You are on page 1of 45

Progress in Neurobiology 98 (2012) 99–143

Contents lists available at SciVerse ScienceDirect

Progress in Neurobiology
journal homepage: www.elsevier.com/locate/pneurobio

The neurobiology of depression in later-life: Clinical, neuropsychological,


neuroimaging and pathophysiological features
Sharon L. Naismith *, Louisa M. Norrie, Loren Mowszowski, Ian B. Hickie
Ageing Brain Centre, Clinical Research Unit, Brain & Mind Research Institute, University of Sydney, Sydney, NSW, Australia

A R T I C L E I N F O A B S T R A C T

Article history: As the population ages, the economic and societal impacts of neurodegenerative and neuropsychiatric
Received 19 December 2011 disorders are expected to rise sharply. Like dementia, late-life depressive disorders are common and are
Received in revised form 3 May 2012 linked to increased disability, high healthcare utilisation, cognitive decline and premature mortality.
Accepted 9 May 2012
Considerable heterogeneity in the clinical presentation of major depression across the life cycle may
Available online 17 May 2012
reflect unique pathophysiological pathways to illness; differentiating those with earlier onset who have
grown older (early-onset depression), from those with illness onset after the age of 50 or 60 years (late-
Keywords:
onset depression). The last two decades have witnessed significant advances in our understanding of the
Depression
neurobiology of early- and late-onset depression, and has shown that disturbances of fronto-subcortical
Late-life depression
Late-onset functioning are implicated. New biomedical models extend well beyond perturbations of traditional
Cognitive monoamine systems to include altered neurotrophins, endocrinologic and immunologic system
Neuroimaging dysfunction, inflammatory processes and gene expression alterations. This more recent research has
Genetic highlighted that a range of illness-specific, neurodegenerative and vascular factors appear to contribute
to the various phenotypic presentations. This review highlights the major features of late-life depression,
with specific reference to its associated aetiological, clinical, cognitive, neuroimaging, neuropathological,
inflammatory and genetic correlates. Data examining the efficacy of pharmacological, non-
pharmacological and novel treatments for depression are discussed. Ultimately, future research must
aim to evaluate whether basic biomedical knowledge can be successfully translated into enhanced
health outcomes via the implementation of early intervention paradigms.
ß 2012 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 100


2. Neural circuitry relevant to depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101
3. Phenotypic features of late-life depression. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103
3.1. ‘Vascular’ depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
3.2. Risk factors for ‘vascular’ and late-onset depression. . . . . . . . . . . . . . . . . . . . . . ......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 105
3.3. Depression as a cardiovascular risk factor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106
3.4. Depression and illness burden . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 106
4. Neuropsychological features of late-life depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
4.1. Predictors of cognitive decline . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ......... . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 108
4.2. The relationship between late-life depression, mild cognitive impairment and dementia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109

Abbreviations: 5HTTLPR, serotonin transporter gene; 5-HT, serotonin; ACC, anterior cingulate cortex; AD, Alzheimer’s disease; BDNF, brain-derived neurotrophic factor;
BOLD, blood oxygen level dependent; Cho, choline; CM, cerebral metabolism; CNS, central nervous system; Cr, creatine; CVD, cardiovascular disease; DLPFC, dorsolateral
prefrontal cortex; DTI, diffusion tensor imaging; ECT, electroconvulsive therapy; EoD, early-onset depression; FA, fractional anisotropy; FDG, fluorodeoxyglucose; fMRI,
functional magnetic resonance imaging; HDL, high density lipoprotein; HMPAO, hexamethyl-propylene amine oxime; HPA, hypothalamic pituitary axis; LLD, late-life
depression; LoD, late-onset depression; MCI, mild cognitive impairment; MD, major depression; MEG, magnetoencephalography; mI, myoinositol; MRI, magnetic resonance
imaging; MRS, magnetic resonance spectroscopy; MTHFR, methylenetetrahydrofolate reductase; MTR, magnetization transfer ratio; NAA, N-acetyl aspartate; OFC,
orbitofrontal cortex; PET, positron emission tomography; PFC, prefrontal cortex; PiB, Pittsburgh B; rCBF, regional cerebral blood flow; RCT, randomised controlled trial; SPECT,
single photon emission computed tomography; SSRI, selective serotonin reuptake inhibitor; VRFs, vascular risk factors; WMLs, white matter lesions.
* Corresponding author at: Clinical Research Unit, Brain & Mind Research Institute, 94 Mallett Street, Camperdown NSW, Australia. Tel.: +61 2 9351 0781;
fax: +61 2 9351 0855.
E-mail address: sharon.naismith@sydney.edu.au (S.L. Naismith).

0301-0082/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.pneurobio.2012.05.009
100 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

5. Structural neuroimaging studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110


5.1. Volumetric findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
5.1.1. Whole brain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
5.1.2. Frontal lobe . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 110
5.1.3. Limbic regions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111
5.1.4. The striatum . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
5.1.5. Significance of volumetric changes in LLD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
5.2. White matter changes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
5.2.1. Macrostructural findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 113
5.2.2. Microstructural findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
6. Macromolecular findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
6.1. Magnetization transfer ratio. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
6.2. Magnetic resonance spectroscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 116
7. Functional neuroimaging studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
7.1. Positron emission tomography (PET) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 117
7.1.1. Associations with clinical features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 118
7.1.2. Medication and treatment effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 118
7.1.3. Summary of PET studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
7.2. Single photon emission computed tomography (SPECT). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
7.2.1. Associations with clinical features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
7.2.2. Medication effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
7.2.3. Relationship to cognition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
7.2.4. Summary of SPECT studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
7.3. Functional magnetic resonance imaging (fMRI) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
7.3.1. Behavioural and cognitive paradigms. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120
7.3.2. Summary of fMRI studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
7.4. Resting state fMRI. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
8. Neuropathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
9. Genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122
9.1. Brain-derived neurotrophic factor gene (BDNF) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
9.2. Serotonin transporter gene (5HTTLPR) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
9.3. Apolipoprotein E gene (Apoe) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
9.4. Genes implicated in systemic vascular risk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 125
10. The role of inflammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 125
11. Prognosis in late-life depression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127
12. Treatment approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
12.1. Pharmacological treatments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
12.2. Non-pharmacological treatments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
13. A focus on prevention for late-life depression? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130
14. Summary and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131

1. Introduction ideation), appetite disturbance or weight change, loss of libido,


sexual dysfunction, non-localized pain, low energy, altered sleep–
As the population ages, the economic and societal impacts of wake cycle and daytime fatigue. Indeed, disturbances of sleep
neurodegenerative and other neuropsychiatric disorders are appear to be linked directly to cognitive impairment (Cho et al.,
expected to rise sharply (Access Economics, 2009; Bloom et al., 2008; Dew et al., 1997; Naismith et al., 2009b, 2011b). Hence, new
2011; Smith, 2011). While dementia is emphasized, late-onset treatments recognise the significance of incorporating sleep and
depressive disorders are also common and disabling (Hickie and circadian realignment into disease management (Hickie and
Scott, 1998; Naismith et al., 2007). Hence, these conditions are now Rogers, 2011).
the focus of considerable public health and clinical attention There is considerable heterogeneity in the clinical presentation
(Hickie et al., 2006; Highet et al., 2002; Kessler et al., 2010; of MD across the life cycle (Hickie et al., 2009). Younger patients
Naismith et al., 2009b; Ustun et al., 2004). The prevalence of may have clinical profiles characterized by high trait anxiety and
clinically significant depressive syndromes in those people over 60 quite variable patterns of circadian, sleep, energy and appetite
years of age (i.e. ‘late-life depression’ or LLD) ranges from 9% to 18% disturbance (Hansell et al., 2011). Those in mid-life present the
and incidence rates of 19.3 per 1000 person years have been more stereotypic picture of anhedonia in combination with sleep
reported (Beekman et al., 1995; Luijendijk et al., 2008; Mulsant and disturbance, weight loss and cognitive and motor impairments
Ganguli, 1999). LLD has been linked to increased rates of suicide (American Psychiatric Association, 1994). In later-life, clinical
and premature mortality (Gareri et al., 2002) and more frequent phenotypes are again more variable (Blazer, 2003). This may well
use of health care with significantly higher health care costs reflect different neuropathological pathways to illness – largely
(Katon, 2003). differentiating those with earlier onset who have grown older from
The core symptoms of major depression (MD) are persistently those who are experiencing clinical depression for the first time.
depressed mood or anhedonia (i.e. loss of pleasure in normal daily Certainly, modelling of genetic and environmental risk factors over
activities). Typically patients also report cognitive impairment the life course suggest distinctly differing pathways in early, mid
(slowed reaction time, poor concentration and memory), dysfunc- and later-life (Gillespie et al., 2004). It would appear that the
tional thoughts (e.g. inappropriate guilt, worthlessness, suicidal relevance of genetic risk to vascular risk factors particularly
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 101

increases with age (especially after age 50 years), while those potential benefits of prevention and early intervention across the
associated with earlier onset forms of depression decline as people life cycle (Insel, 2009b). Clinical staging models for psychotic and
grow older (particularly after age 20 years (Kendler et al., 2009, severe mood disorders (Hickie et al., in press; McGorry et al., 2006)
2008)). have highlighted the need to recognise early clinical phenotypes,
Consequently, treatment responsiveness and longitudinal and related biomedical markers, so that treatments can be
course also vary considerably (Hybels et al., 2009). Additional provided before progression of illness and disability become
clinical concepts such as ‘‘depression without sadness’’ (Blazer, well-established. While these models have not yet been fully
2003) and a depletion syndrome characterized by withdrawal, elaborated for the more common forms of depression, available
apathy and lack of vigour (Adams, 2001) have also been described. data support the notion that cognitive change and disability
Most importantly, for those with the more severe forms of LLD emerge early in the illness course (Hamilton et al., 2011; Hermens
(particularly those of late-onset), neuropsychological deficits et al., 2011).
including executive dysfunction and observable psychomotor A major advance in detailing the various pathophysiologies that
change are prominent (Hickie et al., 2007a; Naismith et al., underpin the clinical phenotype of MD has been to focus on
2003). The risk of progression to permanent cognitive impairment differential pathways to illness. Age of onset has emerged as the
in this type of depression has also been emphasized (Alexopoulos, most robust correlate of these putative trajectories. A key
2005; Baldwin et al., 2006; Kohler et al., 2010a). Minor depression differentiation is between those disorders that have their onset
(depressed mood accompanied by one to three additional early in life (early-onset depression, EoD, typically with first
symptoms), has been found to be more prevalent in older adults symptoms emerging in the adolescent and early adult periods)
than full threshold MD. However, these ‘sub-threshold’ disorders compared with those that emerge for the first time after the age of
are associated with impairments similar to that of MD, including 50 or 60 years (late-onset depression, LoD) (Hickie et al., 2009;
decreased levels of physical function, poorer self-rated health and Insel, 2009b; Krishnan and Nestler, 2010). Despite extensive data
increased days lost due to disability (Beekman et al., 1995). demonstrating that in LLD there are many modifiable risk factors
While a wide range of pharmacological and psychological for illness onset, ‘pre-emptive’ or ‘early intervention’ approaches
treatments for MD are moderately effective, as many as 50% of have not yet been operationalised or extensively evaluated.
patients across all age groups will not achieve remission with their Compared with EoD, the neurobiological models of LoD intrinsi-
first treatment (Roose and Schatzberg, 2005). Ultimately, 20–30% cally place less emphasis on the developmental determinants of
will not achieve full recovery despite access to multiple interven- amygdala circuitry, and other frontotemporal changes. Instead
tions (Whyte et al., 2004). Even with optimal treatment, only 34% they highlight the critical nature of intercurrent medical and other
of the total disease burden of MD (measured as ‘years lived with vascular risk factors that result in structural disruptions of key
disability’) can be averted (Andrews et al., 2004). Those persons fronto-subcortical circuits. Further, these more specific models for
who achieve full recovery remain at high risk for further episodes LoD are not only relevant to onset but have major ramifications for
(Maj et al., 1992). Recovery may well be slower in older patients determining response to treatment, development of new inter-
with conventional treatments (Wilhelm et al., 1994). Hence, ventions and longitudinal course. Importantly, the persistence or
ongoing disability in this group and reliance on other formal and progression of these illness pathways put patients at high risk for
informal caring networks is high (Alexopoulos et al., 2002a; chronic disability and possible development of dementia (Schweit-
Beekman et al., 1997; Steffens et al., 1999a). Recurrence rates in zer et al., 2002; Sheline et al., 2010).
community dwelling elders are more than three times as high as This review highlights the major features of LLD (including
incidence rates (Luijendijk et al., 2008). LoD), with specific reference to its associated aetiological, clinical,
Although much remains to be discovered about the pathophys- cognitive, neuroimaging, neuropathological, inflammatory and
iology of both early and later-onset clinical depression, the last two genetic correlates. Data examining the efficacy of various
decades have seen significant advances. Neurobiological theories pharmacological, non-pharmacological and novel treatments for
have been greatly informed by detailed genetic, neuroimaging, depression will then be discussed with a view to highlighting the
neuropsychology, pathology and interventional studies. These future directions for both research and clinical practice. Depression
indicate that structural and functional abnormalities within key in dementia will not be examined.
fronto-subcortical neural networks underpin many of the clinical
phenomena (Mayberg, 1997, 2007). Concurrently, advances in 2. Neural circuitry relevant to depression
molecular, cellular and systems neuroscience have enriched more
advanced biomedical models. New models extend well beyond The fronto-subcortical model of MD requires the consideration
perturbations of traditional monoamine systems to include altered and integration of the functional neuroanatomical circuits that
neurotrophins, endocrinologic and immunologic system dysfunc- were eloquently described by Alexander in 1986 (Alexander et al.,
tion, inflammatory processes and gene expression alterations 1986). While the motor circuit is perhaps the best understood of
(Belmaker and Agam, 2008; Drevets et al., 2008a; Hickie et al., the networks, the other loops appear to be organised in parallel and
2001a, 1999, 2007b; Insel, 2009a; Krishnan and Nestler, 2010; are termed the oculomotor, dorsolateral prefrontal, lateral
Mayberg, 2003). orbitofrontal and anterior cingulate loops (Alexander et al.,
Genomic variations and early brain development are now 1986; Cummings, 1993; Saint-Cyr, 2003), each of which are
viewed as fundamental factors that ultimately bias individual convergent upon different frontal regions. Behavioural markers of
brain circuitry towards illness resilience or vulnerability (Insel, each circuit can be reproduced by lesions at various points along
2009a). The influence of these intrinsic factors is then shaped by the path, from the cerebral cortex, white matter, or subcortical
ongoing environmental exposures. These extend well beyond the nuclei. The circuits are also discrete (i.e. non-overlapping) and
initial intra-uterine and early childhood period. Specifically, it has spatially constrained. They are widely separated at the level of the
become increasingly clear that major deviations in brain develop- cortex. However, at subcortical levels, the circuits are closer in
ment continue to occur throughout the adolescent and early adult proximity. Thus, subcortical lesions can impair multiple circuits
years (Giedd et al., 1999) in association with the onset of major simultaneously, producing a varied array of symptoms (Royall
psychiatric disorders. Hence, the concept of ‘pre-emptive’ psychi- et al., 2002). Due to the dense interconnections between the
atry has evolved to link evidence of the developmental basis of the circuits, disruption at any level of the fronto-subcortical circuit
major mental disorders with clinical studies focusing on the could result in dysfunction in other areas (Soares and Mann, 1997).
102 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

Thus, in concert with the presence of genetic or other predisposing caudate and putamen receive input from the limbic and paralimbic
factors, such disruption could indirectly or directly contribute to a cortex, hippocampus and amygdala. Early studies noted that there
constellation of depressive, cognitive or movement phenotypes was a higher incidence of depressive disorders in patients with
(Drevets, 1998; Hickie et al., 1997a; Soares and Mann, 1997). lesions or strokes in the basal ganglia or its surrounding regions
Of the five circuits described, at least three are likely to be (Cummings, 1985; Goodwin, 1997). Similarly, diseases primarily
relevant to depression (e.g. see Drevets, 1998, 2000 for reviews) affecting the basal ganglia are often accompanied by depressive
namely those that incorporate the orbitofrontal cortex (OFC), symptoms. For example, MD is the most common psychiatric
dorsolateral prefrontal cortex (DLPFC) and anterior cingulate (ACC) disturbance of Parkinson’s disease, is often a prodromal feature
regions. The OFC is a component of the paralimbic cortical circuit and is evident in 17–25% of patients (see review by Slaughter et al.,
that is involved in several higher-order association functions 2001). Although the basal ganglia have traditionally been
including the integration of emotion, behaviour and various considered to be primarily involved in motor functioning,
sensory processes. In addition to thalamic connections, the OFC has increasing recognition is being given to their role in the integration
reciprocal connections with the amygdala, the temporal neocortex of emotional, cognitive and motor behaviour (Balleine and
and the insula, and has dense connections with the ventromedial O’Doherty, 2010). Specifically, animal studies have highlighted
caudate nucleus. The DLPFC is connected to the ACC, amygdala and the critical role of the dorsomedial and dorsolateral striatum in
dorsolateral caudate nucleus. The ACC circuit receives input from goal-directed action and habit learning respectively (see review by
the hippocampus, amygdala and paralimbic cortex and projects to Balleine and O’Doherty, 2010). While some human studies have
the ventral striatum and nucleus accumbens (Byrum et al., 1999; certainly confirmed the regional specificity of the basal ganglia
Cummings, 1993; Royall et al., 2002). More recently the ACC has (e.g. anterior caudate) in some components of cognition (O’Doh-
been identified as of critical importance in the pathophysiology of erty et al., 2004), this field is in its infancy and no known studies
depression via its two major subdivisions: the dorsal ACC, which have explored these potential relationships in depressive dis-
contributes to a ‘dorsal’ network including the hippocampus and orders. Ultimately, demarcation of specific regions of the basal
DLPFC, and the perigenual ACC, which projects to the amygdala, ganglia that are anatomically and functionally discrete may
hypothalamus and OFC amongst others, comprising the ‘ventral’ provide greater insights into their role in cognitive and/or mood
network (Phillips et al., 2003). The dorsal ‘cognitive/executive’ disturbance.
network is postulated to be important for executive functions, and Comprehensive neurobiological theories of MD need to
plays a key role in the cognitive regulation of emotional responses encompass not only neural circuitry but also the various
whereas the ventral or ‘affective/emotional’ network appears to be neurotransmitter systems. Regardless of their origin, aspects of
involved in assessing the salience of emotional input as well as the fronto-subcortical circuitry funnel information from widespread
generation and regulation of emotional response (Phillips et al., cortical areas and utilise a vast array of neurotransmitters. In this
2003). The fundamental role played by limbic structures is regard, there has been considerable preoccupation with the
illustrated in their dense interconnections within these key neural monoamine hypothesis of depression and accordingly much
circuits, as well as the associations between specific regions and experimental, neuroimaging and treatment-based research has
clinical features of mood disorder. In particular, the amygdala, been focused on serotonergic, noradrenergic and/or dopaminergic
extensively connected to the hippocampus, striatum and the neurotransmission (Hickie et al., 2009).
orbital and medial prefrontal cortices, is of primary importance in Much of our knowledge regarding serotonin (5-HT) functioning
assessing the emotional significance of sensory input, and arose from the implementation of techniques such as post-mortem
modulating monoamine and steroid secretion in response to studies and neuroendocrine response to serotonergic challenge.
aversive or novel stimuli (Drevets et al., 2008b). Furthermore the Such studies revealed deficits in 5-HT1A (frontal regions and
hippocampus, lying at the junction of the dorsal and perigenual temporal pole) and in 5-HT2 binding (frontal, temporal and parietal
ACC as well as the neuroendocrine regulatory networks (specifi- regions) (Bowen et al., 1989). However, as noted by Meltzer et al.
cally the hypothalamic pituitary axis), is a vulnerable region, and (1998) in their review of serotonin in LLD, there are many
extensive evidence points to its key role in mood disorders, inconsistencies in the post-mortem studies, possibly due to
including the memory deficits commonly observed in LLD (Hickie confounds such as use of antidepressant therapies, differences
et al., 2005b; Steffens, 2008; Steffens et al., 2011). in post-mortem delay, tissue preparation, selection of markers for
Recent data suggests that improvements in cognitive function specific receptor subtypes, participant selection bias and the
are associated with discrete functional networks that are separate various mechanisms of action of prior antidepressant therapy (i.e.
from those associated with improvements in mood (Diaconescu agonists versus antagonists). Other studies utilising cerebrospinal
et al., 2010). However, further research is required in order to fluid (e.g. Jones et al., 1990) have, however, confirmed that
elucidate whether abnormalities within specific or multiple loops serotonin is implicated in MD, though there has been a lack of
are associated with different phenotypes of depressive disorder, correlation with clinical symptoms. More recently, serotonin
including those with clear genetic vulnerabilities to early-onset or pharmacologic challenge studies (using serotonin releasing agent
late-onset forms. While some data support the notion that certain fenfluramine) (e.g. Lerer et al., 1996) have confirmed blunted
neurobiological changes (e.g. frontal and limbic) relate to the prolactin response in MD that may also differ according to age,
duration of illness and lack of access to appropriate treatments suggesting an increased vulnerability to MD in older people.
(Drevets et al., 2008a), there has been a paucity of studies More recently, the interactions of the various neurotransmitter
commencing prior to illness onset to fully elucidate the contribu- systems have received increased attention. For instance, via five
tion of neurodevelopmental versus illness-specific processes. different postsynaptic receptors, dopamine has both inhibitory and
In addition to the prefrontal and limbic circuitry, there is excitatory effects on fronto-subcortical functioning. Dopaminergic
considerable data supporting the involvement of the basal ganglia connections between the substantia nigra and limbic system play a
and specifically, the striatum, in the pathophysiology of MD. The role in the integration of emotional input and motor activity,
striatum is comprised of both input and output nuclei, and cortical cognition and motivation. Both cholinergic and serotonergic
input to the striatum has a regional pattern. Whereas the putamen systems are involved in the modulation of these dopaminergic
is primarily innervated by the sensorimotor cortex, the caudate pathways. Additionally, glutamatergic corticostriatal and thala-
nucleus receives preferential input from the frontal, temporal, mocortical projections, via interactions with both cholinergic and
parietal and cingulate cortex. Additionally, ventral parts of the dopaminergic systems, contribute to a negative feedback loop,
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 103

which serves to limit cortical overstimulation (see review by Tekin disease (see Section 3.1) have a poorer response to antidepressants,
and Cummings, 2002). The neurotransmitter systems are begin- particularly SSRIs. In these sufferers with likely frontostriatal
ning to be incorporated into theories of neuroplasticity. For impairments, dopamine-acting agents or psychotropics with
instance serotonin is important in the regulation of N-methyl-D- catecholaminergic activity may assist recovery (Andreescu and
aspartate receptor activity, which in turn modulates synaptic Reynolds, 2011).
plasticity (Bennett, 2010). Furthermore, functional imaging in LLD
patients treated with the selective serotonin reuptake inhibitor 3. Phenotypic features of late-life depression
(SSRI) citalopram implicates an interaction between subcortical
dopamine and serotonin systems with cortical glutamatergic Although there are certainly commonalities in depressive
function (Diaconescu et al., 2010). features throughout the age spectrum, LLD may have distinct
The relevance of considering the interactions of various medical, clinical, cognitive, neuroimaging, neuropathological,
neuropharmacological systems in MD is underscored by the inflammatory and genetic features when contrasted with depres-
increasing availability of dual acting antidepressant agents that sion in younger people (Alexopoulos, 2005; Brodaty et al., 2001;
target multiple systems. For example, the availability of selective Herrmann et al., 2007; Hickie and Scott, 1998; Hickie et al., 2009;
noradrenergic and serotonergic agents has allowed for more Thomas et al., 2009). Upon considering the clinical phenotype of
personalised approaches to treatment, while more recently, there LLD, it is important to note that multiple underlying pathophy-
has been increasing emphasis on considering the specific siologies may be operative (see Table 1) including the long-term
phenotypic features of the depressive disorder (Hickie et al., illness-specific effects of EoD, the underlying brain changes that
2009; Hickie and Rogers, 2011). This is especially relevant in LLD, might give rise to LoD (see Fig. 1a), and the potential role of
where each patient presents with a unique constellation of underlying prodromal neurodegenerative disease. While the age
depressive, cognitive and somatic symptoms, often complicated used to define EoD versus LoD tends to vary across studies, age 50,
by concomitant medical morbidity (Hickie et al., 2009). For 60 and 65 are commonly used. Regardless of the cut-off employed,
example, those with predominant underlying cerebrovascular the concept of LoD has been a catalyst for a plethora of research

Table 1
Clinical, aetiological, and prognostic features of early-onset and late-onset depression.

