tascedda, F., Kriwin, P.,
neuroprotectant agents both in depression and Alzheimer's disease. Mendlewicz, J., 2006. Acetylsalicylic acid accelerates the antidepressant effect of
fluoxetine in the chronic escape deficit model of depression. Int. Clin. Psycho-
pharmacol. 21, 219–225.
4. Conclusion Bruno, V., Battaglia, G., Casabona, G., Copani, A., Caciagli, F., Nicoletti, F., 1998. Neuro-
protection by glial metabotropic glutamate receptors is mediated by transforming
growth factor-beta. J. Neurosci. 18, 9594–9600.
In the present review we have examined recent evidence on the
Butterfield, D.A., Reed, T., Newman, S.F., Sultana, R., 2007. Roles of amyloid beta-peptide-
common molecular mechanisms and cascades between depression associated oxidative stress and brain protein modifications in the pathogenesis of
and Alzheimer's disease. The above short review did not intend to Alzheimer's disease and mild cognitive impairment. Free Radic. Biol. Med. 43, 658–677.
cover the vast amount of data in this field. Cacabelos, R., Barquero, M., García, P., Alvarez, X.A., Varela de Seijas, E., 1991.
Cerebrospinal fluid interleukin-1 beta (IL-1 beta) in Alzheimer's disease and
According to the discussed data we suggest that hyperactivation of neurological disorders. Methods Find. Exp. Clin. Pharmacol. 13, 455–458.
HPA axis, chronic inflammation and deficit of neurotrophin signaling Caraci, F., Battaglia, G., Busceti, C., Biagioni, F., Mastroiacovo, F., Bosco, P., Drago, F., Nicoletti,
can be considered as common pathophysiological mechanisms between F., Sortino, M.A., Copani, A., 2008a. TGF-beta 1 protects against Abeta-neurotoxicity via
the phosphatidylinositol-3-kinase pathway. Neurobiol. Dis. 30, 234–242.
depression and Alzheimer's disease (Fig. 1) and are potential new Caraci, F., Battaglia, G., Busceti, C., Biagioni, F., Mastroiacovo, F., Bosco, P., Leggio, G.M.,
targets for pharmacological treatment of these two disorders. These Drago, F., Nicoletti, F., Sortino, M.A., Copani, A., 2008b. TGF-ß1 signaling as a new
common molecular pathways might be studied in the future for the pharmacological target against Aß induced neurodegeneration. 21st ECNP Congress.
Eur. Neuropsychopharmacol. 18 (S4), 168.
discovery of new biological markers which predict the progression from Caricasole, A., Copani, A., Caraci, F., Aronica, E., Rozemuller, A.J., Caruso, A., Storto, M.,
depression to Alzheimer's disease, and for the identification of early Gaviraghi, G., Terstappen, G.C., Nicoletti, F., 2004. Induction of Dickkopf-1, a negative
Alzheimer's disease patients eligible for disease-modifying therapies. modulator of the Wnt pathway, is associated with neuronal degeneration in
Alzheimer's brain. J. Neurosci. 24, 6021–6027.
Carlson, E., Sherwin, B.B., Chertkow, H.M., 1999. Relationships between dehydroepian-
References drosterone sulfate (DHEAS) and cortisol (CRT) plasma levels and everyday memory in
Alzheimer's disease patients compared to healthy controls. Horm. Behav. 35, 254–263.
Abraham, I., Harkany, E.M., Horvath, K.M., Veenema, A.H., Penke, B., Nyakas, C., Luiten, P.G., Castaneda, A.E., Tuulio-Henriksson, A., Marttunen, M., Suvisaari, J., Lönnqvist, J., 2008. A
2000. Chronic corticosterone administration dose dependently modulates Abeta review on cognitive impairments in depressive and anxiety disorders with a focus
(1–42) and NMDA-induced neurodegeneration in rat magnocellular nucleus on young adults. J. Affect. Disord. 106, 1–27.
basalis. J. Neuroendocrinol. 12, 486–494. Cerpa, W., Dinamarca, M.C., Inestrosa, N.C., 2008. Structure-function implications in
Adler, U.C., Marques, A.H., Calil, H.M., 2006. Inflammatory aspects of depression. Alzheimer's disease: effect of Abeta oligomers at central synapses. Curr. Alzheimer
Inflamm. Allergy Drug Targets 7, 19–23. Res. 5, 233–243.