Early-onset depression Late-onset depression

‘Vascular’ depression Neurodegenerative

Description  Development of depressive and/or anxiety  Development of depression after age 50 or 60  May include more pronounced hippocampal
symptoms early in life, typically before age years and temporal lobe change, particularly with
25  Profile indicative of fronto-subcortical underlying Alzheimer’s pathology
 Profile generally indicative of frontotemporal dysfunction
and amygdala change
Clinical features  Anxiety features  Psychomotor change  May additionally include emerging changes
 Prominent heritability  Executive dysfunction in memory consolidation and language
 Development of awareness to social cues  Apathy
 Treatment resistance
 Absence of family history
Relationship to  Depression is a risk factor for cardiovascular  Increased rates of:  Cerebrovascular disease may play a role in
cardiovascular disease and adverse outcomes including * Cerebrovascular disease risk for depression, cognitive decline and
disease stroke and myocardial infarction * Hypertension more rapid progression of Alzheimer’s
* Diabetes pathology
* Heart disease
* Hypercholesterolaemia
* White matter lesions on neuroimaging
Genetic  Gene susceptibility most operative early in  Genetic susceptibility relates to systemic  For those whose depression occurs in the
vulnerabilities development, with vulnerability emerging in vascular risk, associated with context of incipient underlying
childhood and adolescence methylenetetrahydrofolate and serotonin neurodegenerative change, genetic
transporter genes associations between BDNF and/or ApoE and
volumetric reductions in key brain regions
may be important
Prognosis  Possibly longer time to remission after  Poorer treatment outcomes
treatment with antidepressants  Associated with increased mortality and progression to dementia
 Presence of white matter lesions and cognitive impairment, particularly executive functioning
may predict poor prognosis
Treatment  Possible preferential response to  Management and reduction of vascular risk  Consider role of acetylcholinesterase
serotonergic agents  Possible preferential response to therapies inhibitors
 High-rate of non-response to initial increasing dopamine and/or noradrenaline  Non-pharmacological therapies (e.g.
antidepressant  Consider antidepressant augmentation with cognitive training, problem solving therapy)
 May require lithium augmentation Ca channel blockers may be useful when cognitive dysfunction is
 Non-pharmacological therapy (e.g. cognitive  Additional clinical features such as present
behavioural therapy) useful in targeting melancholia and psychosis may require
anxiety or dysfunctional thoughts consideration of tricyclics, lithium,
antipsychotics and electroconvulsive
therapy
 Non-pharmacological therapies (e.g.
cognitive training, problem solving therapy)
may be useful when cognitive dysfunction is
present

Note: Late-life depression includes those with early-onset depression, with recurrent episodes throughout life, as well as those in whom depression has arisen for the first
time after the age 50 or 60. For the latter group, cerebrovascular disease or early neurodegenerative changes may also contribute to the phenotypic profile.
104 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

Fig. 1. (a) Clinical and cognitive correlates of neurobiological changes documented in late-life depression. (b) Neuroimaging and neuropathological changes documented in
late-life depression. Abbreviations: BDNF, brain-derived neurotrophic factor; 5HTTLPR, serotonin transporter; apoE, apolipoprotein E; 5-HT, serotonin; BOLD, blood oxygen
dependent level; cho, choline; Cr, creatine; GFAP, glial fibrillary acidic protein; DTI, diffusion tensor imaging; fMRI, functional magnetic resonance imaging; mI, myoinositol;
MRI, magnetic resonance imaging; MRS, magnetic resonance spectroscopy; MTI, magnetization transfer imaging; NAA, N-acetyl aspartate; PET, positron emission
tomography; rCBF, regional cerebral blood flow; SPECT, single photon emission tomography; WMLs, white matter lesions. *Includes studies reporting non-specific prefrontal/
frontal changes.
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 105

that has sought to identify the pathophysiological mechanisms other, the role of neuropsychologically determined cognitive
underlying those forms of depression emerging in later-life. In impairments, such as encompassed in the ‘‘depression-dysexecu-
addition to identifying unique clinical features such as absence of a tive syndrome’’ (Alexopoulos et al., 2002c). In addition to having
family history of affective disorder, less personality dysfunction later ages of onset, those with ‘vascular’ depression also tend to be
and more psychomotor change (Brodaty et al., 1997; Hickie et al., older, have treatment resistance, psychomotor change, less family
1995), this research has identified the preponderance of white history of affective disorder, cognitive impairment (particularly
matter lesions (WMLs), seen as ‘hyperintense’ on T2-weighted executive dysfunction) and greater progression to dementia
images. Such lesions have been linked to LLD generally, and more (Hickie and Scott, 1998; Sheline et al., 2010; Steffens and Krishnan,
specifically to those with LoD (e.g. Alexopoulos et al., 1997; Hickie 1998). There is now strong, though not unanimous (Almeida,
et al., 1995; Krishnan et al., 1997; O’Brien et al., 1996) (see 2008), support for the concept of ‘vascular’ depression. With the
structural neuroimaging Section 5.1). Although in this review we ongoing sophistication of neuroimaging techniques, this area
have primarily focused on early versus late-onset of disease, it is continues to be the subject of empirical research. In particular, it
nonetheless important to recognise the limitations of this now aims to better characterize the pathophysiological mecha-
categorical approach to disease conceptualisation. In particular, nisms and disruptions to neural circuitry predisposing to or
one must consider the selection factors that may be operative perpetuating depressive syndromes. Currently, it appears as
when comparing the two phenotypes. That is, those with severe though some degree of lesion burden is required in order to
illnesses may have had high mortality, and premature death due to significantly contribute to depressive syndromes, although this is
several factors such as increased medical comorbidity, substance certainly not absolute and further research will be needed to
misuse, vascular disease, and suicide (Bunker et al., 2003; Shah identify whether the strategic location of lesions or the overall
et al., 2011). Of those who survive, many of these factors, as well burden of lesions is pertinent (Sheline et al., 2008; Tupler et al.,
other detrimental effects may have been operative to impact on 2002). Certainly the literature regarding risk of post-stroke
brain functioning (e.g. neuroendocrine, inflammatory and immu- depression based on stroke location is inconsistent (Bhogal
nological changes, medication effects). Additionally, in the clinical et al., 2004). The observation that many older people may have
context, it can be extremely difficult to ascertain retrospectively cerebrovascular disease and yet do not develop depressive
the timing of depression onset, and commonly, depression in early syndromes also raises the question regarding possible neurobio-
life may have escaped formal medical recognition or treatment, an logical and/or individually specific (e.g. personality) protective
effect which may be less pronounced in future cohorts with greater factors. Additionally, there is some suggestion that VRFs do not
public health recognition, reduced stigmatisation of depression account totally for WML severity, thus suggesting that other non-
and increased rates of treatment for depression (Hickie, 2004; vascular factors may contribute to lesion burden (Teper and
Hickie et al., 2005c, 2009; Highet et al., 2006). Despite these O’Brien, 2008). Nonetheless, it is widely accepted that regardless of
limitations, however, the differentiation of LLD phenotypes has their aetiology, WMLs confer vulnerability to mood disorders by
proved immensely useful, in terms of delineating the pathophysi- disrupting key fronto-subcortical circuitry (see Fig. 1b). In addition
ology and neurobiological profile of LLD. Techniques aimed at to WMLs, a pre-existing vulnerability to depression may be
investigating features linked with hypothesized pathophysiology, necessary as well as environmental stressors.
that do not rely on broad age cut-offs, may now be more useful in
clarifying depression phenotypes and the various pathways to 3.2. Risk factors for ‘vascular’ and late-onset depression
illness onset and longitudinal decline (Box 1).
Regarding individual VRFs for ‘vascular’ depression and LoD,
3.1. ‘Vascular’ depression this literature is extensive and has been the subject of many review
papers (e.g. Alexopoulos, 2003; Camus et al., 2004; Thomas et al.,
While the precise aetiology of WMLs is unclear, it is likely that 2004). In brief, risk factors are those generally considered to be
they predominantly reflect underlying cerebrovascular disease risks for cardiovascular disease (CVD) including hypertension,
including small vessel ischaemic change (Greenstein et al., 2010; diabetes, hypercholesterolaemia, heart disease, obesity and
Thomas et al., 2002). The strong link between WMLs, vascular risk smoking. Data supporting the link between depression and CVD
factors (VRFs) and depression, led to the ‘vascular’ depression stems from both clinical as well as community and epidemiological
hypothesis (Alexopoulos et al., 1997; Hickie et al., 1995; Krishnan studies and further suggests that this relationship is bidirectional.
et al., 1992; Steffens and Krishnan, 1998). Approaches to Hypertension may be particularly relevant to the aetiology of LLD
characterizing ‘vascular’ depression have on the one hand due to the chronic effects of this condition on the small vessels of the
emphasized the importance of neuroimaging changes, ‘‘MRI- brain. However, there are some mixed findings in this area. For
defined vascular depression’’ (Krishnan et al., 2004), and on the instance, a large population-based study (part of the Cardiovascular
Health study, n = 3660) showed that higher CES-D scores (Centre for
Epidemiological Studies Depression Scale) were significantly
associated with the presence of hypertension (Steffens et al.,
Box 1. Clinical characteristics of late-life depression.
1999a). By contrast, some cross-sectional or short-term prospective
 Depression in adults over age 60 studies have not supported this relationship (Kim et al., 2006; Mast
 Fronto-subcortical circuitry abnormalities et al., 2008; Stewart et al., 2001). It is worth noting that hypertension
 May be classified into early-onset or late-onset, but often this is a less stable construct than some other VRFs and this may explain
is difficult to establish conclusively some of the variability in findings. For instance, blood pressure
 Late-onset phenotype may relate to underlying cerebrovas- varies in response to physiological and psychological determinants,
cular disease, evidenced by greater vascular risk factors or and its treatment may also modify depression risk. Additionally, the
white matter lesions on neuroimaging
relationship between hypertension, WMLs and depression may be
 Late-onset phenotype may have more pronounced cognitive
confounded by the presence or absence of pre-existing treatment for
impairment and greater progression to dementia syn-
dromes hypertension. Importantly, data suggests that the cerebral damage
 Late-onset phenotype may have greater treatment resis- induced by VRFs is likely to be irreversible if management is
tance commenced too late, thus potentially confounding many cross-
sectional studies (see Naismith et al., 2009a for a review). In this
106 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

regard prospective longitudinal studies have been informative and 2006) as well as elevated platelet reactivity, or increased immune
have generally supported the association between the accumulation system reactions. Depression is also a risk factor for poor outcome
of VRFs and depression in older adults (e.g. Hickie et al., 2003; Lyness of established CVD (Lichtman et al., 2008), which may in part be
et al., 2000; Mast et al., 2008). due to the above or behavioural risk confounders such as smoking,
A marker of CVD that has been less well studied is carotid lack of exercise or poor adherence. Fewer studies exist that have
intima-media thickness, which has been found to correlate well investigated prospectively the relationship between depression
with atherosclerosis (Bots et al., 1997). Studies in middle-aged and and individual VRFs. Depression has, however, been found to be a
older adults have shown that common carotid intima-media risk factor for diabetes (Knol et al., 2006), though the converse is
thickness is higher in those with LoD depression and is related to not always evident (Kim et al., 2006).
WMLs (Chen et al., 2006), but not to depression severity (Smith While the underlying mechanisms for the bidirectional
et al., 2009c). Whilst carotid intima-media thickness provides relationship between CVD and LLD remain unclear, the link is
some indication of underlying macrovascular disease, measures of certainly plausible when one considers that they share environ-
retinal vasculature (arteriolar and venular diameters) provide a mental risk factors. Interestingly, however, the relationship
marker for cerebral microvascular disease (Ikram et al., 2006). between the two remains strong even when such confounding
Larger retinal venular diameters have also been associated with factors are strictly controlled (Rugulies, 2002). It is likely that
markers of atherosclerosis including raised levels of total shared genetic factors may be important, and in this regard, the
cholesterol and lowered high density lipoprotein (HDL) cholester- methylenetetrahydrofolate reductase (MTHFR) gene (see relation-
ol, smoking as well as inflammatory markers such as C-Reactive ship with genetics in Section 9) has received considerable
Protein (CRP) (Ikram et al., 2004). Moreover, larger retinal venular attention. This gene is involved in homocysteine metabolism;
diameters have been associated with marked progression of WMLs raised levels are considered to be a VRF and contribute to CVD
over a 2-year follow-up period, as evidenced by the Rotterdam (Clarke et al., 1991). Homocysteine is derived from the demethyl-
Study of Aging (7046 adults aged >55 years without dementia) ation of the amino acid methionine. It is metabolized via
(Ikram et al., 2006). Interestingly this association remained after remethylation using B12 and folic acid as co-factors, and cleared
adjustment for VRFs. The relationship between venular diameter via transulfuration to cysteine and glutathione, a process requiring
and WML progression was most attenuated when CRP levels were B6 and B12 (Folstein et al., 2007). A number of studies have
taken into account. The authors thus suggested that this may variously supported the link between homocysteine and LLD
indicate a role for inflammation in the pathogenesis of cerebral generally (Almeida et al., 2008; Bjelland et al., 2003), LoD
small vessel disease (Ikram et al., 2006). However further analysis specifically (Chen et al., 2005) and some have shown a link
of this cohort has not demonstrated any relationship between between the MTHFR gene mutation and LoD (Hickie et al., 2001a).
retinal markers of cerebral microvascular disease and incident LLD The mechanism for this association may lie in cerebral microvas-
(Ikram et al., 2010), results which confirm the findings of two culature changes, via oxidative stress-related endothelial dysfunc-
similarly negative cross-sectional studies (Nguyen et al., 2008; Sun tion (i.e. the impairment of the vascular endothelium’s ability to
et al., 2007). control vasomotor tone) (Welch and Loscalzo, 1998), or via direct
Finally, although not strictly a VRF, stroke is associated with an neurotoxic effects, related to DNA strand breakage or apoptosis
increased risk for depression (see review by Robinson and Spalleta, (Santos et al., 2009). A recent study found that in older
2010), although the data on the role of lesion location and risk for hypertensives, plasma homocysteine levels were associated with
depression is inconsistent (Bhogal et al., 2004). In some studies, up increased rates of progressive WMLs and hippocampal atrophy
to 40% of patients will develop depression in the acute post-stroke (Firbank et al., 2010). However, data from small clinical studies of
period (from a few weeks to 6 months) (see review by Robinson patients with LoD has shown that the association between total
and Spalleta, 2010). Stroke lesions are associated with depletion of plasma homocysteine levels and depression is apparent even after
monoamines, as well as increased production of pro-inflammatory controlling for WMLs, thus suggesting that the relationship
cytokines and cortisol (for review, see Spalletta et al., 2006). Much between homocysteine and depression is not simply underpinned
recent work has been directed at the prevention of post-stroke by WML (potentially vascular) pathology (Chen et al., 2005).
depression with antidepressants, based on the rationale that by Finally, endothelial dysfunction, involved in the development of
modifying the biological factors such as reduced neurogenesis and atherosclerosis and thrombosis (Davignon and Ganz, 2004;
monoamine alterations, depression risk will be reduced (see Rubanyi, 1993), may contribute to the link between vascular
review by Ramasubbu, 2011). Results have been inconsistent, with disease and depression. In support of this notion, measures of
one meta-analysis supporting the role of antidepressants (Chen endothelial dysfunction have indeed been associated with
et al., 2007), and one showing no benefit (Hackett et al., 2008). depression (Rajagopalan et al., 2001). These have also been linked
with raised cortisol (Ghiadoni et al., 2000) as well as pro-
3.3. Depression as a cardiovascular risk factor inflammatory cytokines thus suggesting that homocysteine
metabolism, endothelial dysfunction and the inflammatory system
Of critical significance, an emerging body of literature suggests may have interdependent roles in the pathogenesis of LLD (Santos
that the relationship between depression and CVD is bidirectional et al., 2009). It is likely that other factors are also important (as
(Schrader et al., 2005; Thomas et al., 2004). Depression has been reviewed by Musselman et al., 1998; Teper and O’Brien, 2008). For
shown to have a relatively strong association with the develop- instance, both platelet activation (Bruce and Musselman, 2005)
ment of CVD as indexed by fatal coronary heart disease or and autonomic dysfunction manifested in heart rate variability
myocardial infarction. This finding has been demonstrated by a (Carney et al., 2005) have been associated with depression.
number of longitudinal studies published over the last 40 years
and is particularly apparent in men (Nicholson et al., 2006). 3.4. Depression and illness burden
Reviews have further indicated that the risk is not confined merely
to major depressive disorder but exists across the range of Although there are bidirectional relationships between depres-
depressive symptom severity. This has been suggested to reflect sion and vascular disease, LLD is also clearly associated with other
either common aetiological factors including genetic risk or medical conditions as well as illness burden in late life. For
differential stress responsivity in the hypothalamic pituitary axis instance, depression is associated with increased risk of low bone
(HPA) amongst those with CVD and depression (McCaffery et al., mineral density (a risk factor for osteoporosis) (Wu et al., 2009),
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 107

and metabolic syndrome and insulin resistance (Laudisio et al., persistence, recurrence, and poor treatment responsiveness
2009). Moreover disorders common in elderly populations may be (Baldwin et al., 2004; Sneed et al., 2007). A recent large-scale
frequently comorbid with depression. For instance, chronic randomised controlled trial found that executive dysfunction, as
obstructive pulmonary disease (Chen et al., 2007), chronic renal well as deficits in processing speed and memory predicted
disease (Hedayati et al., 2012), stroke (Ramasubbu, 2011), treatment remission following a 12-week trial of sertraline
Parkinson’s disease (Naismith and Lewis, 2011), hip fracture (Sheline et al., 2010). Executive dysfunction in LLD is therefore
(Kamholz and Unutzer, 2007) and obstructive sleep apnoea (Harris particularly problematic from a prognostic and clinical perspec-
et al., 2009) all have high rates of depression. As the number of tive. Indeed, the term ‘depression-executive dysfunction syn-
comorbid conditions increases, so does the risk for depression drome’ has been proposed to describe this sub-group with
(Luber et al., 2001). Depression is frequently associated with poor prominent executive and psychomotor change, apathy, dispropor-
clinical outcomes, substantial functional impairment and reduced tionate disability and less guilt and insight (Alexopoulos et al.,
quality of life across a range of common comorbid conditions 2002c). In addition to clinical features, executive deficits are
(Hedayati et al., 2012; Lenze et al., 2004; Naismith et al., 2010a; associated with poor psychosocial functioning and difficulties in
Robinson and Spalleta, 2010). In addition, depression is associated instrumental activities of daily living (Kiosses et al., 2000, 2001;
with increased mortality rates (Penninx et al., 1999); for example, Lockwood et al., 2000; Mackin and Arean, 2009). Recent data also
depression has a negative impact on the mortality associated with shows that financial capacity in LLD is largely predicted by
stroke (Ramasubbu and Patten, 2003), and it increases the risk for executive deficits, rather than by depression severity (Mackin and
all causes of cardiovascular mortality when comorbid with Arean, 2009).
diabetes (Pan et al., 2011). Although no known studies have concurrently linked executive
The relationship between depression and co-occurring medical dysfunction and structural brain changes with treatment resis-
illness is likely to be complex and reciprocal. As for the relationship tance in MD, there is evidence that prefrontal cortex reductions
between LLD and vascular disease, not only are the direct biological may mediate the response to antidepressants, thus adding support
effects associated with comorbidity likely to increase the risk of to the notion that executive deficits are likely to reflect more
depression, but LLD itself is associated with a surfeit of destructive permanent disruption of frontal circuitry (Gunning et al., 2009). It
mediators, such as altered glucose tolerance, HPA dysfunction and is also likely that executive deficits in LLD are reflective of the
a pro-inflammatory milieu (Wolkowitz et al., 2011) as discussed deleterious effects of cerebrovascular disease on critical fronto-
above. In addition, behavioural, social and cognitive components subcortical pathways. Several studies have demonstrated signifi-
may lead to poor adherence with treatment plans, reduced social cant associations between WMLs and performance on measures of
contact or maladaptive behaviours, thereby increasing the risk for executive functioning such as cognitive flexibility and phonemic
physical disorder and illness. fluency (Breteler et al., 1994), working memory, inhibition and task
switching (Kennedy and Raz, 2009). In LLD specifically, Kohler et al.
4. Neuropsychological features of late-life depression (2010b) have reported a significant negative association between
increased ratings of WMLs in the deep white matter and
Neuropsychological (cognitive) deficits in older people with periventricular regions and scores on memory and executive
depression have been well described and predominantly include functioning measures over an 18-month period, in 35 older adults
impairments in processing speed, executive functions (i.e. ‘frontal’ with LLD compared to healthy controls. These authors concluded
or higher-order functions) as well as in aspects of learning and that cerebrovascular disease contributes significantly to the
memory (Herrmann et al., 2007; Kohler et al., 2010a; Naismith persistence of such cognitive impairments in LLD. Additionally,
et al., 2006; Sheline et al., 2006) (see Fig. 1a). Indeed, in 18–57% of other studies have provided more indirect evidence to support the
older patients, these deficits may be of sufficient severity to relationship between cerebrovascular disease and executive
warrant a diagnosis of dementia (Alexopoulos et al., 1993). While it functions. For example, Baldwin et al. (2004) reported significantly
was previously thought that such deficits would abate with impaired executive functioning and a higher WML burden in
adequate treatment, this notion is no longer widely accepted. antidepressant non-responders relative to control and responder
Instead, it appears that at least some degree of cognitive groups; however the association between these two outcomes was
dysfunction may persist with symptom resolution (Devanand not reported. Additionally, Salloway et al. (1996) reported a
et al., 2003). significant correlation between subcortical hyperintensity burden
While not easily detected on gross bedside examinations, and general cognitive impairment, using a cognitive index score
evidence indicates that deficits may be more pronounced in those which comprised the summed standardised scores for all cognitive
with later ages of depression onset (Naismith et al., 2003). Greater tests administered, including measures of executive functioning.
deficits on tests of executive functioning and memory (Kohler Deficits in the domains of processing speed are also prominent
et al., 2010a; Naismith et al., 2003; Rapp et al., 2005; Salloway in LLD. Some research has even suggested that such decrements
et al., 1996) have been reported in association with increasing ages may underpin impairments in other domains of functioning
of onset or in those specifically with LoD (compared to EoD). (Sheline et al., 2006), although this may not extend to measures of
However, some cross-sectional data has shown that the effect of episodic memory (Delaloye et al., 2008). Slowed psychomotor
age of onset on cognition is not significant once age and vascular speed is a key feature of ‘vascular’ depression, and a recent meta-
burden are considered (Lesser et al., 1996; Sheline et al., 2006). In analysis suggested that psychomotor speed may be impaired to the
those with LoD, symptom severity may also be an important same degree as executive function in those with LoD (Herrmann
determinant of the extent of cognitive change (Elderkin-Thompson et al., 2007). In addition to executive and memory deficits, slowed
et al., 2003). processing speed may also be associated with poor treatment
Executive dysfunction in LLD and more particularly LoD is response (Hickie et al., 1995; Sheline et al., 2010; Simpson et al.,
common and includes deficits in planning, organization, sequenc- 1997; Story et al., 2008), thus suggesting the utility of considering
ing, response inhibition, problem solving and set-shifting (Herr- cognitive function when determining treatment responsiveness.
mann et al., 2007; Naismith et al., 2003; Pisljar et al., 2008; Changes in processing speed in older patients with depression has
Salloway et al., 1996). Of concern, the presence of executive deficits been linked to underlying neurobiological changes including
is associated with poor clinical outcomes and prognosis. For smaller caudate nucleus volumes (Naismith et al., 2002), and
instance, executive dysfunction appears to be related to symptom reduced regional cerebral blood flow in the striatum (Hickie et al.,
108 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