Aharoni, R., Eilam, R., Domev, H., Labunskay, G., Sela, M., Arnon, R., 2005. The immuno- Chalmers, K.A., Love, S., 2007. Neurofibrillary tangles may interfere with Smad 2/3
modulator glatiramer acetate augments the expression of neurotrophic factors in signaling in neurons. J. Neuropathol. Exp. Neurol. 66, 158–167.
brains of experimental autoimmune encephalomyelitis mice. Proc. Natl. Acad. Sci. Charles, G.A., Lefevre, A., Mirel, J., Rush, A.J., 1986. Suppression of cortisol following
U. S. A. 102, 19045–19050. dexamethasone in demented patients. Psychol. Res. 17, 173–182.
Aisen, P.S., Davis, K.L., Berg, J.D., Schafer, K., Campbell, K., Thomas, R.G., Weiner, M.F., Charney, D.S., Manji, H.K., 2004. Life stress, genes, and depression: multiple pathways lead
Farlow, M.R., Sano, M., Grundman, M., Thal, L.J., 2000. A randomized controlled trial to increased risk and new opportunities for intervention. Science's STKE 2004, re5.
of prednisone in Alzheimer's disease. Alzheimer's Disease Cooperative Study. Copani, A., Bruno, V., Battaglia, G., Lenza, G., Pellitteri, R., Russo, A., Stanzani, S., Nicoletti, F.,
Neurology 54, 588–593. 1995. Activation of metabotropic glutamate receptors protects cultured neurons
Angelucci, F., Brene, S., Mathe, A.A., 2005. BDNF in schizophrenia, depression and against apoptosis induced by beta-amyloid peptide. Mol. Pharmacol. 47, 890–897.
corresponding animal models. Mol. Psychiatry 10, 345–352. Copani, A., Condorelli, F., Caruso, A., Vancheri, C., Sala, A., Giuffrida-Stella, A.M.,
Anttila, S., Huuhka, K., Huuhka, M., Rontu, R., Hurme, M., Leinonen, E., Lehtimaki, T., Canonico, P.L., Nicoletti, F., Sortino, M.A., 1999. Mitotic signaling by beta-amyloid
2007. Interaction between 5-HT1A and BDNF genotypes increases the risk of causes neuronal death. FASEB J. 13, 2225–2234.
treatment-resistant depression. J. Neural Transm. 114, 1065–1068. Copani, A., Caraci, F., Hoozemans, J.J., Calafiore, M., Sortino, M.A., Nicoletti, F., 2007. The
Arosio, B., Bergamaschini, L., Galimberti, L., La Porta, C., Zanetti, M., Calabresi, C., Scarpini, E., nature of the cell cycle in neurons: focus on a “non-canonical” pathway of DNA
Annoni, G., Vergani, C., 2007. +10 T/C polymorphisms in the gene of transforming replication causally related to death. Biochim. Biophys. Acta 1772, 409–412.
growth factor-beta1 are associated with neurodegeneration and its clinical evolution. Cotman, C.W., 2005. The role of neurotrophins in brain aging: a perspective in honor of
Mech. Ageing Dev. 128, 553–557. Regino Perez-Polo. Neurochem. Res. 30, 877–881.
Assal, F., Cummings, J.L., 2002. Neuropsychiatric symptoms in the dementias. Curr. Crochemore, C., Lu, J., Wu, Y., Liposits, Z., Sousa, N., Holsboer, F., Almeida, O.F., 2005.
Opin. Neurol. 15, 445–450. Direct targeting of hippocampal neurons for apoptosis by glucocorticoids is
Baldwin, R.C., Gallagley, A., Gourlay, M., Jackson, A., Burns, A., 2006. Prognosis of late life reversible by mineralocorticoid receptor activation. Mol. Psychiatry 10, 790–798.
depression: a three-year cohort study of outcome and potential predictors. Int. J Cuijpers, P., Smit, F., 2002. Excess mortality in depression: a meta-analysis of community
Geriatr. Psychiatry 21, 57–63. studies. J. Affect. Disord. 72, 227–236.
Ballard, C., Day, S., Sharp, S., Wing, G., Sorensen, S., 2008. Neuropsychiatric symptoms in Czéh, B., Lucassen, P.J., 2007. What causes the hippocampal volume decrease in
dementia: importance and treatment considerations. Int. Rev. Psychiatry 20, 396–404. depression? Are neurogenesis, glial changes and apoptosis implicated? Eur. Arch.
Ballmaier, M., Narr, K.L., Toga, A.W., Elderkin-Thompson, V., Thompson, P.M., Hamilton, L., Psychiatry Clin. Neurosci. 257, 250–260.
Haroon, E., Pham, D., Heinz, A., Kumar, A., 2008. Hippocampal morphology and distin- Davis, K.L., Davis, B.M., Greenwald, B.S., Mohs, R.C., Mathe, A.A., Johns, C.A., Horvath, T.B.,
guishing late-onset from early-onset elderly depression. Am. J. Psychiatry 165, 229–237. 1986. Cortisol and Alzheimer's disease. I. Basal studies. Am. J. Psychiatry 143, 300–305.
Bao, A.M., Meynen, G., Swaab, D.F., 2008. The stress system in depression and Davous, P., 1987. Cortisol and Alzheimer's disease. Am. J. Psychiatry 144, 533–535.
neurodegeneration: focus on the human hypothalamus. Brain Res. Rev. 57, 531–553. de Kloet, E.R., Joels, M., Holsboer, F., 2005. Stress and the brain: from adaptation to
Behl, C., Lezoualc'h, F., Trapp, T., Widmann, M., Skutella, T., Holsboer, F., 1997. disease. Nat. Rev. 6, 463–475.
Glucocorticoids enhance oxidative stress-induced cell death in hippocampal DeBattista, C., Belanoff, J., 2005. C-1073 (mifepristone) in the adjunctive treatment of
neurons in vitro. Endocrinology 138, 101–106. Alzheimer's disease. Curr. Alzheimer Res. 2, 125–129.
Belanoff, J.K., Flores, B.H., Kalezhan, M., Sund, B., Schatzberg, A.F., 2001a. Rapid reversal Dhikav, V., Anand, K.S., 2007. Glucocorticoids may initiate Alzheimer's disease: a potential
of psychotic depression using mifepristone. J. Clin. Psychopharmacol. 21, 516–521. therapeutic role for mifepristone (RU-486). Med. Hypotheses 68, 1088–1092.
Belanoff, J.K., Gross, K., Yager, A., Schatzberg, A.F., 2001b. Corticosteroids and cognition. D'Onofrio, M., Cuomo, L., Battaglia, G., Ngomba, R.T., Storto, M., Kingston, A.E., Orzi, F., De
J. Psychiatr. Res. 35, 127–145. Blasi, A., Di Iorio, P., Nicoletti, F., Bruno, V., 2001. Neuroprotection mediated by glial
Belanoff, J.K., Rothschild, A.J., Cassidy, F., DeBattista, C., Baulieu, E.E., Schold, C., group-II metabotropic glutamate receptors requires the activation of the MAP kinase
Schatzberg, A.F., 2002. An open label trial of C-1073 (mifepristone) for psychotic and the phosphatidylinositol-3-kinase pathways. J. Neurochem. 78, 435–445.
major depression. Biol. Psychiatry 52, 386–392. Duman, R.S., 2004. Depression: a case of neuronal life and death? Biol. Psychiatry 56,
Berton, O., McClung, C.A., Dileone, R.J., Krishnan, V., Renthal, W., Russo, S.J., Graham, D., 140–145.
Tsankova, N.M., Bolanos, C.A., Rios, M., Monteggia, L.M., Self, D.W., Nestler, E.J., Duman, R.S., Monteggia, L.M., 2006. A neurotrophic model for stress-related mood
2006. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat disorders. Biol. Psychiatry 59, 1116–1127.
stress. Science 311, 864–868. Egan, M.F., Kojima, M., Callicott, J.H., Goldberg, T.E., Kolachana, B.S., Bertolino, A.,
Blasey, C.M., Debattista, C., Roe, R., Block, T., Belanoff, J.K., 2009. A multisite trial of Zaitsev, E., Gold, B., Goldman, D., Dean, M., Lu, B., Weinberger, D.R., 2003. The BDNF
mifepristone for the treatment of psychotic depression: a site-by-treatment inter- Val66Met polymorphism affects activity-dependent secretion of BDNF and human
action. Contemp. Clin. Trials 30, 284–288. memory and hippocampal function. Cell 112, 257–269.
70 F. Caraci et al. / European Journal of Pharmacology 626 (2010) 64–71

Elderkin-Thompson, V., Kumar, A., Bilker, W.B., Dunkin, J.J., Mintz, J., Moberg, P.J., Maes, M., 1999. Major depression and activation of the inflammatory response. Adv.
Mesholam, R.I., Gur, R.E., 2003. Neuropsychological deficits among patients with Exp. Biol. Med. 461, 25–46.
late-onset minor and major depression. Arch. Clin. Neuropsychol. 18, 529–549. Maes, M., Yirmyia, R., Noraberg, J., Brene, S., Hibbeln, J., Perini, G., Kubera, M., Bob, P.,
Elliott, E.M., Sapolsky, R.M., 1992. Corticosterone enhances kainic acid-induced calcium Lerer, B., Maj, M., 2009. The inflammatory & neurodegenerative (I&ND) hypothesis
elevation in cultured hippocampal neurons. J. Neurochem. 59, 1033–1040. of depression: leads for future research and new drug developments in depression.