1999, 2007a). Some studies have also examined the role of genetic
risk factors and in this regard there is some data linking the MTHFR Box 2. Neuropsychological features of late-life depression.
gene mutation (a CVD risk factor) to slow processing speed (Hickie
 Fronto-subcortical profile
et al., 2001a; Naismith et al., 2003), though there is also some
 Executive functions, processing speed, learning and memo-
contradictory evidence in this field (Hong et al., 2009).
ry may be affected
Memory deficits as measured by neuropsychological testing  Deficits may persist despite symptom resolution
may not be as severe as deficits within the domains of processing  Deficits may reflect underlying cerebrovascular disease,
speed and executive functioning, but nonetheless do relate to the deleterious illness-specific effects and/or early neurodegen-
patient’s overall complaints of cognitive dysfunction. They are also erative changes
predictive of functional disability (Naismith et al., 2007). In  Late-onset phenotype may have more pronounced cognitive
general, the literature pertaining to memory deficits in LLD is more impairment and greater progression to dementia syn-
heterogeneous. In some reports, memory decline is certainly dromes
associated with later ages of depression onset (Kohler et al.,
2010a). However, this relationship has not been reported in other
clinical (e.g. (Naismith et al., 2003; Rapp et al., 2005), or meta- Furthermore, a more recent study found that duloxetine was
analytic (Herrmann et al., 2007) studies. The lack of clarity associated with significant improvements in verbal learning and
regarding the nature and extent of memory change in LLD may be memory compared to placebo in older depressed adults, although
due to a number of factors. Firstly, there are likely diverse it is not clear whether this effect is separate from the effects on
aetiologies underpinning memory difficulties including the effects mood, particularly as the duloxetine group had higher depression
of cerebrovascular disease, early neurodegenerative changes and scores at baseline (Raskin et al., 2007). Interestingly, some data
the chronic effects of depressive illness on the hippocampus (see shows that even prior to commencing treatment, serum antic-
Table 1). The mechanism by which depression itself may cause holinergicity plays a role in memory decrements in LLD (Nebes
memory decline remains to be determined but there is evidence et al., 1997). Thus, it may be worth considering the potential
suggesting that the down-regulation of the HPA axis may be impact of anticholinergic medications on cognition, an effect that
relevant, as well as the associated neurotoxic effects of gluco- may be more pronounced with the tricyclic antidepressants.
corticoids, and reduced expression of brain-derived neurotrophic While data suggests that the effects of electroconvulsive
factor (BDNF) (Duman et al., 2006). There is emerging evidence therapy (ECT) on episodic memory resolve over time, there is,
linking poorer memory performance and hippocampal functioning however, adequate data supporting the notion that retrograde
in depression (Hickie et al., 2005b). Poorer memory functioning in amnesia may be persistent following ECT. In this regard, future
LLD certainly appears to be associated with reduced hippocampal studies exploring the role of alternative anaesthetics (e.g.
volume. Reduced hippocampal volume was also associated with ketamine) are required (for review, see Gregory-Roberts et al.,
persistence of memory problems at 6-month follow-up intervals 2010). Of critical significance, memory impairments may be a
(O’Brien et al., 2004). Indeed, over 2-year follow-up intervals, prognostic indicator and are associated with treatment respon-
hippocampal volume reduction is more prominent in older people siveness (Culang et al., 2009; Sheline et al., 2010; Story et al., 2008)
with depressive disorders compared to controls, and is predictive and disability (Naismith et al., 2007). Efforts directed at early
of cognitive decline on the MMSE, a measure of gross cognitive detection and treatment of memory deficits are warranted, and
functioning (Steffens et al., 2011). preliminary data suggests that cognitive training strategies may
When evaluating memory functioning in LLD, it is important to prove useful (Naismith et al., 2011a, 2009c) (Box 2).
consider which aspects of memory processing are impaired. That
is, insights regarding underlying pathophysiological changes can 4.1. Predictors of cognitive decline
be gleaned by determining whether difficulties are apparent in
memory ‘encoding’, ‘storage’ or ‘retrieval’. Commonly, learning As noted above, cognitive deficits in LLD and particularly LoD
problems are merely referred to as ‘memory’ problems, wherein often persist despite antidepressant treatment (Devanand et al.,
they may actually reflect ‘encoding’ problems. The differentiation 2003; Nebes et al., 2003) or adequate symptom resolution
of these various aspects of memory is relevant since they likely (Dahabra et al., 1998; Portella et al., 2003; Trichard et al., 1995).
reflect dysfunction within distinct neurobiological circuitry, with Although processing speed and executive function deficits are
the former reflecting dysfunction within prefrontal or frontotem- likely to be specifically implicated in the overall profile (Murphy
poral circuits and the latter being reflective of mesial temporal lobe and Alexopoulos, 2004; Nebes et al., 2003), there is some
dysfunction including hippocampal compromise (Shallice et al., suggestion that selective improvement in executive function
1994; Tulving and Markowitsch, 1997). In addition to aetiological relative to memory may occur (Butters et al., 2000). This failure
and definitional constraints, there are likely other factors mediat- to normalise cognitive performance with treatment is certainly
ing memory performance in LLD. For instance, recent data suggests conceivable if the underlying cause of the cognitive deficits is due
that sleep–wake disturbance may be relevant to both encoding and to structural damage in white matter tracts, which is unlikely to be
memory storage deficits in older people with depression, even reversible.
after controlling for depressive symptom severity (Naismith et al., While a dearth of studies exist in this area, clinical predictors of
2009b, 2011b). Additionally, some data suggest that inpatients longitudinal decline in LoD include increasing severity of depres-
tend to demonstrate greater memory decrements relative to sion at baseline, inflammatory markers such as higher erythrocyte
outpatients (Basso and Bornstein, 1999), and that concomitant sedimentation, and VRFs such as lowered HDL cholesterol and
anxiety may play a key role in memory encoding (Kizilbash et al., increased body mass index (Baldwin et al., 2006). Over a 4-year
2002). With regard to pharmacological treatment, there is period, one study showed that later age of depression onset was
evidence to suggest that both single-acting and dual-acting associated with memory and executive decline (Kohler et al.,
antidepressants may have beneficial effects on memory function 2010a), although not all studies suggest that age of onset alone is
in LLD. A small study investigating the effects of sertraline therapy useful (Brodaty et al., 2001; Dahabra et al., 1998; Krishnan et al.,
found improvements in both short- and long-term storage 1995). In a study examining the role of anxiety over a 4-year
(Doraiswamy et al., 2003), however the lack of a control group period, comorbid Generalised Anxiety Disorder or Panic Disorder
in this study means that practice effects cannot be discounted. was associated with specific decline in memory performance in
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 109

those with LLD. Although participants in this study were not population and therefore constitutes a significant risk factor
stratified by age at onset of depression, the impact of age at onset of (Alexopoulos and Chester, 1992). The risk to dementia conversion
anxiety on cognitive functioning in LLD was investigated and an may be particularly high when depression is accompanied by
association between later age at onset of anxiety and worse cognitive impairment and progressive WMLs (Potter and Steffens,
cognitive function in LLD was found (DeLuca et al., 2005). Other 2007). This data collectively suggests that LLD with cognitive
clinical predictors include apathy, which appears to be a good impairment may represent an early manifestation or prodromal
indicator of dementia risk (Bartolini et al., 2005). Neurobiological feature of cognitive decline and/or dementia (see Panza et al.,
correlates may be particularly relevant and one study showed that 2010).
smaller left hippocampal volumes were related to incident Data from a number of community studies suggests that
dementia over a follow-up period of up to five years (Steffens depression itself presents an independent risk factor for cognitive
et al., 2002b). As expected, WML burden is another predictor of decline (Jorm et al., 1991; Ownby et al., 2006). Indeed, recent
conversion to dementia (Potter and Steffens, 2007), and suggests longitudinal data, as well as a meta-analysis, demonstrated a
the need to target possible underlying CVD early (Naismith et al., population attributable risk fraction of depression for dementia of
2009a). In terms of cognitive predictors, some longitudinal studies around 10%; with estimates that a reduction of 10% in depression
have shown that baseline memory impairment is relevant prevalence could lead to 326 000 less cases of dementia worldwide
(Baldwin et al., 2006; Kohler et al., 2010a). This observation is (Barnes and Yaffe, 2011). Such data highlights the importance of
supported by recent studies demonstrating that decreases in eliminating the impact of depression when considering dementia
hippocampal volume over a 2-year period are predictive of general reduction strategies (Ritchie et al., 2010). Identification of
cognitive decline (Steffens et al., 2011). Additionally, according to a therapeutic targets for improving cognitive functioning in this
recent five year follow-up study of depressed older adults, both ‘at risk’ group is clearly warranted for both symptomatic
memory deficits and executive dysfunction best predict conver- improvement, as well as for potential disease modification
sion to dementia (Potter et al., 2012). (Naismith et al., 2009a).
Personality factors that may increase the risk for dementia have In terms of mechanisms, it is not yet clear how depression
not been well evaluated. Interesting links between personality contributes to dementia, and this may be partly due to the
traits, such as neuroticism and reduced openness, and cognitive heterogeneity in longitudinal illness trajectories (i.e. vascular, AD,
decline have been suggested (Kuzma et al., 2011). Such factors may Lewy body dementia). The notion of a shared aetiological
be associated with dysregulation of cortisol (Tyrka et al., 2006), foundation between LLD and AD has received some attention,
that in turn, has been implicated in dementia. and various studies have investigated the role of amyloid and tau
protein deposition in LLD, and progression to dementia. For
4.2. The relationship between late-life depression, mild cognitive instance, a small post-mortem study found a predominance of AD
impairment and dementia pathology in individuals with LoD and varying levels of cognitive
impairment (Sweet et al., 2004). More recent work utilising
Up to 60% of patients with LLD meet criteria for mild cognitive position emission tomography (PET) has further highlighted the
impairment (MCI) (Adler et al., 2004; Lee et al., 2007), a syndrome potential role of amyloid/tau deposition in the interface between
that is considered to be a prodromal or ‘at risk’ state for dementia. depression and dementia. Utilising different imaging techniques
Indeed, depression itself has been recognised internationally to be (see Section 7.1), Butters et al. (2008) found that half a group of
a prodromal feature for cognitive decline (Steffens et al., 2007) individuals with treated LLD had evidence of cerebral amyloid
with more severe symptoms representing a greater risk for MCI deposition in a cortical pattern characteristic of early AD, and
(Lee et al., 2007); moreover epidemiological studies have Kumar et al. (2011) found that protein (amyloid and tau) binding
supported the ‘prodromal’ nature of depression, demonstrating was significantly higher in individuals with LLD (who did not meet
an association between dementia and depressive episodes criteria for MCI) than in controls, although in a pattern that differed
occurring for the first time in close proximity to dementia onset from that typically observed in patients with AD. Amyloid and tau
(Berger et al., 1999; Chen et al., 1999; Rasmuson et al., 2001). In are of primary importance for neuronal injury and death in AD;
those with MCI, rates of depression vary, with recent reviews animal studies have demonstrated increased amyloid deposition
suggesting a median depression prevalence of 44.3% in hospital- after administration of glucocorticoids (at levels usually associated
based studies and 15.7% in population-based studies (see Panza with stress response), thus suggesting a possible mechanism for
et al., 2010 for a review). the relationship between LLD and dementia (see review by Caraci
Studies have shown that the combination of depression and et al., 2010). It is, however, worth noting that AD pathology is not
MCI is associated with a two-fold risk of developing Alzheimer’s the only pathophysiological mechanism by which cognitive
disease (AD) with an earlier age of onset (Modrego and Ferrandez, decline may ensue. For instance, as noted above, the longitudinal
2004). Other studies suggest that those with LLD tend to link to vascular forms of dementia has been documented (Baldwin
predominantly progress to vascular forms of dementia (Baldwin et al., 2006; Hickie et al., 1997b) and, in turn, cerebrovascular
et al., 2006; Hickie et al., 1997b). Two recent large community- disease has been established as a risk factor for AD. Conversely, a
based studies have demonstrated that increasing levels of recent community study investigating the association between
depressive symptoms are associated with increasing risk of LLD and neuropathological changes found that depression is
dementia (Saczynski et al., 2010) and that recurrent depression associated with subcortical and hippocampal neuronal loss but not
is particularly pernicious (Dotson et al., 2010). Even in those with cerebrovascular or Alzheimer’s pathology (Tsopelas et al.,
patients who may initially demonstrate improvement in mood and 2011). A novel finding of this study was that depression was
cognitive symptoms, follow-up data over mean intervals of eight associated with Lewy bodies in the substantia nigra and locus
years has shown that while patients may initially demonstrate a ceruleus. Therefore, in some patients, synucleinopathy may even
recurrence of depressive episodes, cognitive impairment is contribute to cognitive decline. Overall, the lack of clear
increasingly apparent and eventually, around 79% of patients will neuropathological substrates in LLD again highlights the hetero-
present with a dementia syndrome (Kral and Emery, 1989). geneity of the disease, and the potential for LLD to progress to a
Further, it has been estimated that geriatric depression with multitude of illness trajectories. In this regard, it is also possible
‘reversible dementia’ has a yearly rate of ‘irreversible dementia’ that ongoing immunological and inflammatory changes contribute
two-and-a-half to six times higher than that of the general geriatric to cognitive decline as well as disease-specific mechanisms
110 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

relating to glucocorticoids and neurotrophins (see Table 1). For Taylor, 2006) and will be outlined here with specific reference to
instance, there is evidence to suggest that reduction of BDNF is age of onset, fronto-subcortical circuitry and clinical correlates (see
implicated in the pathophysiology of MD (Duman et al., 2006; Fig. 1a and b).
Nestler et al., 2002) and these reductions appear to relate to
depressive symptom severity. BDNF is a neurotrophin that 5.1. Volumetric findings
promotes neuronal differentiation, growth and survival. It plays
a role in use-dependent plasticity mechanisms such as long-term 5.1.1. Whole brain
potentiation and is critical in cognitive processes such as learning, Although in mid-life samples an association between smaller
memory and executive functions (Duman et al., 2006). Recent whole brain volumes and depression has been demonstrated
work suggests that particular genetic polymorphisms of BDNF are (Hickie et al., 2005b; Naismith et al., 2002), MRI studies of whole
associated with larger hippocampal volumes in depressed elderly brain volumes in LLD have generally not demonstrated significant
compared with non-depressed controls who have the same differences between depressed elders and healthy controls
genotype, and may have a neuroprotective role (Kanellopoulos (Andreescu et al., 2008; Ballmaier et al., 2004b; Krishnan et al.,
et al., 2011). Thus, there appears to be a complex interplay between 1993; Kumar et al., 1998). Of those that have examined LoD
BDNF, depression, hippocampal functioning and cognition. An specifically, Pantel et al. (1997) found that this sub-group had
enriched understanding of how these contributing aetiological significantly smaller whole brain volumes compared to healthy
factors relate to symptoms and brain changes will ultimately help controls. However, more recent studies have failed to confirm this
to develop treatments targeted at neuroprotection and reduction finding (Ballmaier et al., 2004a). Differences in ventricular size
of cognitive decline. between individuals with LLD and controls have been more
In considering the link between depression and dementia, it is consistently found (Alexopoulos et al., 1992; Shima et al., 1984),
also important to recognise that while LLD may be a prodromal or and larger ventricular sizes appear to correlate with the age of
concomitant feature of many neurodegenerative diseases includ- depression onset. Despite the apparent lack of alterations in whole
ing AD, Parkinson’s disease and vascular dementia, the longitudi- brain volumes, there is a wealth of data relating to specific regional
nal trajectory of those presenting with depressive symptoms in volumetric differences associated with LLD. Sophisticated imaging
later-life is not yet well delineated, particularly in non-clinical techniques, including three-dimensional anatomical surface
samples. Indeed, recent large studies of community dwelling older modelling techniques, have demonstrated more clearly some of
adults without dementia at baseline have found that, although these changes, as well as their relationship with age of depression
depression at baseline is associated with an increased risk for onset.
dementia over extended follow-up periods, many of those who
were depressed did not go on to suffer dementia. In the 5.1.2. Frontal lobe
Framingham Heart Study (Saczynski et al., 2010) depressed Numerous studies have demonstrated decreases in frontal lobe
participants had a 50% increased risk for dementia compared to volume in those with LLD (Kumar et al., 2000a, 1998; Lavretsky
non-depressed participants but this represented only 21.6% of the et al., 2005). This has been shown specifically in LoD (Almeida et al.,
depressed group; in other words 79.4% of depressed elders did not 2003), which has also been associated with attenuation of the
develop dementia. Similarly, the Maastricht Aging Study found ‘normal’ frontal volumetric asymmetry (Kumar et al., 2000b).
that out of 180 depressed (defined as falling within the upper However, there are some conflicting results, with Kumar et al.
quartile on the revised 90-item Symptom Checklist) participants in (1999) failing to find any linear relationship between age of
the cohort of 771 non-demented community dwelling elders, only depression onset and total or focal brain volumes. Frontal lobe
14 (8%) developed dementia over a 9-year period (Kohler et al., changes in this group may also be mediated by gender. Lavretsky
2009). Whilst this represented an increased risk for dementia et al. (2004) reported that in depressed elders, males have smaller
compared to non-depressed participants, it also highlights that the frontal lobe volumes compared with females. Taki et al. (2005) also
development of dementia in the context of LLD is by no means found gender differences, with depressed males showing signifi-
automatic. Therefore, primary and secondary strategies aimed at cant decreases in the medial frontal lobe, as well as in the right
identifying ‘at risk’ individuals and targeting modifiable risk precentral gyrus compared with male controls. These results were
factors should be implemented early in this group (Naismith et al., not present in females.
2009a). A number of studies (Ballmaier et al., 2004c; Dotson et al., 2009;
Lai et al., 2000; Lavretsky et al., 2007) have specifically examined
5. Structural neuroimaging studies subregions of the prefrontal cortex (PFC) in LLD. In accordance with
the neural circuitry implicated in depressive disorders, reductions
Major advances in the neurobiology of depression were made in in the OFC, ACC and gyrus rectus have been demonstrated in
the 1990s with the advent and accessibility of magnetic resonance depressed elders compared with controls (Ballmaier et al., 2004c;
imaging (MRI). A large body of structural neuroimaging research Elderkin-Thompson et al., 2009; Lavretsky et al., 2007; Lee et al.,
has ensued and initially included quantitative measurement of key 2003). While not all studies suggest that the LoD phenotype is
structures including volumetric analysis of prefrontal, striatal and associated with specific PFC subregion volumetric reductions
limbic regions (see reviews by Soares and Mann, 1997; Steffens (Ballmaier et al., 2004a), some have shown that grey matter
and Krishnan, 1998), though notably such methods do not inform reductions in frontal superior, orbital, middle and medial superior
us as to the aetiology or nature of the volume loss (e.g. glial, regions correlate with later age of depression onset (Andreescu
neuronal, other). Concurrently, the greater resolution of white et al., 2008). Notably, even sub-threshold depressive symptoms
matter offered by MRI, as opposed to computed tomography, led to appear to relate to volumetric reductions. For instance, depressive
a preponderance of qualitative studies examining the location and symptoms have been found to be associated with reductions in the
severity of WMLs. More recently, further sophistication of cingulate gyrus and OFC, although age also played a role in
neuroimaging acquisition and analysis techniques has enabled mediating this relationship (Dotson et al., 2009).
the more detailed examination of white matter tracts using
diffusion tensor imaging. The specific structural neuroimaging 5.1.2.1. Associations with clinical and cognitive features. There is
findings in LLD have been reviewed by a number of groups some data to support a link between PFC volume reductions and
(Herrmann et al., 2008; Schweitzer et al., 2001; Vaishnavi and clinical symptoms, cognition and functional capacity; although a
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 111

clear regionally specific relationship has not yet emerged. For promote neurotrophins such as BDNF, which in turn, may promote
instance, volumetric reductions of the left frontotemporal region hippocampal neurogenesis, or at least provide some neuroprotec-
have been correlated with psychotic symptoms as well as cognitive tion from the deleterious effects of glucocorticoids (Chen et al.,
impairments in the domains of psychomotor speed and executive 2001; Malberg et al., 2000; Perera et al., 2007; Wang et al., 2008a).
function in those patients with LoD (Simpson et al., 1999). By While there are some negative studies, human studies in
contrast, apathy in LLD has been associated with ACC volume younger patients with depression as well as LLD largely support
reductions (Lavretsky et al., 2007). Volumetric reductions in the the notion that depression is associated with hippocampal volume
medial orbitofrontal gyri were correlated with functional disability reduction. For instance, Sheline et al. (1996) found that duration of
in LLD (Taylor et al., 2003b). Another study showed that smaller depression predicts hippocampal volume loss in women. Consis-
OFC, gyrus rectus and ACC volumes were related to poorer tent with this finding, studies in adults have found that although
executive function (Elderkin-Thompson et al., 2009) in healthy hippocampal dysfunction (as measured by memory performance)
elderly, a relationship that was attenuated in depressed elders. is present in both those with first episode depression as well as
This may indicate that depressed elders are less likely to benefit those with multiple previous episodes of depression, hippocampal
from larger volumes of these key regions in comparison to control volume reductions appear to be apparent only in those with past
subjects. Thus, rather than focussing on volumetric change, it may depressive episodes (MacQueen et al., 2003). Furthermore, in older
be necessary to consider other underlying pathophysiological adults, bilateral hippocampal-entorhinal volume reductions have
contributors to function (Elderkin-Thompson et al., 2009). been found to be associated with greater years of cumulative
depressive illness (Bell-McGinty et al., 2002) and smaller
5.1.2.2. Medication effects. There is some suggestion that antide- hippocampal volumes appear to be linked with treatment
pressant medication may mediate frontal volumetric changes. responsiveness (Hsieh et al., 2002) as well as gross cognitive
Specifically, antidepressant exposure may protect against frontal decline over two year follow-up periods (Steffens et al., 2009).
grey matter loss in LLD. For instance, Lavretsky et al. (2005) found Interestingly the relationship between hippocampal volumes and
that depressed elders had smaller total and grey matter volumes of depression may preferentially relate to the duration of untreated
the OFC but that those with prior antidepressant exposure had illness, as no known relationship has been demonstrated between
larger total OFC volume compared to the drug naı̈ve group. hippocampal volume loss and duration of depression whilst on
Additionally, consistent with data linking executive dysfunction antidepressant therapy, nor with duration of exposure to
with prognosis (e.g. Sheline et al., 2010), volumetric changes in the antidepressant therapy (Sheline et al., 2003).
PFC may be associated with treatment responsiveness. For In particular, when examining LLD, it is important to consider
instance, reductions in dorsal and rostral ACC volumes have been the contributing effects of early neurodegenerative disease, as well
associated with failure of LLD to respond to escitalopram as vascular disease when interpreting hippocampal change. In this
treatment (Gunning et al., 2009). A number of possibilities may regard, studies have found volumetric reductions in both LoD and
account for these findings: smaller regional PFC volumes may EoD (Hickie et al., 2005b; Lloyd et al., 2004; Steffens et al., 2000).
predispose to failure to respond to antidepressant therapy. Some of these have also linked smaller hippocampal size in LoD
Alternatively neurotrophic effects of antidepressant medication with poorer memory performance (Hickie et al., 2005b) or smaller
may lead to the increases seen in grey matter volume. Further- left hippocampal volume and later development of dementia in
more, since a negative association between OFC volume and depressed non-demented elders (Steffens et al., 2002b), as well as
subcortical grey matter lesion severity (basal ganglia) has been early-onset subtypes (Bell-McGinty et al., 2002; Sheline et al.,
demonstrated in LLD (Lee et al., 2003), cerebrovascular disease 1999). Furthermore, a recent large cohort study demonstrated that
may play a key role in mediating volumetric changes. whilst having ever been depressed was associated with smaller
hippocampal volumes, those with EoD had smaller hippocampal
5.1.3. Limbic regions volumes compared with controls whilst those with LoD had
The limbic system comprises a set of brain structures that smaller entorhinal cortex volumes (Gerritsen et al., 2011). Again,
collectively play a role in emotion, behaviour, memory and this data suggests that volumetric changes in LLD can reflect
olfaction and which is linked to other key neurohormonal systems multiple pathophysiological processes. In this regard, there are a
including the circadian system (see review by Gerstner and Yin, range of immunological, inflammatory, vascular and neurodegen-
2010). Of these structures, the hippocampus has been most erative factors of potential relevance.
extensively investigated in MD as well as LLD more specifically. With respect to the ‘neurotoxic’ effects of depressive illness, a
While the amygdala clearly plays a vital role in the EoD phenotype, failure of the termination of glucocorticoid secretion at the end of a
and particularly in the modulation of anxiety, fear and social stressor may be related to degeneration in key brain regions that
behaviours, it has been less well studied in LLD. regulate the HPA axis, including the hippocampus; this degenera-
tion in turn may relate to cumulative exposure to glucocorticoids
5.1.3.1. The hippocampus. The hippocampus is integrally involved throughout the lifespan thus setting up a feed-forward cascade
in mood regulation due to its connections to key frontal and (Sapolsky et al., 1986). As recently reviewed by MacQueen and
subcortical regions including the amygdala, hypothalamus, basal Frodl (2011), in animal models, stress is associated with a decrease
ganglia and PFC. There is evidence to suggest that it plays a direct in the size, length and number of hippocampal dendrites, and
role in the serotonergic HPA functioning (see review by MacQueen disrupts hippocampal neurogenesis, though one must be cautious
and Frodl, 2011). It is an especially plastic and vulnerable region, in extrapolation of these findings to humans since species-
sensitive to the effects of hypoxia, cerebral insult and excess dependent effects have also been observed. As neurogenesis
glucocorticoids, as well as excitatory neurotransmitters, due to the appears to be integral to neuroplasticity, altered neurogenesis may
large numbers of receptors (McEwen and Olie, 2005). The be critically involved in reduced antidepressant responsiveness
hippocampus has been the subject of renewed interest in and may directly impact on pathogenesis such that vulnerability to
neuropsychiatry and regenerative neuroscience in recent years, depression is enhanced and perpetuated. Further support for the
since animal studies have now suggested that this primitive role of stress on hippocampal integrity also stems from data linking
structure is capable of neurogenesis. This research is of particular stress with poor memory, as well as the observation that extended
interest in LLD as a wealth of both animal and human studies have or high dose treatment with glucocorticoids has a negative impact
concurrently demonstrated that antidepressant medications on hippocampus-dependent memory consolidation (for review see
112 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