Fava, M., Kendler, K.S., 2000. Major depressive disorder. Neuron 28, 335–341. Metab. Brain Dis. 24, 27–53.
Fumagalli, F., Molteni, R., Calabrese, F., Maj, P.F., Racagni, G., Riva, M.A., 2008. Neuro- Magariños, A.M., McEwen, B.S., 1995. Stress-induced atrophy of apical dendrites of
trophic factors in neurodegenerative disorders: potential for therapy. CNS Drugs hippocampal CA3c neurons: comparison of stressors. Neuroscience 69, 83–88.
22, 1005–1019. Malberg, J.E., Blendy, J.A., 2005. Antidepressant action: to the nucleus and beyond.
Gallassi, R., Di, S.R., Morreale, A., Amore, M., 2006. Memory impairment in patients with Trends Pharmacol. Sci. 26, 631–638.
late-onset major depression: the effect of antidepressant therapy. J. Affect. Disord. Masugi, F., Ogihara, T., Sakaguchi, K., Otsuka, A., Tsuchiya, Y., Morimoto, S., Kumahara,
91, 243–250. Y., Saeki, S., Nishide, M., 1989. High plasma levels of cortisol in patients with
Geerlings, M.I., Schoevers, R.A., Beekman, A.T., Jonker, C., Deeg, D.J., Schmand, B., Adèr, H.J., senile dementia of the Alzheimer's type. Methods Find. Exp. Clin. Pharmacol. 11,
Bouter, L.M., Van Tilburg, W., 2000. Depression and risk of cognitive decline and 707–710.
Alzheimer's disease. results of two prospective community-based studies in The Matrisciano, F., Panaccione, I., Zusso, M., Giusti, P., Tatarelli, R., Iacovelli, L., Mathé, A.A.,
Netherlands. Br. J. Psychiatry 176, 568–575. Gruber, S.H., Nicoletti, F., Girardi, P., 2007. Group-II metabotropic glutamate
Giovanni, A., Wirtz-Brugger, F., Keramaris, E., Slack, R., Park, D.S., 1999. Involvement of cell receptor ligands as adjunctive drugs in the treatment of depression: a new strategy
cycle elements, cyclin-dependent kinases, pRb, and E2F-DP, in -amyloid-induced to shorten the latency of antidepressant medication? Mol. Psychiatry 12, 704–706.
neuronal death. J. Biol. Chem. 274, 19011–19016. McLaughlin, K.J., Gomez, J.L., Baran, S.E., Conrad, C.D., 2007. The effects of chronic stress on
Goodman, Y., Bruce, A.J., Cheng, B., Mattson, M.P., 1996. Estrogens attenuate and hippocampal morphology and function: an evaluation of chronic restraint paradigms.
corticosterone exacerbates excitotoxicity, oxidative injury, and amyloid beta-peptide Brain Res. 1161, 56–64.
toxicity in hippocampal neurons. J. Neurochem. 66 (5), 1836–1844 May. Mendlewicz, J., Kriwin, P., Oswald, P., Souery, D., Alboni, S., Brunello, N., 2006. Shortened
Green, K.N., Billings, L.M., Roozendaal, B., McGaugh, J.L., LaFerla, F.M., 2006. Glucocorticoids onset of action of antidepressants in major depression using acetylsalicylic acid
increase amyloid-beta and tau pathology in a mouse model of Alzheimer's disease. augmentation: a pilot open-label study. Int. Clin. Psychopharmacol. 21, 227–231.
J. Neurosci. 26, 9047–9056. Milwain, E.J., Nagy, Z., 2005. Depressive symptoms increase the likelihood of cognitive
Greenwald, B.S., Mathe, A.A., Mohs, R.C., Levy, M.I., Johns, C.A., Davis, K.L., 1986. Cortisol impairment in elderly people with subclinical Alzheimer pathology. Dement. Geriatr.
and Alzheimer's disease, II: Dexamethasone suppression, dementia severity, and Cogn. Disord. 19, 46–50.
affective symptoms. Am. J. Psychol. 143, 442–446. Mocali, A., Cedrola, S., Della Malva, N., Bontempelli, M., Mitidieri, V.A., Bavazzano, A.,
Groves, J.O., 2007. Is it time to reassess the BDNF hypothesis of depression? Mol. Psychiatry Comolli, R., Paoletti, F., La Porta, C.A., 2004. Increased plasma levels of soluble CD40,
12, 1079–1088. together with the decrease of TGF beta 1, as possible differential markers of
Gualtieri, C.T., Johnson, L.G., 2008. Age-related cognitive decline in patients with mood Alzheimer disease. Exp. Gerontol. 39, 1555–1561.
disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 32, 962–967. Modrego, P.J., Ferrández, J., 2004. Depression in patients with mild cognitive impairment
Gurevich, D., Siegel, B., Dumlao, M., Perl, E., Chaitin, P., Bagne, C., Oxenkrug, G., 1990. increases the risk of developing dementia of Alzheimer type: a prospective cohort
HPA axis responsivity to dexamethasone and cognitive impairment in dementia. study. Arch. Neurol. 61, 1290–1293.
Prog. Neuropsychopharmacol. Biol. Psychiatry 14, 297–308. Murer, M.G., Yan, Q., Raisman-Vozari, R., 2001. Brain-derived neurotrophic factor in the
Hardy, J., Selkoe, D.J., 2002. The amyloid hypothesis of Alzheimer's disease: progress control human brain, and in Alzheimer's disease and Parkinson's disease. Prog.
and problems on the road to therapeutics. Science 297, 353–356. Neurobiol. 63, 71–124.
Herrup, K., Neve, R., Ackerman, S.L., Copani, A., 2004. Divide and die: cell cycle events as Murray, C.J., Lopez, A.D., 1997. Global mortality, disability, and the contribution of risk
triggers of nerve cell death. J. Neurosci. 24, 9232–9239. factors: Global Burden of Disease Study. Lancet 349, 1436–1442.
Houde, M., Bergman, H., Whitehead, V., Chertkow, H., 2008. A predictive depression Myint, A.M., Kim, Y.K., 2003. Cytokine-serotonin interactions through indoleamine 2, 3
pattern in mild cognitive impairment. Int. J. Geriatr. Psychopharmacol. 23, 1028–1033. dioxygenase: a neurodegenerative hypothesis of depression. Med. Hypotheses 61,
Huberman, M., Sredni, B., Stern, L., Kott, E., Shalit, F., 1995. IL-2 and IL-6 secretion in 519–525.
dementia: correlation with type and severity of disease. J. Neurol. Sci. 130, 161–164. Myint, A.M., Leonard, B.E., Steinbusch, H.W., Kim, Y.K., 2005. Th1, Th2, and Th3 cytokine
Hynd, M.R., Scott, H.L., Dodd, P.R., 2004. Glutamate-mediated excitotoxicity and alterations in major depression. J. Affect. Disord. 88, 167–173.
neurodegeneration in Alzheimer's disease. Neurochem. Int. 45, 583–595. Nagahara, A.H., Merrill, D.A., Coppola, G., Tsukada, S., Schroeder, B.E., Shaked, G.M.,
Imbimbo, B.P., 2009. An update on the efficacy of non-steroidal anti-inflammatory Wang, L., Blesch, A., Kim, A., Conner, J.M., Rockenstein, E., Chao, M.V., Koo, E.H.,
drugs in Alzheimer's disease. Exp. Opin. Investig. Drugs 18, 1147–1168. Geschwind, D., Masliah, E., Chiba, A.A., Tuszynski, M.H., 2009. Neuroprotective
Inestrosa, N.C., Varela-Nallar, L., Grabowski, C.P., Colombres, M., 2007. Synaptotoxicity effects of brain-derived neurotrophic factor in rodent and primate models of
in Alzheimer's disease: the Wnt signaling pathway as a molecular target. IUBMB Alzheimer's disease. Nat. Med. 15, 331–337.
Life 59, 316–321. Nunes, P.V., Forlenza, O.V., Gattaz, W.F., 2007. Lithium and risk for Alzheimer's disease
Karege, F., Vaudan, G., Schwald, M., Perroud, N., La Harpe, R., 2005. Neurotrophin levels in elderly patients with bipolar disorder. Br. J. Psychiatry 190, 359–360.
in postmortem brains of suicide victims and the effects of antemortem diagnosis Okello, A., Edison, P., Archer, H.A., Turkheimer, F.E., Kennedy, J., Bullock, R., Walker, Z.,
and psychotropic drugs. Brain Res. Mol. Brain Res. 136, 29–37. Kennedy, A., Fox, N., Rossor, M., Brooks, D.J., 2009. Microglial activation and amyloid
Kendler, K.S., Karkowski, L.M., Prescott, C.A., 1999. Causal relationship between stressful deposition in mild cognitive impairment: a PET study. Neurology 72, 56–62.
life events and the onset of major depression. Am. J. Psychiatry 156, 837–841. Owen, B.M., 2001. Raised levels of plasma IL-1 beta in major and post viral depression.