MacQueen and Frodl, 2011; Sapolsky et al., 2000). Studies also been found to be smaller in LoD (Ballmaier et al., 2004c),
involving human subjects have demonstrated altered glucocorti- disruption of the efferent and afferent connections between the
coid regulation in patients with both depression (Keller et al., hippocampus and the thalamus, basal ganglia and PFC by
2006) and dementia (Rasmuson et al., 2001), however evidence for ischaemic disease could contribute to depression vulnerability.
cortisol as a mediating mechanism between depression and
hippocampal atrophy is inconsistent. One longitudinal study 5.1.3.3. Relationship with clinical and cognitive features. As noted
examining the relationship between hippocampal volume, cogni- above, reductions in the hippocampus in MD have been directly
tive impairment and hypercortisolaemia, found no association associated with memory performance (Hickie et al., 2005b), a
between hippocampal volume reduction in depressed elders and finding which is not surprising given the well-documented role of
increased cortisol levels (O’Brien et al., 2004). This finding was this structure in memory functioning (Squire, 1992). Hippocampal
replicated in a recent study that found that differences in volumetric reductions have also been associated with failure to
hippocampal and entorhinal cortex volumes in EoD and LoD were respond to antidepressant treatment (Hsieh et al., 2002), and there
not explained by HPA axis activity (Gerritsen et al., 2011). Possibly is a suggestion that the hippocampus may modulate antidepres-
these results may relate to measurement factors, in that cortisol sant effects (Mongeau et al., 1997). The findings of Zhao et al.
levels were measured from salivary samples over a three day (2008) may be seen in this light. An alternative explanation may be
period, and thus may not accurately reflect total exposure to that antidepressants exert neurotrophic effects on the hippocam-
glucocorticoids over a lifetime. Also, other potentially relevant pus, accounting for the lack of shape or volume differences in
steroids such as dehydroepiandrosterone (Magri et al., 2006) were remitted depressed elders compared to controls. Further research
not measured. However these findings suggest that there may be in this area is warranted given that amelioration of hippocampal
other deleterious processes at work. The link between stress and change may ultimately reduce cognitive decline associated with
glutamatergic excess must be considered, particularly given the depression, which in turn would have major impacts on disability,
well-established role of this excitatory neurotransmitter in daily functioning, quality of life, and may possibly even slow rates
learning and memory formation (Pittenger and Duman, 2008). of progression to dementia (Naismith et al., 2009a).
In addition, inflammatory changes associated with depression (see
Section 10) as well as disturbances in neurotrophic factors such as 5.1.3.4. The amygdala. Studies investigating amygdala structure
BDNF (see Section 9.1) are likely to be important. den Heijer et al. in younger samples have yielded variable findings, with both
(2011) found no evidence that smaller hippocampal volumes reductions (Caetano et al., 2004; Sheline et al., 1998; von Gunten
preceded the development of LLD but that depression may lead to a et al., 2000) and increases (Frodl et al., 2003; Lange and Irle, 2004)
faster rate of hippocampal volume decline. The apolipoprotein in volumes of this structure evident. This variation may be related
epsilon 4 allele may also be a relevant contributor to the increased to the effects of antidepressant medication. Indeed, this notion is
likelihood of dementia in older people with depressive symptoms supported by a recent meta-analysis of 13 studies that demon-
(Kim et al., 2002), though further prospective studies are required strated that the proportion of medicated depressed participants in
to support this association. Overall, this data suggests that a study determined the difference in amygdala volume between
hippocampal change in LLD likely has multiple pathophysiological depressed and non-depressed subjects, and found that unmedi-
underpinnings, some of which may directly relate to the duration cated depression is associated with decreased amygdala volumes
of treated or untreated depressive illness, and some of which likely (Hamilton et al., 2008). There has been a paucity of research
reflect age-related or neurodegenerative brain changes (see Table specifically examining amygdala volumes in older depressed
1). samples. This relative negligence is surprising given the critical
role of this structure for emotion regulation, particularly
5.1.3.2. Hippocampal morphology. More recent work utilising processing emotional, fear-related and social information. Addi-
computational image analysis methods have evaluated hippocam- tionally the amygdala is important in the modulation of
pal shape differences. Shape and volumetric differences in the left monoamine and corticosteroid release in response to aversive
hippocampus were found in a group of non-remitted depressed or novel stimuli (Savitz and Drevets, 2009). Of those that have
elders when compared with healthy controls and remitted examined this structure in older samples, some evidence for a
depressed elders (Zhao et al., 2008). These changes roughly reduction in volume is emerging. For example, an investigation of
corresponded with regions CA1 and CA2. The remitted group and patterns of brain atrophy in 14 patients with LoD, showed reduced
controls had no such changes. Consistent with these findings, volumes of the right amygdala in comparison with control
Ballmaier et al. (2008) demonstrated significant regional surface participants (Egger et al., 2008). Hickie et al. (2007b) also noted
contractions bilaterally in the hippocampal head, corresponding to smaller amygdala volumes in 45 older patients with moderate to
the CA1 and CA2 subfields and subiculum in LLD compared with severe depression in comparison to controls. A further study of
controls. Moreover significant reductions in the left hippocampus, morphology has demonstrated significant shape variation in the
concentrated in the anterior subiculum and lateral posterior CA1 left and right amygdalae in depressed elders, most prominently in
subfield, were found in older people with LoD compared with EoD. the anterior portion, a region typically associated with the
In the LoD group, memory performance was significantly related to basolateral nuclei (Tamburo et al., 2009). These core nuclei are
hippocampal deficit patterns. These differential hippocampal highly connected to the hippocampus and prefrontal cortex, are
deficit patterns in LoD and their relationship with memory densely populated with glucocorticoid receptors (Johnson et al.,
performance are interesting in the light of similar patterns of 2005), and have been shown to be glutamatergic. Previously,
CA1 and subiculum involvement found in patients with MCI that Sheline et al. (1998) reported that MD in an adult cohort was
later developed Alzheimer’s disease (Apostolova et al., 2006; Wang associated with volumetric decrease in the basolateral nuclei.
et al., 2003), and add weight to the hypothesis that LoD is part of a Thus, it has been postulated that stress induced glucocorticoid
neurodegenerative process. On the other hand, the CA1 and CA2 toxicity may play a selective role in amygdala dysfunction in LLD
subregions have been suggested to be particularly vulnerable to (Hamilton et al., 2008). The neuropathological finding of reduced
ischaemic damage (see review by Nikonenko et al., 2009), and thus, numbers of oligodendrocytes, which play a role in extracellular
such changes may reflect the ischaemic small vessel disease that is glutamate homeostasis (Damelio et al., 1990), in the amygdala of
seen commonly in LoD. Furthermore since the anterior hippocam- depressed individuals (Hamidi et al., 2004) provides some support
pus has rich connections with the PFC, subregions of which have for this hypothesis. Such reductions may lead to toxic levels of
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 113

extracellular glutamate, thus contributing to neural degeneration. density or synaptic density. Neuropathological studies have
Interestingly, animal studies have demonstrated that inducing demonstrated declines in the density and the size of neurons in
electroconvulsive seizures leads to the generation of oligoden- the PFC as well as reductions in glial cell density in limbic regions
drocytes in the rat amygdala (Wennstrom et al., 2004). Taken (see Section 7). Cerebrovascular disease, as exemplified by WMLs
together with the findings that medicated depression is associat- (see Section 5.2) in fronto-subcortical regions, is likely to disrupt
ed with increased amygdala volumes (Hamilton et al., 2008), this regional blood flow and metabolism as well as disturb local
data provides some suggestion that the amygdala is capable of synaptic transmission. Thus, via disruption to their connections,
neuroplasticity. WMLs may be implicated in the atrophy of cortical and subcortical
regions. Finally early cerebral development may be altered via
5.1.4. The striatum genetic polymorphisms, leading to volumetric reductions in key
Some researchers have shown reductions in the size of the brain regions which then place an individual at risk of depression
striatum in LLD (for reviews see Lorenzetti et al., 2009; Steffens and and cognitive syndromes emerging later in life in the context of
Krishnan, 1998). Significant bilateral volume reductions of the ongoing medical and psychosocial risk factors (see Section 8).
caudate have been demonstrated in elderly depressed subjects; Further studies regarding gene expression are now required in
these reductions are generally localized to the head of the caudate order to determine the mechanisms by which relevant vulnerabil-
nucleus, particularly on the left (Butters et al., 2009; Figiel et al., ity genes are expressed.
1991a; Krishnan and Figiel, 1989; Krishnan et al., 1992; Mendez
et al., 1989; Starkstein et al., 1987). Moreover, a negative 5.2. White matter changes
correlation between age and caudate nucleus volumes in
depressed adults (Krishnan et al., 1992), as well as age at onset 5.2.1. Macrostructural findings
of depression and caudate volumes (Greenwald et al., 1997) has Research in ageing samples indicates that white matter
been demonstrated. volume generally declines with age (Gunning-Dixon et al.,
It has been hypothesised that the functional significance of the 2009), and it appears that the frontal and temporal regions are
relationship between caudate volume reductions and depression is most vulnerable. As aforementioned, WMLs are areas of hyper-
related to degeneration in the dopaminergic connections between intensity appearing on T2-weighted and fluid attenuation
the caudate and cortical and limbic areas important for mood inversion recovery (FLAIR) MRI images; they may occur in both
regulation (Krishnan et al., 1992). Interestingly, increasing volume the deep white and subcortical grey matter as well as in
of anterior WMLs have been found to be associated with smaller periventricular regions. They are typically classified by pattern
total and right caudate volume in depressed elders (Hannestad (e.g. punctate or confluent) and region (periventricular, deep
et al., 2006), suggesting that vascular disease may play a role in white matter, subcortical grey regions) and can be rated visually
caudate atrophy via disruption of frontostriatal connections. No using standardised rating scales (Fazekas et al., 1987; Scheltens
known studies to-date have examined specific subregions of the et al., 1993; Wahlund et al., 2001). More recent research has
caudate nucleus. This may prove to be more pertinent to the utilised volumetric measurement as opposed to visual rating
refinement of comprehensive pathophysiological models of LLD scales as it is able to provide more information on WML location
given the differential roles of specific subregions of the caudate and size (Anbeek et al., 2004), and there is evidence to suggest that
nucleus in aspects of cognitive, emotional and behavioural visual rating scales are less sensitive in differentiating clinical
functioning (Balleine and O’Doherty, 2010). In particular, the groups (van Straaten et al., 2006). Histopathological examination
ventral striatum may play a role in the motivational control of reveals a significant proportion of WMLs represent areas of
action, whilst the dorsal striatum may play a role in the control of infarction, axonal loss, gliosis and/or ischaemic demyelination,
decision-making mediated by reward. Thus, it appears that the and an increased proportion of WMLs in the frontal lobes appear
striatum plays a larger role in executive functions than previously ischaemic in depressed compared to control subjects (Thomas
acknowledged (Balleine and O’Doherty, 2010). One recent study et al., 2002). They are strongly associated with age and VRFs
used three-dimensional surface mapping techniques in a group of (Awad et al., 1986; Liao et al., 1997; Longstreth et al., 2001). The
patients with LLD and control subjects and demonstrated that presence of perivascular spaces indicative of microangiopathy are
volumetric reductions were most pronounced in anterior regions, also common in LLD and recent studies using pressure myography
which in turn were associated with more severe depressive have shown abnormalities of subcutaneous small artery structure
symptoms, but not with age of onset or years of cumulative illness and function, even when compared to controls with similar
(Butters et al., 2009). Further studies using such sophisticated cardiovascular profiles (Greenstein et al., 2010). In clinical
imaging analysis techniques are now required in order to practice, however, it is important to consider that specific
determine the relevance of topographically and functionally neuroradiological investigation must purposively examine for
distinct regions of the caudate nucleus. other underlying causes of WMLs, including those reflecting
infectious or inflammatory conditions or demyelination.
5.1.4.1. Relationship with clinical and cognitive features. There As previously outlined (Section 3), LLD is characterized by more
appears to be a paucity of research in this area. This may partly frequent and intense WMLs in comparison to control subjects, a
reflect the few studies that have been conducted, publication bias finding that is even more prominent in those with later ages of
or may even include lack of reference to specific subregions of the depression onset (Coffey et al., 1993; de Groot et al., 2000; Figiel
caudate nucleus. As noted above, caudate nucleus volumes have et al., 1991b; Herrmann et al., 2008; Hickie et al., 1995; Krishnan
been correlated with depression severity (Butters et al., 2009) in and McDonald, 1995; Kumar et al., 2000a; Steffens et al., 2002a,
some older depressed samples and others have demonstrated links 1999b; Tupler et al., 2002). In the 1990s, several seminal studies
with psychomotor speed (Naismith et al., 2002); a key determinant were published in this area, which collectively highlighted the link
of treatment response (Hickie et al., 1996; Story et al., 2008). between WMLs, VRFs and age at onset of depression (Alexopoulos
et al., 1997; Hickie et al., 1995, 1997b; Krishnan et al., 1997). As
5.1.5. Significance of volumetric changes in LLD outlined above, there has since been a wealth of data that has
Further studies are required in order to determine what explored the association between WMLs and both LLD and LoD;
volumetric reductions actually reflect. For instance, they may these studies have been reviewed elsewhere (e.g. Herrmann et al.,
simply represent changes in neuropil, alterations in glial cell 2008).
114 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

One outstanding issue with regard to the role of WMLs in the to have an important role in long-term outcome of LLD, with
aetiology of LLD pertains to the importance of lesion location as reports that severity of subcortical gray matter hyperintensities at
opposed to lesion volume. Early reports using visual rating scales baseline is associated with scores on the Montgomery-Asberg
linked increasing severity of WMLs with new onset depression Depression Rating Scale over a follow-up period of 64 months in
(Lesser et al., 1991; Nebes et al., 2002). More recent research older adults with depression (Steffens and McQuoid, 2005).
utilising semi-automated volumetric measurement has found In addition to being associated with later ages of depression
greater WML volumes in LLD compared with healthy controls onset and cerebrovascular disease, WMLs also appear to be
(Taylor et al., 2005a). Interestingly in a study evaluating the associated with cognitive decline. For example, one early study
internal validity of ‘vascular’ depression as a unique diagnostic incorporating follow-up (Hickie et al., 1997b) showed that older
subtype, latent class analysis of two independent clinical samples age at onset of depression and extent of WMLs on MRI played a
found that only deep white matter lesion burden (as compared to significant role in progressive cognitive decline and the develop-
executive dysfunction, late-onset of depression and subcortical ment of probable dementia syndromes, as well as the need for
grey matter hyperintensities) was necessary to determine institutional care (Hickie et al., 1997b). Specific associations have
membership to the ‘vascular’ depression group (Sneed et al., been demonstrated with memory impairments, executive dys-
2008). These studies therefore suggest that lesion volume is a key function and impaired processing speed (Hickie et al., 1995; Kohler
determinant of symptom onset. et al., 2010b; Kramer-Ginsberg et al., 1999; Potter et al., 2007).
On the other hand, there is also data to support the critical role Kramer-Ginsberg et al. (1999) found that depressed elderly with
of lesion location. For instance, frontal lesions (Firbank et al., 2004; moderate to severe WMLs in the deep white matter performed
Sheline et al., 2008; Taylor et al., 2003a), and more specifically significantly worse than both depressed patients without such
lesions in the medial OFC (MacFall et al., 2001), are associated with lesions as well as healthy controls (both with and without
LLD depression. In a sample of 22 patients with LoD and controls, moderate to severe deep white matter hyperintensities) on tests of
Dalby et al. (2010) reported that while no significant differences memory and executive function. Interestingly no significant
were found between groups with regard to the number and associations were observed for hyperintensities in periventricular
volume of WMLs, patients displayed a significantly higher lesion or subcortical grey areas. Sheline et al. (2008) examined the
density in the right superior frontal gyrus (the boundary zone of functional significance of WMLs by correlating the lesion burden in
white matter underlying the medial PFC, ACC and the OFC), and in seven subcortical regions (mentioned previously) with perfor-
the white matter area between the left precentral and post-central mance on a comprehensive battery of neuropsychological tests.
gyrus (an area approximating Brodmanns area 6, comprising the Among the depressed patients, three regions were significantly
premotor cortex and supplementary area). It is interesting to note and negatively correlated with executive function including (1)
from this study, however, that five patients demonstrated no right superior longitudinal fasciculus (2) left superior longitudinal
evidence of WMLs, reaffirming that these features are not a fasciculus and (3) left uncinate fasciculus. However, no significant
necessary condition for all patients who develop depression for the correlations were found within the control group. As these areas
first time in later-life. Sheline et al. (2008) also found that the support cortical regions critical for higher level thinking skills (i.e.
strategic location of WMLs, but not whole brain volumes played a executive functions) and emotional processing, such lesion
significant role in LLD and distinguished patients with LLD from locations may increase susceptibility to depression, expressed
non-depressed elderly, irrespective of the amount of VRF burden. functionally as cognitive deficits (Sheline et al., 2008). More
In this study, automated segmentation methods were used to recently WMLs in both the deep white matter and periventricular
compare the total brain and regional WML burden found on MRI areas have been associated with continuing cognitive impairments
between 83 older depressed adults and 32 age-matched controls. in older adults with depression. Using the modified Fazekas criteria
While whole brain volumes, white matter, gray matter and WMLs for assessing WML, Sheline et al. (2010) found that elderly (>60
did not differ between groups, the prevalence of WMLs in seven years) patients with MD were more likely to perform poorly on
regions of the brain was significantly greater in the depressed neuropsychological tests of executive function, processing speed,
group than in the comparison group. The regions identified were (i) episodic memory, language and working memory if they had
the right superior longitudinal fasciculus; (ii) the right superior greater WML burden. Furthermore, severity of cognitive im-
longitudinal fasciculus; (iii) a second region in the right superior pairment, as well as Fazekas scores were predictors of outcome
longitudinal fasciculus; (iv) left superior longitudinal fasciculus; after 12 weeks of antidepressant (sertraline) therapy. By contrast,
(v) left uncinate fasciculus/frontal operculum; (vi) right extreme other studies have shown that neither cortisol levels nor cerebral
capsule; and (vii) right inferior longitudinal fasciculus. This study atrophy are associated with such impairments (Kohler et al.,
is unique in evaluating the white matter tracts and suggests more 2010b). Taken together these findings emphasize the role of
detailed tractography analysis may ultimately be useful to help cerebrovascular disease in conferring vulnerability as well as
delineate which neural circuits and connecting tracts are critical to perpetuating LLD. Possibly confluence of WMLs confers a worse
the vascular model of LLD. prognosis than the presence of punctate lesions (Sachdev et al.,
2006); furthermore the association between specific WML location
5.2.1.1. Relationship with clinical and cognitive features. While a and cognitive deficits suggests that disruption to fronto-subcorti-
prospective study failed to show that WMLs were predictive of cal connections may be relatively more important for the cognitive
mild levels of depressive symptoms longitudinally (Versluis et al., impairments seen in LLD than lesions in the subcortical grey
2006), the aetiological significance of WMLs in LLD, as well as LoD, matter, which presumably represent structural damage in these
is underscored by reports that they may predate the onset of areas. Such structural damage is likely to alter the function of the
depression (Nebes et al., 2002; Teodorczuk et al., 2007), and may subcortical region affected, possibly producing the particular
predict progress over time in those with depression (Godin et al., clinical features of depression such as anhedonia and apathy
2008). Indeed, recent prospective data from the Leukoaraiosis and (Sheline et al., 2010; Taylor et al., 2005a).
Disability in the Elderly (LADIS) multi-centre study has shown that Of significance, WMLs are also related to functional disability.
in several hundred people, more severe white matter changes at Data from the LADIS study, a longitudinal cohort study investigat-
baseline predicted both depressive episodes and depressive ing the relationship between white matter disease and subsequent
symptoms at both two and three year follow-up periods depression and disability in older adults, suggests that greater
(Teodorczuk et al., 2010). Additionally subcortical disease appears baseline WMLs independently predict functional decline and
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 115

disability in this group (Inzitari et al., 2007). In older adults with 5.2.2. Microstructural findings
severe depressive disorders, the extent of WMLs predicts the long- As discussed above, the strategic location of WMLs may be a key
term disability experienced by these patients (Hickie et al., 1997b), component in conferring vulnerability and/or perpetuating de-
and after controlling for age, gender, depression severity and pressive syndromes in later-life (Dalby et al., 2010; Sheline et al.,
medical comorbidity, subcortical WMLs have been found to be 2008). However, given the association between ageing and white
significantly associated with impairments in instrumental activi- matter integrity, more subtle and diffuse white matter compro-
ties of daily living (Steffens et al., 2002a). Of interest is the recent mise such as microstructural disturbance, could play a role in the
data emerging from the LADIS study, which suggests that in older disorder. This has been assessed using diffusion tensor imaging
adults with WMLs, the relationship with incident depression over (DTI), a variation of MRI that measures the magnitude and
the longer term is somewhat attenuated as the levels of functional orientation of diffusion of water in tissues (Gunning-Dixon et al.,
disability increase (Teodorczuk et al., 2010). The authors suggest 2009). Diffusion occurs equally in all directions if no barriers to
that, over the longer term, a potential secondary pathway emerges, diffusion exist (isotropy). Barriers include myelin sheaths, cell
whereby WMLs lead to functional impairments, which then go on membranes and white matter fibre tracts, and, when present, tend
to amplify depressive symptoms, leading to further functional to cause diffusion to follow the long axis of the barrier (anisotropy).
impairments (Teodorczuk et al., 2010). Thus fractional anisotropy (FA), which is a measure of the strength
Early reports showed that increasing severity of WMLs were of the directional dependence of diffusion and tissue disruption,
associated with decreased response to treatment (Hickie et al., allows for a quantification of brain tissue microstructure.
1995; O’Brien et al., 1998; Simpson et al., 1997). Additionally some DTI studies in LLD utilising a region of interest protocol have
studies have implicated regional specificity in the relationship consistently found widespread reduced FA primarily in frontal
between WMLs and treatment response. Patankar et al. (2007) (Alexopoulos et al., 2009; Bae et al., 2006; Nobuhara et al., 2006;
found that basal ganglia hyperintensities were associated with Taylor et al., 2004; Yang et al., 2007) but also temporal (Nobuhara
failure to respond to antidepressant monotherapy. This mirrors et al., 2006; Yang et al., 2007) regions. Furthermore a more recent
research linking subcortical gray matter hyperintensities with technique, an automated ‘tract-based spatial statistics’ procedure
failure to respond to ECT (Steffens et al., 2001). More recent work that potentially improves DTI analysis and interpretation, also
has found that depressed elders who failed to respond to 12 weeks demonstrated lower FA in frontal, temporal and midbrain regions
of escitalopram therapy exhibited significantly greater WML (Colloby et al., 2011). One study has demonstrated diffuse
burden compared with both normal controls and those depressed abnormalities throughout the whole brain (Yuan et al., 2007).
elders who did experience remission of depression (Gunning- The role of WMLs contributing to the observed reduced FA values
Dixon et al., 2010). There have been conflicting reports (Salloway was investigated by Shimony et al. (2009), who excluded WMLs
et al., 2002; Sneed et al., 2007), however it is possible that use of from the DTI analysis by segmentation techniques. They found that
visual rating scales may have contributed to the difference in even ‘‘normal appearing’’ white matter displayed diffuse micro-
results; in particular a recent double-blind, placebo controlled trial structural damage in LLD. The authors concluded that WMLs
of citalopram in patients aged 75 years or older found that only represent the ‘tip of the iceberg’, implying that their presence
executive function deficits predicted response to the antidepres- indicates that even normal appearing white matter is undergoing
sant and no significant relationship existed between WML load and microstructural change.
treatment response (Sneed et al., 2007). The authors raised the
interesting point that, given the high rate of lesion load in their 5.2.2.1. Relationship with clinical and cognitive features. DTI abnor-
sample combined with the average age of participants (79.6 years), malities in multiple frontostriatal–limbic regions have been
MRI based diagnoses of ‘vascular’ depression may only be useful in correlated with cognitive impairments in LLD, including proces-
younger populations where increasing WML load is likely to be sing speed (Shimony et al., 2009) and poor response inhibition on
relatively specific (Sneed et al., 2007). Perhaps intertwined with the ‘Stroop’ task (Alexopoulos et al., 2002b; Murphy et al., 2007).
treatment responsiveness, it appears that lesion burden confers Furthermore, reduced FA of the right and left frontal white matter
increased risk for illness course or chronicity of depression. For regions lateral to the ACC has been associated with failure to
instance, an early cohort study found that on follow-up (6 months– respond to a 12 week course of citalopram (Alexopoulos et al.,
2 years), only 24% of a small group (n = 37) of older adults with 2002b), and reduced FA of the corpus callosum, left superior corona
severe depression were in remission, whilst in 19% of patients, the radiata and right inferior longitudinal fasciculum with failure to
depression had not improved or had become worse (Hickie et al., respond to 12 weeks of escitalopram (Alexopoulos et al., 2010).
1997b). Increased WML load has also been associated with a more Contradicting these results, failure to remit to 12 weeks of
chronic course of depression (Heiden et al., 2005; Lavretsky et al., sertraline was found to be associated with higher FA values in the
2008). ACC and superior frontal gyrus in a LLD group (Taylor et al., 2008a).
There also seems to be an important link between WML and These results are unexpected given these regions have previously
factors associated with homocysteine metabolism in patients with been found to demonstrate depression-related DTI abnormalities.
depression. For instance, in a clinical sample of 47 older patients Possibly, methodological differences (voxel based analysis versus
with MD, low vitamin B12 levels (but not folate) were predictive of region of interest approaches) may play a role, however the
more extensive WMLs on imaging (Hickie et al., 2005a). Total authors suggest that in some instances of LLD, impaired functional
plasma homocysteine levels have also been independently related connectivity may be secondary to structural changes, and these
to deep WMLs in healthy men aged 60–64 years (Sachdev et al., may be responsive to the neurotrophic effects of antidepressants,
2004). On the other hand, although the extent of WMLs was whereas in other cases functional connectivity deficits occur in the
positively correlated with plasma homocysteine concentration, the face of intact structural connections. In this case, poorer response
association between higher plasma homocysteine and depression to antidepressants may be expected (Taylor et al., 2008a).
remained even after controlling for WMLs, suggesting that the
relationship may not simply be due to vascular disease (Chen et al., 6. Macromolecular findings
2005). Thus consideration of a multifactorial approach that takes
into account the complex relationship between VRFs, dietary As detailed so far, white matter and volumetric abnormalities
factors, and structural brain changes is likely to be paramount to at a macro- and micro-structural level in frontostriatal and
primary and secondary intervention approaches. limbic circuitry are implicated in aetiological models of LLD.
116 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