Kessing, L.V., 1998. Cognitive impairment in the euthymic phase of affective disorder. Acta Psychiatr. Scand. 103, 226–228.
Psychol. Med. 28, 1027–1038. Ownby, R.L., Crocco, E., Acevedo, A., John, V., Loewenstein, D., 2006. Depression and risk
Kessing, L.V., Andersen, P.K., 2004. Does the risk of developing dementia increase with for Alzheimer disease: systematic review, meta-analysis, and metaregression
the number of episodes in patients with depressive disorder and in patients with analysis. Arch. Gen. Psychiatry 63, 530–538.
bipolar disorder? J. Neurol. Neurosurg. Psychiatry 75, 1662–1666. Pariante, C.M., 2003. Depression, stress and the adrenal axis. J. Neuroendocrinol. 15,
Kessing, L.V., Søndergård, L., Forman, J.L., Andersen, P.K., 2009. Antidepressants and 811–812.
dementia. J. Affect. Disord. 117, 24–29. Pasinetti, G.M., 2002. From epidemiology to therapeutic trials with anti-inflammatory
Kessler, R.C., Berglund, P., Delmer, O., Jin, R., Merikangas, K.R., Walters, E.E., 2005. drugs in Alzheimer's disease: the role of NSAIDs and cyclooxygenase in beta-
Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the amyloidosis and clinical dementia. J. Alzheimer's Dis. 4, 435–445.
National Comorbidity Survey Replication. Arch. Gen. Psychiatry 62, 590–592. Patil, S.T., Zhang, L., Martenyi, F., Lowe, S.L., Jackson, K.A., Andreev, B.V., Avedisova, A.S.,
Klafki, H.W., Staufenbiel, M., Kornhuber, J., Wiltfang, J., 2006. Therapeutic approaches to Bardenstein, L.M., Gurovich, I.Y., Morozova, M.A., Mosolov, S.N., Neznanov, N.G.,
Alzheimer's disease. Brain 129, 2840–2855. Reznik, A.M., Smulevich, A.B., Tochilov, V.A., Johnson, B.G., Monn, J.A., Schoepp, D.D.,
Köhler, S., Thomas, A.J., Barnett, N.A., O'Brien, J.T., 2009. The pattern and course of 2007. Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: a
cognitive impairment in late-life depression. Psychol. Med. 6, 1–12. randomized Phase 2 clinical trial. Nat. Med. 13, 1102–1107.
Krishnan, V., Nestler, E.J., 2008. The molecular neurobiology of depression. Nature 455, Pittenger, C., Duman, R.S., 2008. Stress, depression, and neuroplasticity: a convergence
894–902. of mechanisms. Neuropsychopharmacology 33, 88–109.
Landfield, P.W., Blalock, E.M., Chen, K.C., Porter, N.M., 2007. A new glucocorticoid Pomara, N., Doraiswamy, D.M., Tun, H., Ferris, H., 2002. Mifepristone (RU486) for
hypothesis of brain aging: implications for Alzheimer's disease. Curr. Alzheimer Alzheimer's disease. Neurology 58, 1436.
Res. 4, 205–212. Poon, W.W., Blurton-Jones, M., Tu, C.H., Feinberg, L.M., Chabrier, M.A., Harris, J.W., Jeon, N.L.,
Lee, K.M., Kim, Y.K., 2006. The role of IL-12 and TGF-beta1 in the pathophysiology of Cotman, C.W., in press. β-Amyloid impairs axonal BDNF retrograde trafficking.
major depressive Disorder. Int. Immunopharmacol. 6, 1298–1304. Neurobiol. Aging. doi:10.1016/j.neurobiolaging.2009.05.012.
Leonard, B.E., 2007. Inflammation, depression and dementia: are they connected? Quintanilla, R.A., Orellana, D.I., González-Billault, C., Maccioni, R.B., 2004. Interleukin-6
Neurochem. Res. 32, 1749–1756. induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35
Leyhe, T., Eschweiler, G.W., Stransky, E., Gasser, T., Annas, P., Basun, H., Laske, C., 2009. pathway. Exp. Cell Res. 295, 245–257.
Increase of BDNF serum concentration in lithium treated patients with early Rapp, M.A., Schnaider-Beeri, M., Purohit, D.P., Perl, D.P., Haroutunian, V., Sano, M., 2008.