Complementing these findings, alternative imaging modalities phosphate containing molecules and provides a measure of
have started to provide further insights into possible in vivo and bioenergetic changes, suggests altered energy metabolism in
molecular changes. In particular, magnetization transfer imaging LLD (Forester et al., 2009), and points to mitochondrial dysfunction
(MTI), producing the magnetization transfer ratio (MTR), and as playing a potential role in the pathophysiology of LLD.
magnetic resonance spectroscopy (MRS) have begun to enhance This technique permits in vivo neurochemical evaluation of the
our understanding of LLD by providing information regarding the brain. Different molecules have unique magnetic resonance signals
biophysical and biochemical characteristics of particular brain that can be quantified by taking the area under the signal curve
regions (see Fig. 1b). (Hoptman et al., 2006); as values are not absolute, they are typically
reported as ratios to a standard metabolite. Nearly all studies of LLD
6.1. Magnetization transfer ratio have employed 1H NMR (proton spectroscopy), which measures
compounds such as choline (cho), N-acetyl aspartate (NAA),
MTI allows the examination of the macromolecular structure of myoinositol (mI) and creatine (Cr). The cho signal is likely a
cerebral white matter based on the interaction of the normally composite signal with contributions from free choline, phosphocho-
observed tissue water signal and protons contained in large line and glycerophosphocholine (Kumar et al., 2002); as these
macromolecules (mainly myelin) (Hoptman et al., 2006). Water in choline-containing metabolic compounds are essential constituents
biological tissues falls into two classes: ‘bound’ water (i.e. water of the cell membrane, as well as second messenger transmitters, cho
associated with macromolecules), and ‘free’ water (i.e. water that ratios are thought to reflect membrane turnover, degree of
is free to move about within biological compartment restraints). myelination and cell density (Miller, 1991). Thus increased cho/Cr
These two forms of tissue water produce different magnetic seen in the frontal white matter may reflect altered neuronal
resonance signal characteristics so may be separated out for membrane structure (Kumar et al., 2002). mI is involved in
analysis. Two images are acquired: a control acquisition, which is phosphoinositol cycling, thus plays an important role in the second
like a usual MRI study, and a MT acquisition (an ‘off-resonance’ messenger systems (Ross, 1991); it is also considered a marker of
radiofrequency pulse that interacts only with the bound water and glial cells. Increases in the mI/Cr may therefore represent
cancelling it out). As ‘free’ water is the predominant component of disruptions in the receptor-second messenger system complex
the MRI signal due to the broad distribution of the ‘bound’ water (Kumar et al., 2002) or glial dysfunction. Finally NAA is located
signal across a wide range of frequencies, the MTR, the difference in predominantly in neurons, therefore it acts as a marker of neuronal
signal intensity between the two images, reflects the concentration viability and density; NAA has also been demonstrated to be
of macromolecules in the tissue as well as their chemistry (see expressed by oligodendrocytes, thus suggesting that reduced NAA/
Hoptman et al., 2006). There are few studies utilising MTR in the Cr may not only represent neuronal loss but also disruption in the
examination of LLD. Kumar et al. (2004) examined normal formation and maintenance of myelin (Venkatraman et al., 2009).
appearing white matter and subcortical regions in eight patients Early studies demonstrated raised levels of cho in a large voxel in the
with LLD and eight healthy age-matched controls. Significantly subcortical region (including the caudate, internal capsule and parts
lower MTRs were noted in the depressed group, located in the genu of the temporal lobe) in LLD compared with controls; the increased
and splenium of the corpus callosum, the right caudate nucleus cho/Cr normalised after treatment with nefazodone (Charles et al.,
and putamen and the occipital white matter. A more recent study 1994). Biochemical abnormalities may be more apparent in frontal
(Gunning-Dixon et al., 2008) found that LLD is characterized by white matter. For instance, significantly increased ratios of mI/Cr
reduced MTRs in multiple left sided frontostriatal and limbic and cho/Cr were found in the left dorsolateral frontal lobe white
regions, including the white matter lateral to the lentiform nuclei, matter in a LLD group compared with controls (Kumar et al., 2002).
dorsomedial and dorsolateral prefrontal, dorsal anterior cingulate, No significant differences were noted in the anterior cingulate grey
periamygdalar, subcallosal, lentiform, insular and posterior matter bilaterally. In the only known study to stratify the depressed
cingulate regions. Additional regions associated with decreased group by age of onset of depression, Murata et al. (2001) found that
MTR in the left hemisphere include the thalamus, splenium of the compared with EoD, LoD was associated with poorer cognition and
corpus callosum, inferior parietal, precuneus and middle occipital more severe depression, as well as increased WMLs, which in turn
regions. Use of MTI in disorders characterized by damaged myelin were associated with decreasing levels of NAA/Cr in the white
and axonal loss such as multiple sclerosis has demonstrated a matter of the frontal lobes. Interestingly no difference in cho/Cr was
range of MTR abnormalities, suggesting that lower MTRs reflect reported. Extending these findings, LLD has been associated with
compromised myelin integrity and axonal damage (Filippi and significantly lower NAA/Cr in the left frontal white matter and
Agosta, 2007). Finding such markers of myelin damage in the white higher cho/Cr and mI/Cr in the left basal ganglia compared with
matter and subcortical nuclei of patients with LLD dovetails with controls. Moreover the mI/Cr correlated with global cognitive
the ‘tip of the iceberg’ concept; and suggests that the pathophysi- function in the LLD group but antidepressant therapy had no
ological changes underpinning LLD may be more widespread than significant effect on MRS biochemical ratios (Chen et al., 2009).
initially recognised. Moreover changes in the macromolecular pool Contrasting previous work, Venkatraman et al. (2009) examined
of the white matter may be related to metabolic changes (Wyckoff biochemical disturbances in remitted LLD, and found reductions in
et al., 2003). total NAA, cho and Cr in the medial PFC (anterior cingulate),
accompanied by elevations in NAA and mI in the left medial
6.2. Magnetic resonance spectroscopy temporal lobe (amygdala). These findings are interesting for two
reasons: firstly they are somewhat conflicting with results of studies
Taken together, results from these few studies suggest that LLD in acute LLD, demonstrating biochemical changes including reduced
may encompass disruptions in neuronal membrane structure, as cho signal in the anterior cingulate. These data therefore point to the
well as neuronal loss, glial dysfunction and possible dysregulation possibility of both state and trait neurochemical markers of
in the second messenger system. The pattern of biochemical depression (Venkatraman et al., 2009). Secondly, the results indicate
abnormalities may be significant, findings of altered mI and cho/Cr that even with resolution of acute depression, underlying biochem-
in the absence of NAA/Cr disturbance emphasizes the potential ical abnormalities thought to reflect neuronal and glial changes
importance of the glial compartment (Kumar et al., 2002). remain. These findings support the neuropsychological data cited
Additionally, research using Phosphorus (31P) MRS, which is used above, which displays persistence of deficits despite symptom
to quantify concentrations of metabolites including high energy resolution (Box 3).
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 117

7.1. Positron emission tomography (PET)


Box 3. Structural neuroimaging studies.

 Volumetric MRI studies show increased ventricular size and PET involves the use of radionuclides that produce positrons,
reductions in prefrontal, hippocampal, amygdala and stria- which in turn emit photons during the process of decay. These
tal (including caudate) volumes in patients with LLD photons can be sensed by radiation detectors and in conjunction
 Volumetric changes in LLD appear to be associated with with computer reconstruction are used to produce three-dimen-
clinical/cognitive features and medication effects sional tomographic images of the brain. Unlike SPECT, PET allows
 White matter lesions are more frequent and severe in LLD, for absolute quantification of perfusion or metabolic rate as well as
supporting the ‘vascular’ depression model
high temporal and spatial resolution (D’haenen, 2001; Nobler et al.,
 While white matter lesions may disrupt fronto-subcortical
circuitry critical for mood and emotional circuits, both lesion 1999b; Soares and Mann, 1997).
location and lesion volume may be important for the ex- In terms of investigating cerebral changes in depression, PET
pression of clinical features and cognitive impairment and has also offered a methodology by which direct examination of
for influencing treatment response serotonergic functioning is possible. This has occurred in the
 Even in the absence of white matter lesions, DTI studies context of increasing interest in the interaction between cognitive
show microstructural changes are evident in patients with decline and mood dysregulation, influence of age- and disease-
LLD and relate to cognitive and clinical features related processes on the serotonergic system (Meltzer et al., 1998)
 Emerging MTI and MRS studies suggest widespread mac- as well as the integration of neuropharmacological and anatomical
romolecular changes and in vivo biochemical abnormalities
information (Soares and Mann, 1997). While the majority of
may be implicated in LLD pathophysiology
studies have focused on younger samples, such studies have
generally indicated decreased 5-HT transporter density and
binding sites in depressed patients relative to healthy controls
(see Nobler et al., 1999a). The development of radioligands such as
7. Functional neuroimaging studies altanserin and [11C]WAY-100635 have enabled targeted investi-
gation of pre- and post-synaptic serotonergic function. Studies
Functional neuroimaging studies conducted in MD have using [18F]altanserin PET have, however, produced inconsistent
enabled more sophisticated heuristic frameworks for modelling results. For instance, one study showed no significant difference in
MD circuitry than that offered by structural imaging alone 5-HT2A binding potential between 11 patients with LLD and a
(Krishnan and Nestler, 2010). Functional imaging has encom- control group across 12 brain regions (Meltzer et al., 1999). By
passed a range of methods including positron emission tomogra- contrast, Sheline et al. (2004) showed significantly reduced (i.e.
phy (PET), single photon emission computed tomography (SPECT) 39%) hippocampal receptor binding in a group of 16 patients with
and functional magnetic resonance imaging (fMRI), typically LLD, relative to controls. No significant differences were found
conducted at rest or within a treatment, emotion-activation or between early- and late-onset sub-groups. In terms of the 5-HT1A
cognitive-induction paradigm. More recently, resting state fMRI receptor, Meltzer et al. (2004) demonstrated significantly reduced
has also been employed as well as magnetoencephalography binding potential in the dorsal raphe nucleus in 16 depressed
(MEG), although to-date few studies using these techniques have elderly patients compared to 17 healthy controls.
been published in LLD specifically. A limitation of this body of More recent PET studies using various ligands including
literature is the considerable variability in study designs, the [18F]
fluorodeoxyglucose (FDG), [18F]setoperone as well as
heterogeneity of participant samples (e.g. medicated versus [18F]
altanserin have provided some further understanding of
untreated, variable age of onset, comorbid VRFs) as well as the changes in cerebral activation in LLD more specifically, although
various approaches taken in analysing data. Nonetheless, these sample sizes are typically small. For example, de Asis et al. (2001)
techniques provide further elucidation of the specific brain regions used a high sensitivity [15O]H2O (8 mCi) slow bolus rCBF imaging
and neural networks involved in depression (see Figure 1b) and technique to demonstrate bilateral rCBF deficits in the dorsal
antidepressant treatment mechanisms, as well as providing anterior cingulate and hippocampus in six patients with severe
diagnostic clarification and treatment guidance from a pathophys- depression compared to five controls, both at rest and during a
iological perspective. word generation task. In a sample of 19 patients with LLD, Smith
Major advances in the use of radioligands have allowed for et al. (2004) found that relative to controls, the depressed sample
specific investigations into regional cerebral blood flow (rCBF) and had increased glucose metabolism bilaterally in frontal and post-
cerebral metabolism (CM). They have allowed for the direct central cortices, as well as in the left middle frontal and middle
examination of relevant neurotransmitter systems notably 5-HT, temporal cortices, inferior parietal lobule, occipital cortex and
which has long been linked to the pathophysiology of mood cerebellum. Additionally, Smith et al. (2009a) measured cerebral
disorders. Many studies in the 1990s quantified rCBF or CM resting glucose metabolism using FDG-PET, and reported cerebral glucose
state abnormalities in LLD samples. The majority of studies hypermetabolism in anterior and posterior cortical regions in the
consistently reported global or topographical reductions in frontal context of cerebral atrophy in 16 depressed older adults with
(PFC, ACC) or subcortical (caudate) regions, with the topographical moderate to severe depression relative to 13 control subjects. Also
reductions most conspicuous in older and more severely depressed using FDG-PET, other investigators have shown that in compari-
patients. Metabolic reductions were often corrected with antide- son to control subjects, treatment-resistant LoD is associated with
pressant medication (Drevets, 1998; see Nobler et al., 1999b; decreased metabolism in the anterior cingulate, temporal and
Soares and Mann, 1997). While similar reductions have been parietal regions, as well as increased metabolism in other regions
demonstrated in younger samples, Soares and Mann (1997) and including the dorsal frontal region and basal ganglia (Fujimoto
Nobler et al. (1999b) suggest that the greater extent and et al., 2008). Furthermore, an earlier study by Kumar et al. (1993)
topography of deficits in older depressed patients may relate to demonstrated a pattern of regional cerebral metabolic changes in
an interaction between normal aging processes and depression eight participants with LLD, which was comparable in magnitude
pathophysiology. The basis of this interaction may relate to to the pattern seen in a group of eight participants with AD.
changes in cerebral vasculature associated with normal ageing in Changes were characterized by widespread decrements in major
combination with the proposed relationship between ischemic neocortical, paralimbic and subcortical regions relative to healthy
vascular changes and depression. controls.
118 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

7.1.1. Associations with clinical features may reflect a compensatory up-regulation of the 5-HT2A receptor
As noted in a recent review by Smith et al. (2009a), the available as a result of prior pharmacotherapy. Further investigations
data linking perfusion or metabolism abnormalities with clinical include D’haenen’s (2001) account of two PET studies showing
features and treatment efficacy in LLD is limited, although has been decreased 5-HT2A receptor binding following tricyclic treatment.
reviewed elsewhere (for example, see Nobler et al., 1999a,b, 2000). Using SSRIs, however, increased receptor binding has been
Examples of reported associations between clinical features and reported. Specifically, reduced 5-HT1A binding potential in the
physiological measures include symptom severity, sleep quality dorsal raphe nucleus may predict time to remission following
and treatment responsiveness. For example, in the empirical study paroxetine treatment (Meltzer et al., 2004).
by Smith et al. (2009a) described above, greater cerebral In the context of treatment effects, PET has also been used to
metabolism in anterior and posterior cortical regions relative to illustrate Mayberg’s (1997) attribution of sleep and other somatic/
controls was also associated with more severe symptoms of vegetative disturbances to dysregulation of a ventral compartment
depression and anxiety. Meltzer et al. (2004) investigation of 5- comprising paralimbic cortical, subcortical and brainstem regions
HT1A receptor binding in 16 depressed older adults (described within a limbic-cortical model of depression. According to
above) further demonstrated a positive association between the Mayberg’s review, various PET paradigms in depressed patients
magnitude of the binding potential measured in the dorsal raphe as well as post-fluoxetine treatment have indicated that depressive
nucleus and the severity of depression. This latter somewhat illness is associated with increased ventral paralimbic activity,
paradoxical finding may reflect a failure of adaptive mechanisms in while improved mood and particularly remission of sleep and
patients with more severe depression. A failure to induce adaptive vegetative disturbances correlates with ventral paralimbic sup-
changes was also postulated to underlie differences in cerebral pression in fluoxetine-responders. Additionally, PET has been
glucose metabolism alterations over a treatment course of six utilised in sleep deprivation studies, which have examined
weeks between middle-aged SSRI treatment responders and non- whether acute sleep deprivation (particularly in conjunction with
responders, as reported by Mayberg et al. (2000). antidepressant treatment) can be used as a therapeutic strategy to
improve depressive symptoms, possibly by recruiting dopaminer-
7.1.2. Medication and treatment effects gic and serotonergic systems, which, in turn, promote neurogen-
PET studies have perhaps been most elegantly employed to esis (Wu et al., 2008). Initial applications of this paradigm within
elucidate the efficacy of various treatment paradigms in terms of the LLD literature were promising. For example, in a sample of six
affective and cognitive symptom remission as well as alterations in elderly depressed patients, Smith et al. (1999) reported a
neural circuitry. An example of this application is the recent study significant decrease in depression severity after total sleep
by Diaconescu et al. (2010), who further clarified the effect of SSRI deprivation, after recovery and after antidepressant treatment,
treatment in LLD in an exploration of the functional networks effects which coincided with decreases in cerebral glucose
associated with affective and cognitive improvement following metabolism in the right anterior cingulate and medial frontal
eight weeks of treatment with citalopram. Sixteen older depressed gyrus following sleep deprivation. The decrease in metabolism in
patients with a mean age of onset of 60 years underwent FDG-PET the anterior cingulate persisted after recovery sleep and after two
as well as psychiatric evaluation and neuropsychological assess- weeks of paroxetine treatment. These promising results were
ment. Consistent with expectations, PET results demonstrated confirmed in subsequent studies by this group (Smith et al., 2002)
decreased cerebral glucose metabolism in the anterior cingulate, although a more recent large randomised controlled trial (RCT) (of
middle temporal gyrus, precuneus, amygdala and parahippocam- sleep deprivation, paroxetine and placebo) failed to corroborate
pal gyrus; as well as increased glucose metabolism in the putamen, these findings, suggesting that there were no differences in clinical
occipital cortex and cerebellum. Further analyses of correlations response or remission after controlling for baseline depression
between treatment-related changes in glucose metabolism and severity (Reynolds et al., 2005). A PET sub-study involving 16 of
clinical symptom (i.e. affective/cognitive) improvement also these patients (Smith et al., 2009b) further indicated a lack of
identified distinct functional networks. Specifically, a subcorti- evidence for an accelerated response to antidepressant treatment
cal-limbic-frontal network was associated with improved depres- in combination with sleep deprivation.
sion and anxiety symptoms, whilst a separate medial temporal- Addressing the depression–dementia interface, the Pittsburgh B
parietal-frontal network was associated with cognitive improve- ([11C] 6-OH-BTA-1) (PiB) ligand may offer the opportunity to
ments (i.e. verbal fluency, verbal learning and memory). Mayberg examine the presence of b-amyloid pathology in those patients
et al. (2000) also utilised FDG-PET to investigate the time course of with LLD who have preclinical forms of dementia or in those with
cerebral metabolic changes associated with SSRI (fluoxetine) cognitive impairment. This compound has shown high tracer
treatment, albeit in a mid-life sample of 10 depressed males. retention in cortical areas in patients with clinical diagnoses of
Results indicated a significant change in cerebral glucose metabo- probable AD and low retention in age-matched controls. In a small
lism after one week of treatment, with both increases and decreases clinical sample of nine patients with remitted MD, Butters et al.
seen in the subcortical, limbic–paralimbic and (to a lesser extent) (2008) showed that in those meeting criteria for MCI, 50%
cortical regions. A second follow-up after six weeks of fluoxetine demonstrated PiB retention levels similar to those observed in
treatment generally indicated a reciprocal pattern of cortical AD, while another three patients with MCI showed intermediate
metabolic increases and limbic–paralimbic decreases. Three pat- levels of PiB retention. More recently, the 2-(1-(6-[(2-[18F]fluor-
terns of change over time were identified, including (a) comparable oethyl)(methyl)-amino]-2-naphthyl)radiethylidene) malononi-
changes in glucose metabolism at one and six week scans; (b) trile ([18F]FDDNP) radioligand has also been utilised to
reversal of the one week pattern of changes at the six week scan; and, investigate amyloid and tau distribution in critical brain regions
(c) unique changes evident only at six weeks. The pattern of reversal in individuals with LLD. Kumar et al. (2011) reported significantly
was thought to suggest a process of adaptation to sustain serotonin higher [18F]FDDNP binding overall and specifically in the posterior
reuptake inhibition in specific regions over time. cingulate and lateral temporal regions in 20 LLD patients compared
Antidepressant medication may also mediate 5-HT binding to 19 healthy controls matched for age, sex and level of education.
potential, as demonstrated in a study by Sheline et al. (2004), Importantly, the authors noted that [18F]FDDNP has previously
whereby six untreated patients showed reduced hippocampal 5- been shown to discriminate between those with AD and healthy
HT2A receptor binding in comparison to 10 previously treated controls, and has demonstrated a progression in binding from MCI
depressed patients. The authors postulated that this difference patients to those with dementia. These findings from the LLD
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 119

sample provided further evidence of a pathophysiological pathway showed significant regional reductions in 18 older patients relative
mediated by amyloid and tau distribution that may predispose to age-matched controls, with the greatest decrements observed in
older adults to mood and associated cognitive/behavioural the anterior cingulate, superior frontal and superior temporal gyri
syndromes. Further larger studies utilising these compounds are and the caudate nucleus. Similarly, Curran et al. (1993) demon-
now needed in order to determine their prognostic value for strated reductions in frontal and temporal cortices as well as the
determining longitudinal dementia risk. anterior cingulate, caudate and thalamus in 12 older medicated
male patients; although the authors noted a possible interaction
7.1.3. Summary of PET studies effect of continuing antidepressant or benzodiazepine treatment.
In summary, while PET imaging has been widely utilised to More recently, reductions in frontal, temporal and parieto-
study cerebral activation in LLD, studies have typically included occipital regions were reported in 25 depressed patients older
small sample sizes, and have largely focused on the serotonergic than 55 years relative to healthy controls (Ishizaki et al., 2008).
system as a key source of pathophysiology. Such studies have Overall, this data yields reasonably consistent support for cortical
suggested reduced serotonergic binding in frontal (PFC and ACC), rCBF reductions across frontal, temporal and parietal areas as well
hippocampal and brainstem (dorsal raphe nucleus) regions and as reduced rCBF in the basal ganglia.
some have reported hypermetabolism in similar (e.g. prefrontal) or
other regions such as the basal ganglia, temporal and parieto- 7.2.1. Associations with clinical features
occipital regions. While few clinical features have been studied in Findings have been inconsistent regarding the relationship
detail, several studies suggest that depressive symptom severity between clinical features and cerebral activation in depressed
relates to cerebral metabolism alterations. PET has also been elderly. This may be due to the heterogeneity of clinical variables
utilised to explicate the efficacy of antidepressant treatment, with investigated and reported in the literature. In terms of baseline
additional exploration of other features of medication effects such clinical parameters (e.g. age, age at onset, gender, number of
as the time course of cerebral metabolism changes and associa- episodes, depression severity or global cognition scores), no
tions with affective/cognitive symptom improvement. The litera- significant correlations were reported in a sample of 18 older
ture pertaining to effects of sleep deprivation and treatment patients with on average mild depressive symptoms (Awata et al.,
responsivity is more disparate possibly due to the small sample 1998). Likewise, Navarro et al. (2001) did not find correlations with
sizes typically studied. clinical features in a sample of 30 patients with more severe
Ultimately, continued development of new PET biomarkers will depressive symptoms. Despite these findings, however, some data
afford the opportunity to investigate the pathogenesis of depres- suggests a relationship between rCBF and treatment responsive-
sion from new perspectives. One ligand with promise for ness. For instance, the former study reported robust and extensive
examining the vascular and inflammatory mechanisms that are reductions in anterior cerebral regions in a sub-group with
likely to relevant to aetiology is the [11C](R)-PK11195 ligand. This refractory depression relative to a non-refractory group and to
ligand displays up-regulation of peripheral benzodiazepine controls, indicating that the nature of hypoperfusion, particularly
binding sites, which provides a cellular marker for microglial in the anterior cingulate and prefrontal regions, may be related to
activation, thus representing a measure of inflammatory activity refractoriness and chronicity of depression. Furthermore, left
(Banati and Hickie, 2009). Initial studies utilising this ligand in anterior frontal abnormalities associated with acute depression
multiple sclerosis displayed increased binding in association with have been shown to normalise with remission, as seen at 12-
focal white matter pathology as well as in grey matter (Banati et al., month follow-up (Navarro et al., 2002).
2000). The PK11195 ligand has since been utilised across a range of There may be some prognostic value in SPECT abnormalities as
disorders including AD (Edison et al., 2008), MCI (Okello et al., illustrated by a study by Ritchie et al. (1999) where the finding of
2009), Parkinson’s disease (Gerhard et al., 2006a) and progressive greater reductions in rCBF in patients with dementia and
supranuclear palsy (Gerhard et al., 2006b). However, to-date, no depression was paralleled by clinical differences between the
known studies have utilised this ligand to measure potential groups over time. That is, in the two years prior to dementia
inflammatory activity that may be critical to the pathogenesis of diagnosis, fewer affective and reactive symptoms and greater
LLD generally, or LoD more specifically. psychomotor change was apparent. Finally, some data from
middle-aged samples suggests that atypical depression may
7.2. Single photon emission computed tomography (SPECT) manifest with differential HMPAO tracer uptake compared to
typical depression (Pagani et al., 2007). Such results underscore the
Similar to PET, SPECT utilises radioligands (typically hexam- importance of considering disease heterogeneity when analysing
ethyl-propylene amine oxime [HMPAO]) for brain imaging. SPECT neurobiological findings and highlight the need to consider the
yields lower resolution images in comparison to PET; however it is presenting clinical and aetiological symptom complex.
significantly less expensive and therefore utilised more frequently
in research trials. 7.2.2. Medication effects
Resting state studies attempting to quantify changes in rCBF in To our knowledge, only a small number of studies have utilised
MD have generally indicated regional reductions in the frontal and SPECT to examine the effects of antidepressant treatment
temporal lobes as well as subcortical areas (e.g. caudate). In older specifically in elderly depressed samples. For example, Ishizaki
samples specifically, Upadhyaya et al. (1990) examined 18 patients et al. (2008) examined 25 depressed older adults and demonstrat-
aged over 66 years as well as 12 controls using HMPAO SPECT. ed that after 14 weeks of fluoxetine treatment, significant rCBF
Their data suggested global deficits as well as regional reductions improvements were apparent in areas that were reduced at
in the frontal, temporal and parietal regions. These regional baseline including the left DLPFC, precentral regions and right
reductions were also reported by Lesser et al. (1994) who used parieto-occipital regions. In an earlier study, Nobler et al. (2000)
HMPAO SPECT to assess 39 non-medicated patients aged over 50 utilised the planar 133Xenon inhalation technique to assess global
years in comparison with 20 age-matched controls, and by CBF in 20 older depressed patients treated with antidepressant
Ebmeier et al. (1998) in 39 patients relative to controls. The latter medication over six to nine weeks. This RCT of TCA (nortriptyline)
group of patients was divided by age of onset with results and SSRI (sertraline) treatments was not associated with global
indicating that reductions in bilateral temporoparietal cortex CBF changes, although notably the sample size was very small
perfusion were associated with LoD. Awata et al. (1998) further (n = 6 and 5 respectively). However, treatment responders showed
120 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

significantly greater regional CBF reductions in selective prefrontal have prognostic utility in terms of longitudinal course or
and anterior temporal regions compared to non-responders, and progression to dementia.
the magnitude of change correlated with the extent of clinical
improvement on the Hamilton Depression Rating Scale. Other 7.3. Functional magnetic resonance imaging (fMRI)
studies have also investigated differences in rCBF between
antidepressant responders and non-responders, though these Functional MRI (fMRI) elicits patterns of blood oxygen level
have included general adult samples rather than older adults dependent (BOLD) activation during periods of rest, activity or
specifically. For example, in 65 middle-aged depressed patients, cognitive functioning. BOLD is an MRI marker of deoxyhaemoglo-
Joe et al. (2006) noted that a sample of 35 citalopram responders bin and reflects changes in cerebral blood flow and oxygenation.
initially demonstrated increased perfusion in the left frontal and These haemodynamic alterations resulting from neural activity
insula regions, right posterior cingulate and inferior frontal cortex, manifest as changes in BOLD signal intensity, with areas of higher
the latter of which persisted four weeks later. By contrast, 30 non- BOLD signal intensity reflective of increased concentration of
responders showed a significant rCBF increase in the left posterior oxygenated haemoglobin in the particular brain region. Relative to
cingulate and putamen. A group-by-time analysis demonstrated other functional imaging techniques (e.g. magnetic resonance
significantly different changes in rCBF in the two groups indicating spectroscopy, electroencephalography, nuclear neuroimaging),
opposite directed courses, with responders showing a decrease and fMRI is less invasive, has less radiation exposure, has higher
non-responders showing an increase in the posterior cingulate, left spatial resolution and is more widely available.
inferior frontal gyrus and left putamen. It was postulated that the
posterior cingulate region may be implicated in the pathophysiol- 7.3.1. Behavioural and cognitive paradigms
ogy of depression and may have predictive value for treatment. To-date, studies utilising fMRI have largely focussed on the PFC
Using a similar methodology, Brockmann et al. (2009) studied 93 (Ebmeier et al., 2006) and have utilised emotion-based paradigms.
middle-aged depressed adults. In this sample, initial responders Surprisingly, few of these studies have been conducted in older
also demonstrated significantly higher rCBF in prefrontal, tempo- and/or LoD samples.
ral, and insula cortex regions in comparison to non-responders. Using an emotional paradigm, Brassen et al. (2008) examined
However, these differences did not persist after four weeks. 13 older antidepressant naı̈ve female patients with LoD and 13
Overall, these studies demonstrate that SPECT has some capacity to controls. They employed an emotional evaluation task of positive,
differentiate between treatment responders and non-responders, negative and neutral words in association with fMRI and showed
whereby responders show higher perfusion in key cortical and that although there were no performance differences between
possibly also, basal ganglia regions. groups, the depressed group showed decreased BOLD activation in
the ventromedial PFC in response to negative words, compared to
7.2.3. Relationship to cognition positive stimuli, relative to control participants. Follow-up data
Regional decrements in frontal and temporal cortices as well as after seven months for this sample showed that the attenuated
the anterior cingulate, caudate and thalamus in male depressed response had normalised and was associated with a significant
patients have been associated with impairment of performance on improvement in depressive symptoms. In a study specifically
Part B of the Trailmaking Test, a set-shifting and visuomotor task examining executive functioning in LoD, Wang et al. (2008b)
(Curran et al., 1993). Also using SPECT, Hickie et al. demonstrated employed an emotional odd-ball paradigm in 12 older patients
that reductions in striatal rCBF are related to performance on a with current MD, 15 patients with remitted MD and 20 controls.
reaction time task, particularly when the task is associated with a Depressed patients demonstrated reduced BOLD activation in
more complex choice- as opposed to simple-reaction time regions involved in executive functions (i.e. right middle frontal
requirement (Hickie et al., 1999). Using an MRI and SPECT co- gyrus, cingulate and inferior parietal areas) relative to controls.
registration method with a region of interest analysis, Hickie et al. Reduced activity in the middle frontal gyrus appeared to be state-
(2007a) later demonstrated that such striatal changes are not only dependent, as attenuated activity persisted in remitted partici-
associated with increasing task complexity but are also associated pants only in the anterior portion of the posterior cingulate and
with performance on the Trailmaking test (postulated to reflect inferior parietal regions. Consistent with this observation of
fronto-subcortical functioning) as well as with WML load. While posterior cingulate and parietal areas being implicated in the
few studies have examined the predictive longitudinal utility of pathophysiology of the disorder, Woo et al. (2009) recently
SPECT, some data suggests that LLD may intensify the decrements investigated the impact of depressive symptoms on the role of the
in brain function occurring with neurodegenerative processes. posteromedial cortex in an active memory task. Sixty-two older
Specifically, Ritchie et al. (1999) reported greater reductions in left participants with sub-threshold depressive symptoms underwent
temporal rCBF in elderly people who had both depression and fMRI scanning whilst completing a face-name associative memory
dementia, compared to those with dementia alone. encoding task incorporating both ‘familiar’ and ‘novel’ conditions.
Data indicated a cluster within the posteromedial cortex whereby
7.2.4. Summary of SPECT studies reduced BOLD activation during encoding was associated with
Overall, studies utilising SPECT have generally reported depressive symptom severity, history and anxiolytic use. Since this
decreased rCBF in frontal, temporal and subcortical (e.g. caudate) sample did not include cases of clinical depression, however,
regions in samples of older adults with LLD, relative to healthy, further studies in clinical samples would be required to enhance
non-depressed controls. Data regarding relationships to clinical the relevance of these findings with respect to neurobiological
features has been sparse and to-date has been inconsistent. Some models of LLD. One recent study by Aizenstein et al. (2011)
studies in middle-aged samples suggest that antidepressant attempted to do this by relating the characteristic structural
treatment is associated with improved rCBF in key cortical regions, changes of LLD (namely white matter hyperintensities) with
although data in LLD specifically is lacking. Few studies have functional neuroanatomical models of depression. Not only was
examined the relationship between SPECT-defined rCBF changes greater activity in the subgenual cingulate observed in depressed
and cognitive dysfunction, although in those that have data elderly compared with healthy controls in response to a facial
suggests that changes in the striatum do relate to performance on expression affective reactivity task, but also task-related
tasks thought to reflect fronto-subcortical functioning. Further activity was positively correlated with the burden of white matter
studies are required in order to determine whether SPECT changes disease. The depressed group also showed a significantly greater
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 121

interaction between WML and fMRI activity effect than controls. across the entire striatum (Balleine and O’Doherty, 2010). An
This data supports the notion that WMLs disrupt key regions additional consideration is the dynamic interaction between the
required for the control of limbic activity. frontal and striatal regions in mediating performance. Ongoing
While the striatum has been implicated in various structural cognitive paradigms that seek to inform our knowledge of how
volumetric investigations, as well as in research using PET and functions are partitioned within this region (Owen, 2000) may help
SPECT (as described in Sections 6.1 and 6.2 above), few fMRI to further delineate the conditions upon which the broader fronto-
studies have specifically sought to probe the frontostriatal circuitry subcortical networks are involved in symptom expression and
in MD generally or in LLD specifically. However, some investigators cognitive dysfunction. Similarly, studies may need to increasingly
have attempted this task using motor sequencing paradigms, focus on homogenous samples including those with LoD or
which assess implicit sequence learning, a cognitive function that melancholia, where both subtypes have been associated with
has been shown across many disease groups to be sensitive to distinct cognitive profiles (Exner et al., 2009; Naismith et al., 2003).
frontostriatal dysfunction. One fMRI study conducted by Aizen-
stein et al. (2005) utilised an implicit learning task in 11 elderly 7.3.2. Summary of fMRI studies
patients with MD and 12 matched controls. This study reported no Overall, there has been a relative dearth of fMRI studies that
difference between groups in striatal activation during this task. have specifically sought to examine the neurobiological circuitry
However, the groups did differ in BOLD activation during an implicated in LLD. Of those that exist, it seems that reduced PFC
explicit learning task, where increased striatal activation and activation may underpin emotion-specific effects (i.e. negative
decreased prefrontal cortex activation were evident in the emotions) and may relate to depressive ‘state’ phenomena. Other
depressed group. In a later study of 13 elderly males with LLD posterior circuitry changes have also been reported, even in the
and 13 controls, Aizenstein et al. (2009) again reported decreased remitted state and in those with sub-threshold symptomatology.
prefrontal activation, as well as reduced functional connectivity Reductions in posterior regions have also been associated with
between dorsolateral PFC and dorsal anterior cingulate regions, in memory encoding. A small but concise set of studies has
the depressed group during an executive-control task involving specifically attempted to probe the frontostriatal circuits, and
response inhibition. More recently, Naismith et al. (2010b) also has demonstrated that frontal, as opposed to striatal mechanistic
employed a motor sequencing task with fMRI in an older depressed changes likely underpin implicit sequence learning deficits,
sample of 22 patients and controls. While this data supported that although there are some circumstances in which increased striatal
of Aizenstein et al. (2005) in demonstrating no between group- activity may be required. Further studies in this area may be able to
effects in striatal activity, patients with MD did show performance delineate specific neural network correlates of various phenotypic
decrements in implicit sequence learning (Naismith et al., 2006), a expressions and may also elucidate the threshold upon which such
finding which appeared to relate to increased activation of the network dysfunction begins to manifest (e.g. task complexity,
DLPFC (middle frontal gyrus). This data highlights the need to effortful tasks, treatment responsiveness).
consider the broader neural networks when evaluating fronto-
subcortical circuitry in MD, and that different components of the 7.4. Resting state fMRI
network may underpin dissociable cognitive and clinical phenom-
ena. Resting state fMRI is an emerging novel technique for
Data from the above studies is in accordance with that found by determining functional connectivity between brain regions and
Rostami et al. (2009), where there was no evidence of striatal networks. It examines the level of co-activation between the
activation in a goal-directed implicit learning task despite functional time series of anatomically separated brain regions
significant learning-related activity in prefrontal, precentral and during rest. Studies show that even at rest, there is a high level of
occipital regions. In this regard, it has been suggested that the basal connectivity suggestive of communication between motor, func-
ganglia are more integral to the performance (i.e. reaction time) tional (i.e. visual and auditory), emotion and cognitive networks
component and general demands of implicit learning tasks, rather (see review by van den Heuval and Hulshoff Pol, 2010). To-date,
than being responsible for the specific learning component, which there is a paucity of studies utilising this technique in MD and even
may be mediated by a more distributed network including the less in LLD, although many are currently in progress. In addition to
frontal and cerebellar regions (Exner et al., 2002). Certainly, our functional connectivity analysis, a new technique known as
prior work has suggested that the striatum likely underpins some assessment of regional homogeneity has recently been reported
of the functional changes associated with slowed reaction time within this developing area. Yuan et al. (2008) described this
(Hickie et al., 1999, 2007a; Naismith et al., 2002). However, in MD, technique as a means of exploring the time series of the regional
it is possible that more diffuse network changes mediate tasks that BOLD signal, rather than its density across larger areas. Abnormal
contain a greater cognitive load. Thus, although the striatum may regional homogeneity may therefore reflect changes in the
be critical to the pathophysiology of MD, broader cortical– temporal aspects of neural activity in particular regions of interest.
subcortical network dysfunction may be necessary to impede In the first known application of this resting state fMRI technique
more complex cognitive performance tasks. In this regard, in MD (Yuan et al., 2008), 18 patients with remitted LoD and 14
evidence from other areas of cognitive neuroscience suggests that controls showed that those with LoD demonstrated an atypical
successful acquisition of implicit learning sequences may depend pattern of neural activation, characterized by decreased regional
initially on cortical and cerebellar involvement, while the basal homogeneity over the frontal, temporal and parietal lobes, and
ganglia may be more integral to the maintenance of learning (see increased homogeneity in the putamen, frontal and parietal lobes.
review by Forkstam and Petersson, 2005). An alternative explana- Of interest, regional homogeneity in fronto-subcortical areas
tion for the lack of striatal change in association with reduced (superior frontal gyrus and putamen) was negatively associated
implicit learning is that the topographical components of the with performance on both Parts A and B of the Trailmaking Test
striatum may need to be further detailed. That is, perhaps there are (reflecting visuomotor speed and mental flexibility/set-shifting) in
altered patterns of activation that are only evident in specific the depression sample, suggesting that this technique may further
components of the network. Moreover, since there is some aid in understanding the pathophysiology of executive function in
evidence to suggest that some striatal regions show reduced LLD. Using a somewhat different approach focusing on the caudate
activity during task completion, while others show increased as the ‘seed’ region of interest, Kenny et al. (2010) utilised resting
activity, a ‘cancelling out’ may occur when activity is measured state fMRI in a sample of 33 subjects, 16 of whom had a recent
122 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

Box 4. Functional neuroimaging studies. Box 5. Neuropathological studies.

 Have enabled more sophisticated modelling of MD patho-  Few studies exist and unlike studies in younger patients,
physiology and treatment mechanisms those with LLD show no specific glial density alterations in
 PET ligands have identified alterations in serotonergic bind- the ACC, DLPFC or OFC
ing in various regions including frontal, hippocampal, basal  For cortical regions, decreased density of pyramidal neurons
ganglia and brainstem regions has been found in the OFC, while reductions in pyramidal
 SPECT studies have generally reported decreased regional neuron size have been found in the DLPFC
cerebral blood flow in frontal, temporal and subcortical  For subcortical regions, decreased neuronal density is evi-
regions in LLD, where relationships between decreased rCBF dent in the caudate nucleus, and decreased glial density has
and some clinical features such as psychomotor speed have been found in the amygdala
emerged  Increases in hippocampal neuronal and glial density have
 fMRI data in LLD is relatively sparse but thus far indicates also been found
prefrontal and posterior circuitry changes in relation to  No known studies have examined the microglia specifically
cognitive and emotional tasks
 Resting state fMRI and MEG are emerging techniques for
investigating functional connectivity in LLD
 Variability in study designs, heterogeneity of samples and non-significant reductions have been reported in the entorhinal
data analyses should be considered
cortex (Bowley et al., 2002).
Two studies have reported a reduction in glial density and in the
glia/neuron ratio in the amygdala of patients with MD (Bowley
et al., 2002; Hamidi et al., 2004). Although neither study was
depressive episode. This study found that the connectivity specifically focused on LLD, all specimens came from subjects aged
between the caudate and frontal regions was more diverse in between 59 and 90 years (mean age of 77.7 years). Interestingly it
the depressed, compared to the control group (Box 4). appears that the changes in glial density in the amygdala are due to
reductions in the density of oligodendrocytes (Hamidi et al., 2004).
8. Neuropathology Oligodendrocytes are well known for their role in myelination;
satellite oligodendrocytes, a population located next to neuronal
Neuropathological studies, although few, provide a glimpse of cell bodies, appear to function in the metabolism of synaptic
the cellular changes that may underlie the metabolic and glutamate (D’Amelio et al., 1990), additionally they appear to be
structural changes noted so far (see Fig. 1b). However, for cortical sensitive to excess levels of glutamate as well as ischaemic injury
regions, it appears that such changes may differ according to age. (Dewar et al., 2003). Taken together with structural neuroimaging
While previous studies involving younger samples have shown a data suggesting shape alterations in the core nuclei of amygdalae
reduction in astrocyte and oligodendrocyte density in the ACC, OFC of subjects with LLD (Tamburo et al., 2009), this suggests that
and DLPFC (Cotter et al., 2005; Rajkowska et al., 1999), no glutamatergic toxicity may play a role in LLD pathophysiology.
significant differences in glial density in the DLPFC (Rajkowska It has been postulated that consideration of neuronal and glial
et al., 2005b) or OFC (Khundakar et al., 2009) have been found in cell pathology in MD must be considered dynamically. That is, glial
LLD. By contrast, LLD has been associated with a significantly pathology may be most prominent early in the disease, whereas
decreased packing density of pyramidal neurons in the OFC, neurons may become affected in later stages, possibly due to an
specifically cortical layers IIIc and V (Rajkowska et al., 2005a). excess in extracellular glutamate and glial dysfunction (Rajkowska
Although examination of the DLPFC has shown no significant and Miguel-Hidalgo, 2007). Indeed, a recent meta-analysis
differences between subjects with LLD and controls in the neuron supported this view using the glial marker protein S100B, found
density (Van Otterloo et al., 2009), morphometric analysis of the in serum. This study showed that S100B was more pronounced in
DLPFC has revealed reduced pyramidal neuronal size (Khundakar older subjects compared to younger subjects, though there was no
et al., 2009), particularly in cortical layer V. These changes were not relationship with duration of illness or age of onset (Schroeter
associated with any alterations in the glial population or non- et al., 2011). Overall, studies to-date have been somewhat limited
pyramidal neurons. In the DLPFC and OFC, cortical layers III and V in terms of the range of glial cells and processes they can examine
contain pyramidal neurons that project to the striatum. Accord- and no known studies have examined the microglia. Further
ingly, neuronal density has been reported to be decreased in the clinicopathological work in LLD is still required, and in particular
striatum in LLD, with specific reductions in the dorsolateral and needs to provide detailed characterisation of the clinical and
ventromedial aspects of the caudate nucleus. No differences were cognitive profile of patients. Such work may also help to elucidate
evident in those with EoD compared to LoD and the authors did not the unique contributions of illness-specific (e.g. inflammatory,
find alterations in glial density (Khundakar et al., 2011). glucocorticoid) processes, as well as those associated with
In the hippocampus, Stockmeier et al. (2004) reported a 35% concomitant cerebrovascular and emerging neurodegenerative
increase in both glial and neuronal density in the brains of 21 older disease and underlying changes in glial cell, particularly microglia
patients (mean age 57.4 years, range 30–87) with MD when functioning (Box 5).
compared to controls, with a significant decrease in neuronal soma
size. The authors suggested that the differential pattern in the 9. Genetics
hippocampus compared with other frontal structures may be due
to altered neuropil, including a loss of dendritic branching and glial Increasingly, there is recognition of the interactions between
processes. This may be related to the reduction in neurotrophic genetic variations and specific environmental factors that bias
factors implicated in the pathophysiology of depression. No known brain development towards illness vulnerability or resilience. A
studies have found any significant differences between late and number of genetic polymorphisms have been identified as having
early-onset depression groups with regard to neuronal pathology potential importance in the aetiology, management and outcome
(Khundakar et al., 2009; Rajkowska et al., 2005a), suggesting that of LLD (see Fig. 1a). To-date, these include genes that are
reductions in neuronal density may represent a final common considered to be important for neuronal plasticity and survival,
pathway for differing trajectories of illness. Smaller but serotonin transmission, lipid transport and systemic vascular risk.
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 123

However interpretation of the literature to-date is challenging due et al., 2008b). Such models are certainly immensely complex and
to the complexity of methodology, with issues in the measurement need to incorporate multi-faceted data points. In younger
of environment, the diagnosis of depression and study design, euthymic samples the met allele has been associated with smaller
among others (for review, see Lotrich, 2011). hippocampal volumes (Bueller et al., 2006; Pezawas et al., 2004),
and cognitive changes, particularly delayed memory (Egan et al.,
9.1. Brain-derived neurotrophic factor gene (BDNF) 2003). Two recent studies in a clinical population have produced
conflicting results. Benjamin et al. (2010), investigating a large
As previously mentioned, BDNF is the most abundant sample of patients with LLD, found no significant relationships
neurotrophin in the brain, and of critical importance for neuronal between BDNF genotype and hippocampal structure, memory or
plasticity and survival (Duman et al., 2006). BDNF also regulates working memory. In contrast, Kanellopoulos et al. (2011)
the mesolimbic dopaminergic pathway and nucleus accumbens demonstrated that depressed val/val homozygotes had larger
that are involved in the identification and response to emotional hippocampal volumes than non-depressed controls with the same
environmental stimuli (Berton et al., 2006). In non-geriatric genotype. This data highlights the need to consider the broader
samples there is increasing evidence to suggest that the BDNF gene–environment associations in large community and/or
genotype mediates the response to social stressors (Aguilera et al., genetically informed (e.g. twin) studies, whereby the interactions
2009). BDNF has been implicated in the pathogenesis of depres- between gene expression, risk and protective factors and complex
sion, and appears to be an important mediator of therapeutic illness trajectories can be examined. A recent study has attempted
response to antidepressants (Duman et al., 2006). The BDNF gene to unravel some of these connections; Gatt et al. (2009) used path
has been mapped to chromosome 11; a single nucleotide modelling in a non-clinical sample to investigate the relationship
polymorphism (G/A), resulting in an amino acid switch from between the BDNF val66met polymorphism and early life stress,
valine to methionine at position 66 of the coding exon of the BDNF and its effects on brain structure, cognition and the phenomenol-
gene (Maisonpierre et al., 1991). The BDNF val66met polymor- ogy of depression. In those with the met allele, increased exposure
phism (met allele) reduces the activity-dependant secretion of to early life stress resulted in reductions of the volume of
BDNF that is important for hippocampal based synaptic plasticity, hippocampal gray matter as well as its lateral prefrontal
learning and memory (see Egan et al., 2003). projections, higher levels of neuroticism and elevated body
In an older community dwelling cohort with no depression, arousal. In turn, reductions of hippocampal volume were
impairments in processing speed and delayed recall as well as associated with elevated levels of depressive symptoms, which
reduced performance on tests of general intelligence were were themselves associated with declines in working memory;
significantly associated with the met allele (Miyajima et al., similarly a pathway from elevated arousal to heightened neuroti-
2008). The met allele has been associated with increased risk of LLD cism to increased depression was demonstrated. Further longitu-
in both inpatient and outpatient samples, however no association dinal studies examining the effect of gene–environment
with severity of depression or age of onset has been demonstrated interactions on brain structure and function, as well as the role
(Hwang et al., 2006; Taylor et al., 2007), and not all studies have that social connection and support may play are now required.
replicated this result (Surtees et al., 2006). The met allele may be
associated with increased incidence of depression after stroke 9.2. Serotonin transporter gene (5HTTLPR)
(Kim et al., 2008), and is associated with significantly greater
WMLs in older adults, independent of diagnosis of depression Polymorphisms in the promoter region of the serotonin
(Taylor et al., 2008b). Additionally both serum and plasma transporter gene (5HTTLPR) may be important in LLD, as the gene
concentrations of BDNF have been demonstrated to be significant- product is critical for serotonergic neurotransmission, mainly via
ly lower in older adults with LoD compared with healthy age- uptake of serotonin. Polymorphisms of 5HTTLPR, which most
matched controls (Diniz et al., 2010b; Shi et al., 2010). The use of commonly include a 44 base pair insertion (L allele) or deletion (S
peripheral BDNF as a biomarker for affective disorders has many allele), affect gene expression rather than changing the structure of
methodological issues (Gass and Hellweg, 2010) including the serotonin transporter protein (Heils et al., 1995). The S allele is
whether peripheral BDNF actually provides an accurate represen- inefficient in transcribing the serotonin transporter in neurons
tation as to cerebral BDNF concentrations. However both animal (Heils et al., 1995), with the L allele associated with increased
and human studies have correlated BDNF serum levels with serotonin reuptake. Generally the literature suggests that the S
cortical BDNF expression (for review see Gass and Hellweg, 2010), genotype confers increased risk for depression in younger adults,
attesting to serum levels as an adequate proxy. via interactions with ‘‘stress’’ (Caspi et al., 2010, 2003; Kendler
The literature to-date regarding BDNF in LLD has been most et al., 2005b), although this relationship is by no means universal,
informative regarding treatment responsiveness. Specifically, the as evidenced by some large studies with negative findings (see
met allele has been found to be associated with increased odds of Risch et al., 2009). With regard to older adults, in a study
remission at six months in depressed elders (Taylor et al., 2010a); a examining those with LoD, no significant association was found
relationship that was noted to be independent of WMLs and between the S allele and depression in the whole sample. However
perceived social support. Similarly BDNF met carriers were more there did appear to be gender differences, with the S allele being
likely to achieve remission than val/val homozygotes after 12 associated with depression development in men, and recurrent
weeks of treatment with escitalopram (Alexopoulos et al., 2010). In depression in women (Steffens et al., 2002c). A prominent question
the same group, microstructural abnormalities in the corpus relates to the nature of the stressor in the relationship (Lotrich
callosum, left superior corona radiata and right inferior longitudi- et al., 2011). Much of the early work has found that the S/S
nal fasciculum were correlated with failure to achieve remission. genotype interacts with severe childhood stress and trauma to
However there was no significant interaction between val66met increase the risk of depression in adult life (Caspi et al., 2003). By
status and microstructural abnormalities in predicting remission. extension this increased risk then passes into old age as recurrent
It has been postulated that polymorphisms in the BDNF gene episodes of depression leading to an increased risk for LLD
impact on early cerebral development leading to permanent (Luijendijk et al., 2008). However the role of acute stressors is less
changes in cellular development and plasticity (Egan et al., 2003), clear. Increased medical burden in older adults, considered a
thus placing carriers preferentially at risk of depression in the specific stressor, appears to interact with the S/S genotype to
context of further neurobiological or psychosocial risks (Taylor increase the risk for depression (Kim et al., 2009) whereas the
124 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