Alzheimer's disease. J. Alzheimer's Dis. 16, 649–656. Increased neurofibrillary tangles in patients with Alzheimer disease with comorbid
Lyketsos, C.G., Steele, C., Baker, L., Galik, E., Kopunek, S., Steinberg, M., Warren, A., 1997. depression. Am. J. Geriatr. Psychiatry 16, 168–174.
Major and minor depression in Alzheimer's disease: prevalence and impact. Redondo, S., Santos-Gallego, C.G., Tejerina, T., 2007. TGF-beta1: a novel target for
J. Neuropsychiatry Clin. Neurosci. 9, 556–561. cardiovascular pharmacology. Cytokine Growth Factor Rev. 18, 279–286.
Maccioni, R.B., Rojo, L.E., Fernández, J.A., Kuljis, R.O., 2009. The role of neuroimmuno- Rivelli, S., Jiang, W., 2007. Depression and ischemic heart disease: what have we learned
modulation in Alzheimer's disease. Ann. N. Y. Acad. Sci. 1153, 240–246. from clinical trials? Curr. Opin. Cardiol. 22, 286–291.
F. Caraci et al. / European Journal of Pharmacology 626 (2010) 64–71 71

Robert, P.H., Schuck, S., Dubois, B., Lepine, J.P., Gallarda, T., Olie, J.P., Goni, S., Troy, S., 2003. Tesseur, I., Zou, K., Esposito, L., Bard, F., Berber, E., Can, J.V., Lin, A.H., Crews, L., Tremblay, P.,
Validation of the short cognitive battery (B2C). Value in screening for Alzheimer's Mathews, P., Mucke, L., Masliah, E., Wyss-Coray, T., 2006. Deficiency in neuronal TGF-
disease and depressive disorders in psychiatric practice. Encephale 29, 266–272. beta signaling promotes neurodegeneration and Alzheimer's pathology. J. Clin. Invest.
Rojo, L.E., Fernández, J.A., Maccioni, A.A., Jimenez, J.M., Maccioni, R.B., 2008. Neuro- 116, 3060–3069.
inflammation: implications for the pathogenesis and molecular diagnosis of Thomas, A.J., O'Brien, J.T., 2008. Depression and cognition in older adults. Curr. Opin.
Alzheimer's disease. Arch. Med. Res. 39, 1–16. Psychiatry 21, 8–13.
Saarelainen, T., Hendolin, P., Lucas, G., Koponen, E., Sairanen, M., MacDonald, E., Agerman, Tong, L., Balazs, R., Soiampornkul, R., Thangnipon, W., Cotman, C.W., 2008. Interleukin-1
K., Haapasalo, A., Nawa, H., Aloyz, R., Ernfors, P., Castrén, E., 2003. Activation of the beta impairs brain derived neurotrophic factor-induced signal transduction.
TrkB neurotrophin receptor is induced by antidepressant drugs and is required for Neurobiol. Aging 29, 1380–1393.
antidepressant-induced behavioral effects. J. Neurosci. 23, 349–357. Tsai, S.J., 2007. Glatiramer acetate could be a potential antidepressant through its
Sapolsky, R.M., 1986. Glucocorticoid toxicity in the hippocampus: reversal by neuroprotective and anti-inflammatory effects. Med. Hypotheses 69, 145–148.
supplementation with brain fuels. J. Neurosci. 6, 2240–2244. van Praag, H.M., 2004. Can stress cause depression? Prog. Neuro-Psychopharmacol.
Sapolsky, R.M., 2000. Glucocorticoids and hippocampal atrophy in neuropsychiatric Biol. Psychiatry 28, 891–907.
disorders. Arch. Gen. Psychiatry 57, 925–935. Vivien, D., Ali, C., 2006. Transforming growth factor- signalling in brain disorder.
Schatzberg, A.F., Lindley, S., 2008. Glucocorticoid antagonists in neuropsychotic Cytokine Growth Factor Rev. 17, 121–128.
disorders. Eur. J. Pharmacol. 583, 358–364. Vollmar, P., Haghikia, A., Dermietzel, R., Faustmann, P.M., 2008. Venlafaxine exhibits an
Schiepers, O.J., Wichers, M.C., Maes, M., 2005. Cytokines and major depression. Prog. anti-inflammatory effect in an inflammatory co-culture model. Int. J. Neuropsycho-
Neuropsychopharmacol. Biol. Psychiatry 29, 201–217. pharmacol. 11, 111–117.