relationship between psychosocial stress and 5HTTLPR polymor- direct association between the S allele and hippocampal volume in
phisms is less obvious (Surtees et al., 2006). It has been suggested a healthy sample of older adults, there was a significant interaction
that individuals with the S/S genotype are more sensitive to the between waking cortisol levels and the S allele on reduced
depressogenic effects of acute stressful life events (e.g. divorce/ hippocampal volume and delayed memory function (O’Hara and
separation, major financial problems, serious illness/injury, job Hallmayer, 2007). At somewhat of a tangent to this work,
loss) because they have an increased sensitivity to the impact of amygdala hyperactivation to negative stimuli may also be
mild stressors (Kendler et al., 2005a). heightened in the presence of the S allele (Munafo et al., 2008).
CVD, a common form of stress in older adults, is associated with Moreover the S/S genotype has been associated with reduced gray
somewhat contradictory findings. The L/L genotype has been matter in the amygdala and perigenual cingulate (Pezawas et al.,
associated with greater platelet activation, which is thought to 2005). Given the amygdala’s role in modulating glucocorticoid
increase the risk of thrombus development, as well as increased release (Savitz and Drevets, 2009), this body of work highlights the
depression in older adults with a low cardiovascular burden potential importance of the HPA axis, mediating the interaction of
(Whyte et al., 2001). Moreover the L/L genotype has been stress and 5HTTLPR polymorphisms on risk for depression.
demonstrated to be associated with increased cardiac events While few studies have examined the potential prognostic
post coronary artery bypass grafting (Phillips-Bute et al., 2008). relevance of 5HTTLPR polymorphisms, there is some data
Thus the presence of the L allele appears to increase the risk for suggesting that, together with white matter change, presence of
exposure to adverse cardiovascular events, thus negating its the S allele may be linked to remission. More specifically, lower
‘‘positive’’ effect (Lotrich et al., 2011). However there are remission rates after treatment with SSRI antidepressants were
conflicting results, as the S allele has also been related to seen in those with the S/S genotype (Alexopoulos et al., 2009). This
increased risk for further vascular disease post myocardial data is consistent with earlier work by the same group, which had
infarction, a relationship that appears to be mediated in part by shown that presence of the S allele was associated with poorer
depressive symptoms (Nakatani et al., 2005). Further complicat- response to the noradrenergic and specific serotonergic antide-
ing the literature regarding the relationship between the S and L pressant, mirtazapine; possibly related to poor tolerability rather
alleles and vascular disease is the finding that heterozygosity (i.e. than reduced efficacy of this agent (Murphy et al., 2004).
the S/L genotype) was associated with increased risk for higher
cholesterol levels, as well as heart disease in healthy older male 9.3. Apolipoprotein E gene (Apoe)
samples (Comings et al., 1999).
Of relevance to the pathophysiological link between the The apolipoprotein E gene (Apoe) encodes a lipid transport
serotonin transporter gene and neurobiological changes in LLD, protein that is involved in the maintenance and repair of neuronal
a number of studies have investigated the relationship between cells (Yuan et al., 2010). There are three alleles: e2, e3 and e4, with
5HTTLPR polymorphisms, structural brain changes and risk for data suggesting that the e4 allele is less efficient. A considerable
cerebrovascular disease. With regard to structural brain changes in body of literature has indicated that the e4 allele is a risk factor for
depression, some studies have shown that the 5HTTLPR genotype AD, with about 65% of individuals with non-related sporadic AD
relates to hippocampal volumes although there have been some and up to 80% of familial AD sufferers carrying this allele,
inconsistencies, which may possibly relate to the aetiological path compared with only 20% in healthy control populations (Farrer
to depression including age of illness onset. Data from adult MD et al., 1997). Despite clear evidence of an association, the
samples has shown that L/L homozygotes with depression have mechanism for the association between e4 and AD remains
smaller hippocampal volumes than L/L control participants (Frodl unclear, although it may be mediated by the role played by e4 in
et al., 2004). Within depressed samples, hippocampal volumes the redistribution of lipids between cells in the central nervous
have also been found to be smaller in L/L, compared to S/S system (CNS), which are important in neuronal repair and
homozygotes (Frodl et al., 2008), although this has not been a maintenance of synaptic-dendritic connections (Mahley et al.,
consistent finding (Hickie et al., 2007b). Upon specifically 2006).
examining these relationships in LLD, Taylor et al. (2005b) Studies that have sought to examine whether the e4 allele is
reported that among L/L homozygotes, those with LoD had relevant to the pathogenesis of LLD have generally produced
significantly smaller right hippocampal volumes than those with inconsistent results. For instance, some data has suggested that the
EoD and controls. Hickie et al. (2007b) reported that in a sample of e4 allele is more common in individuals suffering from LoD as
47 older patients with moderate to severe depression, the presence compared with EoD (Krishnan et al., 1996). However these results
of at least one S allele was associated with smaller caudate nucleus have not been replicated in more recent research (Hickie et al.,
volumes. However, there was no relationship between the S allele 2001a; Steffens et al., 2003).
and aetiological factors such as age of onset, summed VRFs, Other investigations have focused on the link between the e4
duration of depression episode or years of illness. Given the data allele and volumetric changes in LLD; these have primarily focused
highlighting the links between 5HTTLPR polymorphisms and on the hippocampus, alterations of which are implicated very early
vascular disease, the relationships with white matter changes in in the course of AD. The e4 allele has been associated with
LLD are important. Alexopoulos et al. (2009) reported that reductions of hippocampal volume in some (Kim et al., 2002), but
depressed S allele carriers had lower DTI-measured FA (that is, not all (Hickie et al., 2005b) studies. It has been suggested that the
microstructural white matter change) than L allele homozygotes in e4 allele may mediate hippocampal shape alterations in the region
fronto-limbic brain areas; and, extending findings related to of the CA1 subfield (Qiu et al., 2009). A small longitudinal study in
heterozygosity, the S/L genotype has been associated with depressed, non-demented elders (n = 45) found that the e4 allele
increased total and WML volumes in a sample of older adults was associated with more rapid hippocampal volume loss on the
with depression (Steffens et al., 2008). right over two years (Kim et al., 2002). Other studies examining
The possible role of the HPA axis underpinning the relationship more extensive brain regions in 37 patients with remitted LoD
between stress, S/S genotype and depression is suggested by some found that in comparison to non e4 carriers, those with an e4 allele
studies in non-clinical samples. Alexander et al. (2009) showed had smaller volumes of the medial and middle frontal gyrus and
that the L/S genotype was associated with HPA axis hyperreactivity left inferior occipital gyrus although there were no differences in
in a stressful situation, when the individual had previously been neuropsychological performance between these groups (Yuan
exposed to multiple stressful life events. Although there was no et al., 2010). The lack of relationship between Apoe4 and cognitive
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 125

decline in older patients with MD replicates earlier work (Hickie


Box 6. Genetic research in late-life depression.
et al., 2001a; Naismith et al., 2003). However more recently,
depressed e4 carriers were shown to have greater cognitive decline  The serotonin transporter gene may be implicated in depres-
than those without the e4 allele (Corsentino et al., 2009; Niti et al., sion onset, basal ganglia changes, vascular risk, WMLs and
2009). These findings have been recently extended to show that treatment responsiveness
the e4 allele moderates the relationship between hippocampal  The Apoe4 allele may be linked to hippocampal volume loss
volume and cognitive decline, such that smaller hippocampal and later depression onsets
volumes in depressed e4 allele carriers, have greater cognitive  BDNF polymorphisms may be associated with risk for LLD
and response to treatment responsiveness
decline (Sachs-Ericsson et al., 2011).
 The MTHFR gene mutation is integral to homocysteine
Although the precise mechanism by which the Apoe4 genotype metabolism, relates to cardiovascular risk and has been
may be pertinent to the structural brain changes in LLD is associated with later ages of depression onset and slowed
unknown, there is some data suggesting that the expression of psychomotor speed
Apoe4 may relate to cerebrovascular disease. Replicating a
suggested link between cerebrovascular disease and Apoe
genotype (Nebes et al., 2001), Steffens et al. (2003) found a
significant association between the e4 allele and the volume of More recent studies have begun to focus on other genotypes
gray matter hyperintensities in elderly depressed patients. that are relevant to cardiovascular health. Of these, genes that are
Despite these structural associations, however, the e4 allele seemingly integral to blood pressure regulation have been of
was not found to be associated with the development of dementia interest including the renin-angiotensin system genes, angiotensin
in a longitudinal study of depressed older adults (Steffens et al., II receptor vascular types I and II (AGTR1 and AGTR2). Early studies
2007). Additionally, the Apoe4 allele does not appear to relate to in this area have suggested that there may indeed be a link with
concomitant cerebrovascular disease in LoD (Traykov et al., 2007), WMLs, particularly in progression of WMLs in men (Taylor et al.,
suggesting it plays a unique and differential role in cognitive 2010b), as well as with LLD outcomes (Kondo et al., 2007) (Box 6).
decline.
As has been observed with the serotonin transporter gene, some 10. The role of inflammation
data suggests that Apoe genotype may play a role in antidepressant
response in LLD. A double-blind 8-week randomised controlled Despite the multitude of evidence relating to structural and
trial involving 246 patients over the age of 65 years with MD, metabolic abnormalities in LLD, the pathophysiological mecha-
examined the effect of Apoe genotype on response to mirtazapine nisms underpinning such changes are poorly understood. Recently,
or paroxetine. Interestingly, those patients with the e4 allele prolonged and exaggerated immune responses have been postu-
showed a rapid onset of mirtazapine action whereas those on lated to promote such changes in neurobiological circuitry that
paroxetine were slow to respond (Murphy et al., 2003). predispose to LLD or facilitate metabolic changes that mediate
depression (Alexopoulos and Morimoto, 2011). The role of the
9.4. Genes implicated in systemic vascular risk inflammatory system and immune activation in the aetiology of
MD was suggested well over a decade ago (Hickie et al., 1990b;
In light of the considerable wealth of data linking LLD, and Maes, 1995). There is now ample evidence demonstrating a
more specifically LoD, with cerebrovascular disease, there has relationship between pro-inflammatory cytokines, such as the
been relevant interest in those genes that appear to predict interleukins and interferons and MD specifically (Dantzer et al.,
systemic vascular risk. One that has received attention is the 2008; Thomas et al., 2005). Commonly studied cytokines include
MTHFR gene, which encodes the enzyme 5-methylenetetrahy- interleukin (IL)-1, IL-6, tumour necrosis factor alpha (TNFa) and
drofolate reductase. This enzyme is associated with the metabo- CRP. These intracellular signalling polypeptides, produced by
lism of homocysteine, utilising B12 and folic acid as co-factors. A macrophages, monocytes and other immune activated cells, play a
relatively common polymorphism (C677T) produces an enzyme central role in the regulation of the immune response. Increased
with reduced activity leading to increased homocysteine levels. A levels of pro-inflammatory cytokines have been found in older
large number of studies have linked the C677T mutation and/or adults (Krabbe et al., 2004); such an age-related ‘‘pro-inflamma-
homocysteine levels with cardiovascular and cerebrovascular tory state’’ has been suggested to be associated in part with high
disease as well as to WMLs. Studies examining the presence of this levels of VRFs and morbidity (Ferrucci et al., 2005).
genotype in LLD have noted an increased prevalence in compari- Although holistic examination of the role of immunological
son to controls (Almeida et al., 2008) and have noted it to be and/or inflammatory responses cannot be tested entirely, the
associated with later ages of onset (Hickie et al., 2001a). Some results of studies to-date investigating inflammatory markers in
neuropsychological data has also linked the C677T genotype with LLD are less consistent than in younger or mixed populations. After
slowed processing speed in older depressed samples (Naismith adjusting for common confounders, one study (Penninx et al.,
et al., 2002, 2003). However, a recent analysis of data from the 2003) found CRP to be elevated in depression. This cross-sectional
NCODE study found no association between MTHFR genotype and community study also demonstrated raised IL-6 and TNFa levels to
neurocognitive performance (specifically MMSE, and tests of be associated with depressed mood; moreover if two or all three
processing speed, delayed recall and executive function) in older markers were elevated, the odds ratio of MD was 2.45 compared
adults with MD (Hong et al., 2009). Despite this, an interaction with those with no elevation in markers. However a number of
between age and genotype was discovered, such that the TT other studies did not find any association with raised levels of CRP
genotype was associated with increased WML volume compared and depression in older adults (Almeida et al., 2007; Bremmer
with the CC genotype for a given age. Since homocysteine levels et al., 2008; Forti et al., 2010; Kop et al., 2002; Pan et al., 2008;
may be particularly linked to WMLs even in those with small Steptoe et al., 2003; Thomas et al., 2005; Tiemeier et al., 2003).
vessel disease (Wong et al., 2006), the influence of this genotype Interestingly, in a large cross-sectional community study of men
may be particularly relevant to processes mediated by subcortical aged over 70 years, raised levels of CRP were not associated with
structures (Naismith et al., 2002), which are in turn especially clinical significant depressive symptoms once confounders had
vulnerable to ischaemia (Kumral et al., 1999; Steffens et al., been adjusted for, but the risk of depression was increased in men
1999b). carrying a genetic polymorphism resulting in lower serum
126 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

concentrations of CRP (Almeida et al., 2009). Four studies reported particularly caregiving burden, is associated with depression in
elevation of IL-6 in depressed subjects (Bremmer et al., 2008; older adults (Marin et al., 2011). Furthermore it appears that such
Dentino et al., 1999; Penninx et al., 2003; Tiemeier et al., 2003); depressed and stressed older adults may then be primed to
with another four studies not finding any significant difference in respond to undergo an exaggerated inflammatory response to
the levels of IL-6 between depressed and controls subjects further stressors and challenges (Gouin et al., 2008).
(Empana et al., 2005; Forti et al., 2010; Pan et al., 2008; Steptoe Despite the associations between stress and VRFs and increas-
et al., 2003). ing levels of peripherally circulating pro-inflammatory factors, it is
Only two studies have investigated the association between not clear whether peripheral elevations in inflammatory factors
depression and IL-1b, both reported an increase in the depressed are positively associated with cerebral levels, or how raised
group, independent of confounders (Diniz et al., 2010a; Thomas cytokine levels contribute to depressed mood. Although the blood
et al., 2005). These two studies compared IL-1b levels in EoD and brain barrier is relatively impermeable to cytokines, saturable
LoD. In the study by Diniz et al. (2010a), the mean IL-1b level was transport mechanisms exist that allow limited penetration of IL-1b
raised in the EoD group compared with the LoD group, but the and TNFa (Banks et al., 1991). Additionally, peripherally circulat-
difference only approached significance. One explanation for ing cytokines may influence cerebral processes via the vagus nerve
higher levels in EoD group may be ‘‘a cumulative increase in (Maier and Watkins, 1998), and pro-inflammatory cytokines may
inflammatory response’’ related to the increased rates of recur- be able to enhance permeability at vascular sites (de Vries et al.,
rence seen in this group (i.e. mean of 3.0 episodes in EoD compared 1996). Recent findings suggest that peripheral cytokine elevations
to 1.1 in LoD) (Diniz et al., 2010a). Thomas et al. (2005) did not are reflected in the brain; post-mortem microarray mRNA
report any difference in IL-1b between their EoD and LoD subjects, expression analysis, carried out in the BA10 region of brains from
or any correlation between IL-1b and age of depression onset. individuals with melancholic depression as well as controls,
These contrasting findings suggest that further research is required demonstrated altered expression of a number of genes, particularly
in order to delineate the contribution of inflammatory changes in up-regulation of the expression of pro- and anti-inflammatory
those disorders emerging later in life. cytokines (Shelton et al., 2011). The CNS immune system therefore,
Although cross-sectional approaches have failed to yield although protected in some degree by the blood brain barrier, has
consistent results, two longitudinal studies have demonstrated some link to the periphery. Microglia, the primary immune defence
an association between the development of depression in cells in the CNS (Allen and Barres, 2009), exist in both quiescent
community dwelling older adults and pro-inflammatory markers. and primed states (Kreutzberg, 1996). Primed microglia may be
In a population-based cohort of 85 years old, participants who did rapidly activated by stimulation with an antigen, and secrete
not report depressive symptoms at baseline had cytokine levels cytokines and growth factors, as well as stimulate an activation
determined by ex vivo whole blood stimulation with bacterial cascade, that allows recruitment of other glial cells, particularly
lipopolysaccharide (LPS) (this method reflects the capacity of the astrocytes, and secretion of more cytokines and neurotrophic
innate immune system) (van den Biggelaar et al., 2007). High factors to bring about repair and facilitate homeostasis. As
circulating levels of CRP at baseline predicted accelerated increase mentioned above, the age-related pro-inflammatory state is
of depressive symptoms; additionally higher IL-1b and lower IL- associated with increased numbers of activated microglia as well
1ra production after LPS challenge was associated with the as increased production of pro-inflammatory cytokines (Sparkman
development of self-reported depressive symptoms over a 5-year and Johnson, 2008).
period. On the other hand, in a population-based cohort of older The role of glia in the neurobiology of depression is increasingly
adults asymptomatic at baseline, only levels of IL-1ra predicted the a focus of research (see review by Paradise et al., in press).
development of significant depressive symptomatology 6 years Increases in glial fibrillary acidic protein (GFAP), an astrocytic
later (Milaneschi et al., 2009). Those in the two highest quartiles of marker, have been discovered in cortical layer one of the DLPFC in
IL-1ra at baseline had a 2.3- and 2.8-fold increased risk of persons with LLD (Davis et al., 2002). Furthermore in younger
depression compared with those in the lowest two quartiles. IL-1ra people with depression, levels of GFAP in the PFC were significantly
is an antagonist of IL-1ra and IL-1b, and represents a marker of lower compared to controls (Si et al., 2004), whilst there was no
immune activation. It is produced in large quantities in the liver significant difference in the level of GFAP between depressed and
under the same circumstances that lead to the production of IL-1ra control subjects older than 60 years at time of death. One
and IL-1b, and may be considered a more reliable indicator of explanation for these findings may be that early in the course of
inflammation than IL-1, as it remains in the circulation for longer depression an imbalance in toxic (glucocorticoid/glutamate) and
periods (Granowitz et al., 1991). The contradictory findings of both trophic factors may lead to a reduction in glia, with resultant
reduced IL-1ra production after LPS challenge and increased neuronal damage and injury. Compensatory mechanisms later in
circulating IL-1ra levels being associated with increased risk of the course of depression then lead to gliosis and increased glial
depression may result from the differing methods of cytokine density (Rajkowska, 2000), with levels becoming comparable with
measurement, and suggests that on one hand risk for depression older subjects without a history of depression. Alternatively,
may depend on the ability to produce IL-1ra to an immune differences in GFAP expression may result from differing
challenge, conversely, high circulating levels of IL-1ra indicate a underlying pathophysiologies in EoD and LoD. Neuropathological
state of immune activation, which increases depression risk. In any studies investigating the pathophysiology of WMLs have demon-
case it would appear that the IL-1 signalling network has a role in strated evidence of ischaemia in the white matter of the DLPFC but
the development of depressive symptoms in older adults. not the ACC or occipital cortex (Thomas et al., 2003), and that all
Whilst the accumulation of VRFs as well as other inflammatory deep WMLs in a depressed group of adults with LLD were
conditions may contribute to increasing levels of pro-inflammato- ischaemic in origin as compared with only one-third of WMLs in a
ry cytokines, stress, particularly psychological stress, is likely to be healthy aged matched control group (Thomas et al., 2002).
important. In fact increasing inflammatory markers may be a key Moreover, compared with healthy age-matched controls, ischae-
mechanism in the development of depression in the face of mic lesions in depressed elders were significantly more likely in
increased stress. In a longitudinal community study, chronically the DLPFC than the ACC. Additionally up-regulation of the
stressed older caregivers were demonstrated to have a four-fold intracellular adhesion molecule-1, a vascular marker of inflamma-
increased rate of increase of IL-6 levels compared to non-stressed tion, in the grey and white matter of the DLPFC in subjects with LLD
older adults (Kiecolt-Glaser et al., 2003). Chronic stress, and more (Thomas et al., 2000), suggests that cerebral ischaemia may induce
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 127

inflammatory changes. This is partially supported by the GFAP


Box 7. Inflammatory research in late-life depression.
findings in layer one of the DLPFC described above. Finally
microglial activation has been demonstrated in periventricular and  Cross-sectional studies yield inconsistent results regarding
deep subcortical WMLs in ageing brains (Simpson et al., 2007), the association between CRP, IL-6 and LLD
with the response associated with periventricular lesions in  Some data linking IL-1b with LLD, with no clear link to LoD
particular, possibly reflecting immune activation related to specifically
disruption to the blood brain barrier. Taken together, these  Longitudinal data shows baseline IL-1ra and IL-1ra predicts
findings suggest a mechanism by which vascular risk increases the increase of depressive symptoms over time, with mixed
results for CRP and IL-1b
risk for LLD. Cerebrovascular disease, represented by WMLs, may
 Glial fibrillary acidic protein may be increased in the DLPFC
lead to localized microglial activation and the production of and may be indicative of altered astrocytic activity
inflammatory cytokines, thus priming the individual’s innate  Intracellular adhesion molecule-1 may be up-regulated and
immune response, which then responds in an exaggerated fashion may be indicative of vascular inflammatory activity
to future stressors – how this then culminates in the clinical  Altered functioning of the HPA axis is supported by data
picture of depression remains unclear. Potential explanations showing U-shaped curves for hypercortisolaemia and hypo-
include altered tryptophan metabolism, leading to reduced cortisolaemia
serotonin synthesis, via the activation of indoleamine 2,3-
dioxygenase by pro-inflammatory enzymes (Dantzer et al.,
2008), and excess glutamate, leading to excitotoxicity, due to Overall, the inflammation hypothesis of MD requires further
inflammatory mediator associated impaired expression of excit- empirical examination. However, a convergence of data does
atory amino acid transporter on astrocytes (McNally et al., 2008). support some role for immunological changes in the pathogen-
Once delivered in the brain or alternatively once local esis of this disease, warranting efforts devoted to further
production is increased, pro-inflammatory cytokines exert their understanding the interaction between such factors and medical
effects in a number of ways. Animal studies have demonstrated comorbidity, ageing, and microglial responses, and the emer-
that cytokines potently activate the HPA axis (Dantzer et al., gence and/or perpetuation of depressive disorders. As recently
2008), via inducing glucocorticoid receptor resistance thus highlighted by Alexopoulos and Morimoto (2011), this model
reducing the ability of glucocorticoids to down-regulate cortico- certainly leads to testable hypotheses and data regarding the
trophin releasing hormone, and resulting in a feed-forward mechanisms by which inflammatory responses may promote
cascade in cortisol production. Moreover the induction of depressive symptoms. In turn, this may allow greater opportu-
glucocorticoid receptor resistance in circulating immune cells nities for specific interventions targeting the underlying
would lead to failure of the inhibitory effect of glucocorticoids on pathophysiology, and may even be suited for early intervention
cytokine production and action, with a consequent intensified approaches (Box 7).
pro-inflammatory state. Although studies in younger adults have
frequently reported hyperactivity of the HPA axis as well as high 11. Prognosis in late-life depression
cortisol levels (Holsboer, 2000), there are relatively few studies
examining the role of the HPA axis in LLD. Generally these studies The prognosis of LLD is typically poor and is characterized by
have demonstrated HPA axis hyperactivity and high cortisol chronicity, mortality and increased risk for cognitive impairment
levels in depressed older adults (O’Brien et al., 2004), with and progression to dementia (Djernes et al., 2011; Hickie et al.,
cortisol levels returning to normal with resolution of the 1997b; Reynolds et al., 2008; Schoevers et al., 2009). In a three year
depressive episode. However there have been contradictory cohort study, LoD was found to be associated with a high rate of
findings of hypocortisolaemia in frail elderly with LLD (Morrison mortality and dementia. VRFs, including lower HDL, raised
et al., 2000), as well as those with chronic and recurrent erythrocyte sedimentation rate (ESR) and a higher score on the
depression (Oldehinkel et al., 2001). More recent research has Hachinski Index (a measure of ischaemic disease), were associated
demonstrated a U-shaped association between depression and with worse prognosis in this group (Baldwin et al., 2006).
cortisol levels in older adults with LLD. Two population-based Eleven longitudinal studies have investigated the prognosis of
studies of adults aged 65 and over have both found that LLD LLD as related to the age at onset of first episode. Of these, six
symptoms are associated with both hyperactivity and hypoac- found a difference in prognosis, in either direction when the
tivity of the HPA axis (Bremmer et al., 2007; Penninx et al., 2007). cohort was either divided into EoD and LoD (four studies) or age of
Bremmer et al. (2007) found in a European population-based onset was analysed as a continuous variable. Two studies reported
study that plasma cortisol levels were associated with MD in a U- LoD to have a worse prognosis: Alexopoulos et al. (1996) found
shaped fashion. Hypocortisolaemic depression was associated those with LoD had less chance of recovery at two years and, with
with being female, recurrent depression, smoking and joint regard to short-term prognosis, Conwell et al. (1989) found
disease, whilst hypercortisolaemic depression was associated subjects with LoD had a longer hospital stay and worse symptoms
with older age, male sex, CVD and cognitive impairment at discharge. In contrast, four studies found those with EoD had a
(although this last association was only of borderline signifi- worse prognosis: Reynolds et al. (1998) found that those with EoD
cance) (Bremmer et al., 2007). A community study, conducted in took longer to achieve remission although no overall differences
Italy investigated the relationship between urinary cortisol and were found between groups in rates of remission, relapse,
depressive symptoms and had similar findings: individuals at recovery or recurrence; Dew et al. (1997) reported earlier onset
each end of the cortisol spectrum reported higher depressive of depressive disorder (as a continuous measure) predicted a
symptoms (Penninx et al., 2007). Moreover hypocortisolaemic poorer treatment response and Brodaty et al. (1993) found EoD to
depression was associated with relatively lower depressive have a worse prognosis in the elderly (measured at one year and
severity but increased physical frailty. Neither study looked two to four year follow-up periods, using a categorical ‘global
specifically at the age at onset of depression; however the outcome’ scale). In the earliest study, Kay et al. (1955) also
association with chronicity and hypocortisolaemia on the one reported EoD had a better prognosis in several domains, although
hand and cardiovascular risk and hypercortisolaemia on the these latter findings are limited by the lack of valid diagnostic
other, further highlights the likelihood that LoD and EoD are criteria and omission of statistical tests to confirm differences
underpinned by differential mechanisms. between groups.
128 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