Sheline, Y.I., Gado, M.H., Kraemer, H.C., 2003. Untreated depression and hippocampal Walz, J.C., Frey, B.N., Andreazza, A.C., Cereser, K.M., Cacilhas, A.A., Valvassori, S.S.,
volume loss. Am. J. Psychiatry 160, 1516–1518. Quevedo, J., Kapczinski, F., 2007. Effects of lithium and valproate on serum and
Sheline, Y.I., Barch, D.M., Garcia, K., Gersing, K., Pieper, C., Welsh-Bohmer, K., Steffens, D.C., hippocampal neurotrophin-3 levels in an animal model of mania. J. Psychiatr. Res.
Doraiswamy, P.M., 2006. Cognitive function in late life depression: relationships to 42, 416–421.
depression severity, cerebrovascular risk factors and processing speed. Biol. Psychiatry Weggen, S., Eriksen, J.L., Das, P., Sagi, S.A., Wang, R., Pietrzik, C.U., Findlay, K.A., Smith, T.E.,
60, 58–65. Murphy, M.P., Bulter, T., Kang, D.E., Marquez-Sterling, N., Golde, T.E., Koo, E.H., 2001. A
Shim, Y.S., Yang, D.W., 2006. Depression as prognostic factor: 6 months follow-up in a subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase
geriatric institution. Arch. Gerontol. Geriatr. 43, 277–283. activity. Nature 414, 212–216.
Simard, M., Hudon, C., van Reekum, R., 2009. Psychological distress and risk for Weggen, S., Eriksen, J.L., Sagi, S.A., Pietrzik, C.U., Ozols, V., Fauq, A., Golde, T.E., Koo, E.H.,
dementia. Curr. Psychiatry Rep. 11, 41–47. 2003. Evidence that nonsteroidal anti-inflammatory drugs decrease amyloid beta
Singh, V.K., Guthikonda, P., 1997. Circulating cytokines in Alzheimer's disease. J. Psychiatr. 42 production by direct modulation of gamma-secretase activity. J. Biol. Chem. 278,
Res. 31, 657–660. 31831–31837.
Sometani, A., Kataoka, H., Nitta, A., Fukumitsu, H., Nomoto, H., Furukawa, S., 2001. Wilson, R.S., Arnold, S.E., Schneider, J.A., Kelly, J.F., Tang, Y., Bennett, D.A., 2006. Chronic
Transforming growth factor-beta1 enhances expression of brain-derived neuro- psychological distress and risk of Alzheimer's disease in old age. Neuroepidemiology
trophic factor and its receptor, TrkB, in neurons cultured from rat cerebral cortex. 27, 143–153.
J. Neurosci. Res. 66, 369–376. Wyss-Coray, T., 2006. Tgf-Beta pathway as a potential target in neurodegeneration and
Sotiropoulos, I., Cerqueira, J.J., Catania, C., Takashima, A., Sousa, N., Almeida, O.F., 2008. Alzheimer's. Curr. Alzheimer Res. 3, 191–195.
Stress and glucocorticoid footprints in the brain-the path from depression to Xiang, Z., Ho, L., Yemul, S., Zhao, Z., Qing, W., Pompl, P., Kelley, K., Dang, A., Qing, W.,
Alzheimer's disease. Neurosci. Biobehav. Rev. 32, 1161–1173. Teplow, D., Pasinetti, G.M., 2002. Cyclooxygenase-2 promotes amyloid plaque
Starkstein, S.E., Jorge, R., Mizrahi, R., Robinson, R.G., 2005. The construct of minor and deposition in a mouse model of Alzheimer's disease neuropathology. Gene Expr. 10,
major depression in Alzheimer's disease. Am. J. Psychol. 162, 2086–2093. 271–278.
Sutcigil, L., Oktenli, C., Musabak, U., Bozkurt, A., Cansever, A., Uzun, O., Sanisoglu, S.Y., Yeh, H.L., Tsai, S.J., 2008. Lithium may be useful in the prevention of Alzheimer's disease
Yesilova, Z., Ozmenler, N., Ozsahin, A., Sengul, A., 2007. Pro- and anti-inflammatory in individuals at risk of presenile familial Alzheimer's disease. Med. Hypotheses 71,
cytokine balance in major depression: effect of sertraline therapy. Clin. Dev. 948–951.
Immunol. 2007, 76396. Yu, S., Holsboer, F., Almeida, O.F., 2008. Neuronal actions of glucocorticoids: focus on
Swanwick, G.R., Kirby, M., Bruce, I., Buggy, F., Coen, R.F., Coakley, D., Lawlor, B.A., 1998. depression. J. Steroid Biochem. Mol. Biol. 108, 300–309.
Hypothalamic-pituitary–adrenal axis dysfunction in Alzheimer's disease: lack of
association between longitudinal and cross-sectional findings. Am. J. Psychiatry 155,
286–289.

You might also like