The effect of age of onset of depression on prognosis and with the first treatment (Roose and Schatzberg, 2005), warranting
treatment response is therefore uncertain. Age may have a the need to consider alternative non-pharmacological options in
complex relationship with depression and is highly correlated some sub-groups. A recent systematic review of treatment for
with a number of other known risk factors known to affect refractory depression in older adults found that lithium was the
prognosis. Elderly depressed patients with an early first onset of only treatment with consistent evidence for an additional effect
depression are more likely to have had a greater lifetime number of (Cooper et al., 2011). This is indicative of the lack of focus on this
relapses and an overall longer duration of illness; both of these key patient group. Traditionally, other physical treatments,
factors are important predictors of poor prognosis. Alternatively, notably ECT, have been preferentially used in older subjects with
patients presenting with depression for the first time in later-life severe and treatment-resistant depression. Such patients more
have an increased risk of physical comorbidities (Lavretsky et al., often present with psychotic features or disturbance of psycho-
1998; Subramaniam and Mitchell, 2005; Tupler et al., 2002) and motor function (psychomotor retardation or agitation) that do
physical comorbidity has been associated with a worse prognosis predict a comparatively better response to ECT (Hickie et al.,
(Iosifescu et al., 2003). Complicating the clinical picture is the 1990a). Interestingly, ECT has often been suggested as an
possibility of emerging neurodegenerative disease in some adjunctive treatment for other neurological and neurodegenera-
patients, with prevalence rates for AD alone doubling every five tive disorders including Parkinson’s disease and delirium (see
years after the age of 60 (Lobo et al., 2000; National Institute on Popeo and Kellner, 2009). New direct brain stimulation models for
Aging, 1999). If VRFs are untreated, depressive symptoms may treatment-resistant depression (Mayberg et al., 2005) have been
become more chronic and cognitive decline may also continue. directly modelled on the success of similar interventions for
Therefore the effect of age of onset on prognosis may be partially patients with Parkinson’s disease.
mediated by the presence of early illness, physical comorbidities, Other data is largely limited to small non-randomised trials or
untreated VRFs and emerging incipient neurodegenerative disease. case studies. Nimodipine (a calcium channel blocker) augmenta-
tion in those with ‘vascular’ depression has demonstrated
12. Treatment approaches encouraging results (Taragano et al., 2005), as has Aripiprazole
augmentation in SSRI non-remitters with LLD (Rutherford et al.,
12.1. Pharmacological treatments 2007). A trial of Galantamine, a cholinesterase inhibitor, augmen-
tation produced negative results (Holtzheimer et al., 2008).
While there have been a very large number of studies Methylphenidate has been used in older adults and medically ill
investigating response to pharmacological treatments in middle- patients for many years, despite no clear evidence of efficacy. A
aged persons, there is still a remarkably small number of studies recent review of clinical trials of methylphenidate in older adults
specifically focused on assessing interventions in older persons, found that the data to support its use is of relatively poor quality,
particularly those with LoD. Increasingly over the last two decades, but suggests that it may have efficacy for depressive symptoms,
as much larger studies have moved to evaluate populations outside fatigue, apathy and cognitive slowing in a variety of medically ill
specialist settings the differential between active treatments and populations (Hardy, 2009). Other agents that may show promise in
placebo response has declined. It is assumed that this has resulted LLD are those that concurrently target the sleep–wake system,
from the movement away from assessment of interventions whilst also possessing low side-effect profiles. In this regard, the
among a more severely ill group (who best fit a more traditional novel antidepressant Agomelatine, may show some promise for its
‘neurobiological’ model) to much more heterogeneous (and less capacity to concurrently target melatonergic receptors (Hickie and
persistently-ill) populations. The same phenomena may well Rogers, 2011). However, no known studies to-date have examined
emerge among studies of older persons with depression, as both the efficacy of this agent for LLD.
preventive/early intervention studies (Christensen et al., 2011; Extensive research now suggests that the presence of VRFs,
Cockayne et al., 2011; Walker et al., 2010) and broader communi- small vessel vascular disease or cognitive dysfunction is a strong
ty-based studies are now underway. predictor of impaired response to SSRI treatments, highlighting the
Although drawing on the current data base, pharmacological relevance of these variables for management plans (Sheline et al.,
treatments appear to be effective for older persons with MD (Roose 2010). In general, WMLs, VRFs, executive dysfunction, depression
and Schatzberg, 2005), with a Cochrane meta-analysis (Wilson recurrence and later ages of illness onset are associated with
et al., 2001) of 17 studies demonstrating efficacy of SSRIs, TCAs and particularly poor treatment outcomes (Alexopoulos et al., 2008;
monoamine inhibitors (MAOIs) over placebo in the treatment of Hickie et al., 1997b; Potter and Steffens, 2007; Sheline et al., 2010).
LLD; more recent data suggests response rates may be little better However, some evidence suggests that better outcomes can be
than placebo (Nelson et al., 2008). Several comparator or ‘head-to- achieved if treatment trials are prolonged or more complex
head’ studies of different classes of antidepressant including SSRIs (Anstey and Brodaty, 1995; Driscoll et al., 2005). Unfortunately,
to TCAs (Bondareff et al., 2000) and mirtazapine (a noradrenergic little attention has been paid to the role of other neurotransmitter
and specific serotonergic antidepressant) to SSRI (Schatzberg et al., systems in LLD that may offer viable options for treatment targets,
2002) have failed to consistently demonstrate the superior efficacy and which may be better tailored to the underlying neurobiology
of one class of antidepressants over another, although different of the disease. A previous emphasis has been placed on
tolerability and side-effect profiles are seen between classes dopaminergic systems, particularly as it related to relief of anergia
(Mottram et al., 2006). Moreover, a recent large-scale general and psychomotor retardation (Brown and Gershon, 1993). Hence,
practice-based review of treatment of depression in older persons the older literature emphasizes the use of stimulant medications
(Coupland et al., 2011; Hickie, 2011) indicates the extent to which and antidepressants with evidence of increased dopaminergic
widespread prescribing of SSRIs are associated with important activity (e.g. Antleman and Chiodo, 1981). In more recent times
side-effects (e.g. falls, hyponatraemia, agitation), and the extent to agents acting on glutamatergic or GABA systems have been
which some classes may be associated with more significant risks proposed (see review by Javitt et al., 2011). However, more human
including increased risk to stroke, seizures, fracture and premature data regarding the pathophysiological roles of these various
death. systems in depressive disorders is required (possibly through
Compared to middle-aged patients, older persons may take magnetic resonance spectroscopy and/or neuropathological stud-
longer to respond to antidepressant medication, and unfortunately ies). The additional role of agents that focus on specific cognitive
as many as 50% of patients with LLD will not achieve remission features, including memory dysfunction (e.g. acetylcholine)
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 129

requires systematic evaluation in cohorts with such features. In 12.2. Non-pharmacological treatments
younger samples, there is certainly preliminary evidence suggest-
ing that agents targeting glutamate, GABA and dopamine may Non-pharmacological approaches for treatment of depression
demonstrate antidepressant efficacy (Berman et al., 2000; Zarate and cognition in LLD have been limited. This may reflect previous
et al., 2004), thereby warranting further evaluation in older, albeit consensus statements implying that psychotherapeutic interven-
more complex samples. tions may be less efficacious in older adults (NIH Consensus
Electroconvulsive therapy is an effective option in those with Development Panel on Depression in Late Life, 1992) or the more
psychomotor retardation, psychotic phenomena or severe disease general practice bias against providing psychological interventions
(Hickie et al., 1996). However, MRI-associated white or grey nuclei to older persons with depression (Hickie et al., 2001b). However a
changes predict a poorer response (Hickie et al., 1997b). Other recent meta-analysis of psychotherapy and pharmacotherapy
clinical predictors of poor response include anxiety, hopelessness, trials in LLD demonstrated comparable effect sizes for both forms
limitations in physical function, chronicity of current episode, of treatment (Pinquart et al., 2006) and expert guidelines have
social isolation, and poverty (Alexopoulos et al., 2008). Repetitive generally recommended the use of both forms of treatment (see
transcranial magnetic stimulation (rTMS) is a low risk, non- review by Alexopoulos, 2011). Moreover, for elderly patients
invasive approach that involves the stimulation of specific brain suffering minor depression and dysthymia, psychotherapeutic
regions via the passage of magnetic pulses through the skull, and interventions displayed larger effects with regards to clinician-
does not involve a general anaesthetic or seizure. A recent review rated depression than pharmacological strategies. Despite data
on the use of rTMS in MD found that although rTMS is more suggesting that antidepressants may demonstrate little superiority
effective than sham treatment for the treatment of depression, it over placebo (as reviewed above), as well as evidence that
may be less effective than other conventional antidepressant psychological therapy has similar efficacy to medication in mild to
therapies (Loo and Mitchell, 2005). The literature is less clear on moderate depression (McCusker et al., 1998) in older people,
the benefits of rTMS in LLD. Two small controlled studies in elderly psychotherapeutic interventions are less frequently offered to this
patients reported that rTMS produced no additional antidepres- age group (Baldwin and Wild, 2004). In fact, a combination of
sant effects when compared with sham treatment (Mosimann pharmacological and non-pharmacological strategies may have
et al., 2004), and did not lead to improvement in depression optimal benefits, as evidenced by a study comparing monotherapy
severity (Manes et al., 2001). However a case series of 49 older Cognitive Behavioural Therapy (CBT), desipramine (a TCA), or a
adults who received high frequency rTMS delivered to the left combination of the two. Although all three groups showed
DLPFC as an adjuvant to pharmacotherapy, reported a modest but improvement, the combination group demonstrated the greatest
statistically significant mean reduction in the Hamilton Depression improvement (Thompson et al., 2001).
Rating Scale scores from baseline to the end of treatment (Milev The two most extensively researched non-pharmacological
et al., 2009). The largest study to-date, a double-blinded RCT of treatments for LLD are CBT (Serfaty et al., 2009) and Interpersonal
rTMS in geriatric patients with ‘vascular’ depression, demonstrated therapy (IPT) (Reynolds et al., 1999). Briefly, CBT focuses on
the efficacy of rTMS of the left DLPFC compared with sham challenging negative thoughts and changing the behaviours that
treatment (Jorge et al., 2008). Both older age and smaller frontal reinforce them. Dysfunctional patterns of thought and behaviour are
gray matter volumes were associated with poorer response to identified, and interventions include challenging unhelpful thinking,
rTMS although interestingly the burden of WMLs was not reducing avoidant behaviour and introducing positive behaviour
associated with treatment response. These findings suggest that patterns (Hepple, 2004). IPT, on the other hand, takes as its focus
rTMS may be an effective alternative treatment in LLD when disturbances in current relationships; a range of interventions to
accompanied by cerebrovascular disease. improve communication and expression of emotion are employed in
Recent years have witnessed an increasing interest in the role order to reduce symptoms and improve levels of function (Hepple,
that genetic polymorphisms may play in antidepressant response. In 2004). Both therapies are best delivered in approximately 10–20
middle-aged subjects, there is as yet no clear set of predictors of sessions. Recently there has been increased interest in briefer and
response to pharmacological interventions (Licinio and Wong, alternative psychotherapeutic interventions, such as problem
2011). Consistent with the more general treatment literature, there solving therapy (PST). This treatment trains patients to identify
is little evidence for any strong associations between specific problems that are affecting their wellbeing, and provides education
markers and treatment response in older persons, though some regarding how to select and then enact a plan for solving the problem
associations have emerged, as outlined above. In a study of 246 older (Arean et al., 2010). It is based on the assumption that if patients are
outpatients treated with either paroxetine or mirtazapine, several better able to manage their lives, they will experience less stress and
polymorphisms at the ABCB1 (i.e. ATP-binding cassette, subfamily B, a reduction in depressive symptoms. There is evidence to suggest
member 1 transporter (ABCB1); a gene that produces P-glycopro- that PST has efficacy in older adults with LLD (Arean et al., 1993) and
tein, which affects the transport of drugs out of cells) locus appeared may be particularly suited to those with executive dysfunction. For
to predict treatment outcome in geriatric patients taking paroxetine instance, a recent RCT investigated the efficacy of PST for LLD with
(which is a substrate for P-glycoprotein) (Sarginson et al., 2010). concomitant executive dysfunction (Arean et al., 2010). Two hundred
However, these results were no longer significant after Bonferroni and twenty one adults were randomly assigned to weekly sessions of
correction. Similarly a reported finding of an association between either supportive therapy or PST for 12 weeks. Although reductions
polymorphisms in the FK506 binding protein 5 (FKBP5; a gene that is in depression severity were comparable between the two groups at
involved in the regulation of glucocorticoid receptor sensitivity, and week six, by weeks nine to twelve PST was more effective in reducing
response to antidepressant therapy in MD), was not replicated in a depressive symptoms and produced greater response and remission
sample of 246 geriatric patients (aged 65 years and older) treated for rates than supportive therapy. Additionally, this form of therapy was
eight weeks in a double-blind randomised trial of mirtazapine and associated with reductions in disability that were sustained
paroxetine (Sarginson et al., 2009). Despite their implicated role in (Alexopoulos et al., 2011). These results have important implications
pathogenesis, analysis of genetic polymorphisms of the 5HTTLPR for offering non-pharmacological strategies to those with cognitive
gene has also returned null findings with regard to outcome of impairment and pharmacological treatment resistance.
antidepressant treatment in LLD, though in most studies, the S allele Rather than address the depressive features directly, investi-
does appear to be associated with a slower treatment response (see gators are now beginning to address other factors such as
Gerretsen and Pollock, 2008 for review). disturbed sleep, disrupted circadian cycles and reduced physical
130 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

activity which are critically linked to LLD. Lieverse et al. (2011) of underlying aetiological risk factors as well as factors which
recently published a double-blind placebo-controlled RCT, which perpetuate symptom persistence (Naismith et al., 2009a). To-date,
examined the role of bright light therapy for improving depressive there have been few studies that actively seek to reduce the
symptoms. In a sample of 89 patients aged over 60 years, three incidence of LoD. A limited range of studies has focused on broad
weeks of 1-h bright light was associated with improvement in interventions such as increased mental health literacy and folate
mood that was sustained for a further three weeks after treatment supplementation (Christensen et al., 2011). Increasingly, a focus on
cessation and which was associated with concomitant changes in providing targeted non-pharmacological, novel supplements (fish
melatonin rise and salivary cortisol. These data suggest that oils, folate) or traditional antidepressants to populations who are
therapies directly addressing the sleep–wake system have signifi- at risk due to either prior depressive symptoms or presence of VRFs
cant antidepressant effects. Exercise-based treatments have been has emerged (Cockayne et al., 2011; Walker et al., 2010). Since data
well evaluated in older adults, and look to be particularly effective suggests that even mild depressive symptoms are risk factors for
in the treatment and management of mood disturbance (Chodzko- later illness, approaches that utilise clinical disease staging may
Zajko et al., 2009; Lautenschlager et al., 2004). Evidence suggests also provide more effective frameworks for tracking the trajecto-
that regular physical exercise (such as walking or jogging) may be ries of illness progression and providing optimal periods for
as effective in reducing depressive symptoms as antidepressant intervention delivery (Naismith et al., 2009a). Providing education
therapy in depressed older adults (Blumenthal et al., 1999), though regarding modifiable risk factors for healthy brain ageing
beneficial effects of physical activity training appear to be (including VRFs) may be one way in which older age groups
correlated with the intensity of the intervention, and ideally may become more cognizant of the factors which contribute to
should include aerobic, muscle strengthening and flexibility cognitive decline, depression and dementia, and may offer
exercises (Chodzko-Zajko et al., 2009). Importantly, these thera- potential for prevention. Data suggests that older ‘at risk’ people
peutic effects may be sustainable, as demonstrated by Singh et al. can benefit from such educational programs in terms of knowledge
(2001) who reported persistently reduced depressive symptoms at (Norrie et al., 2011), though it is unclear whether such programs
20-week and 26-month follow-up assessments in individuals who translate into altered behaviour.
participated in a 10-week supervised exercise program. Further- Clearly, if the disability associated with persistent depression in
more, exercise has also been associated with reduced rates of later-life (with related cognitive decline and risk to dementia) is to
relapse over a 10-month follow-up period, particularly if be averted, effective new interventions (e.g. pharmacological, non-
individuals remained physically active over the follow-up period pharmacological and physical) need to be developed. Clearer
(Babyak et al., 2000). It has been suggested that physical exercise differentiation of the underlying pathophysiological models is
impacts on psychological well-being by improving psychosocial required to develop personalised treatments that will target the
constructs such as self-efficacy and functional independence, as deficits arising from dysfunction in the underlying circuitry.
well as by mediating metabolic and other physiological processes Clinical trials need to be stratified more carefully, with regards to
associated with depression and with ageing generally (Chodzko- underlying MRI parameters, neuropsychological function and
Zajko et al., 2009; Lautenschlager et al., 2004). While evidence has other neurobiological makers. In particular, older patients need
thus far suggested an important role for physical exercise and to be more clearly differentiated with regards to actual age of onset
activity in managing depression in older adults, further RCTs are of depression (i.e. early-onset grown old, versus genuine late-
needed to delineate its efficacy in specific groups such as those onset). Additionally, innovative health service models that target
with LoD and/or concomitant cerebrovascular disease. In addition, the specific (behavioural, cognitive, physical) needs of older
generalised exercise programs may not be suitable for depressed persons need to be designed and evaluated. The development of
individuals with medical comorbidities and/or VRFs or those such models for younger patients (McGorry et al., 2007) has
whom have been sedentary for some time. These individuals may already had a profound effect on clinical research directions in this
initially require supervised and/or individually tailored exercise cohort and is an excellent template for these later-onset disorders.
programs to ensure safety and compliance.
Due to the high preponderance of cognitive impairment in LLD, 14. Summary and future directions
some investigators have sought to improve neuropsychological
functioning in this group using cognitive training techniques. One To-date, neurobiological research in LLD has provided invalu-
recent study employing cognitive training demonstrated signifi- able insights into the key pathophysiological factors that underpin
cant improvements in learning and memory in older people with a this clinical disorder. In particular, it is increasingly clear that there
history of major depressive disorder and cognitive decline without is a need to focus attention on the pathways to depression that
dementia (Naismith et al., 2011a). These improvements corre- emerges for the first time in later-life. These data strongly
sponded with medium to large effect size changes (i.e. ÿ0.74 to implicate fronto-subcortical regions in the aetiology and perpetu-
ÿ0.82), which was considered to be a clinically meaningful ation of this disorder. This model only partially overlaps with other
improvement. However, there was no improvement in executive neurobiological models that have been developed for younger
functioning, which would be optimal if dysfunction in this domain patients with depressive disorders. Specifically, the key role of
indeed relates to other aspects of daily functioning such as disruption of subcortical and hippocampal elements in older
disability. Certainly, across the area of neuropsychiatry and patients (as compared with other areas of frontotemporal cortex) is
neurodegenerative disease, these techniques are showing promise emphasized.
as a way to slow cognitive decline (see review by Mowszowski This review has also highlighted the pronounced role depres-
et al., 2010), and this form of treatment warrants further sion plays in the emergence of neurodegenerative disease. A
investigation in LLD. converging body of structural and functional neuroimaging
research implicates various aspects of PFC functioning in
13. A focus on prevention for late-life depression? neuropsychological and clinical symptoms while the basal ganglia
and hippocampus also appear to have specific clinical correlates.
It is proposed that future paradigms targeting early interven- More recent research has provided a preliminary insight into
tion and management of both minor and major depression potential changes in the synaptic complex, neurometabolite
occurring in later-life need to shift from purely diagnostic models changes, and astrocytic markers of disease, and although more
toward those which incorporate early screening and management work is required, some data has linked HPA functioning,
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 131

inflammatory and glial markers to depressive symptoms and support, sleep and circadian systems) as well as the more
illness trajectories. Concurrently, research conducted largely in commonly associated medical and vascular risks. Similarly,
clinical samples has begun to unravel likely gene candidates that emerging research focusing on gene–environment interactions
now need further examination and validation in larger genome- must be incorporated as comprehensively into neurobiological
wide association studies. Future models of LLD must synthesise models of late-onset disorders as it is for early-onset disorders.
these findings into coherent testable frameworks within which the Primary and secondary prevention strategies must increasingly
interaction of these multi-layered systems can be examined. focus on how such strategies directly prevent or alleviate
Unfortunately, treatment options for patients with LLD are detrimental impacts on brain functioning (Naismith et al.,
under-evaluated. Consequently, we are still somewhat faced with 2009a). Indeed, the ‘translational gap’ between knowledge and
limited efficacy data that relate to this specific age and syndromal clinical practice remains wide and must be addressed if we are to
group (Mottram et al., 2006; Nelson et al., 2008; Pinquart et al., see public health impact (Insel, 2009b). The use of e-health
2006; Taylor and Doraiswamy, 2004). The same issues that are technologies (and other self-care strategies) may also provide a
challenging for younger cohorts (i.e. separation of response viable way for early screening and management of depression.
between active and non-specific treatments, emergence of new Current trials are underway to evaluate such approaches (Cock-
side-effect profiles with new treatments) are emerging as larger ayne et al., 2011). Similarly, large-scale prevention and early
numbers of older patients with depression receive active treat- intervention trials are urgently required if we are to reduce the
ments. Since the focus to-date has been largely on modifying burden of depression on medical and cognitive outcomes.
monoamine systems (typically serotonergic) for LLD, future Ultimately, it is clear that improved earlier identification and
research focusing on other critical neurotransmitters is now management of depression will be associated with lower preva-
warranted. Examination of the value of treatments in these critical lence rates of dementia, and consequently, enhanced independent
periods may be enhanced by greater reference to personalised living, less reliance on formal and informal caring systems, lower
treatment approaches that investigate individual patterns of levels of disability and decreased health care utilisation.
heterogeneity and treatment responsiveness as well as the more
traditional approaches of pooling group data (Insel, 2009b). Conflict of interest
Further development of targeted physical treatments (e.g. direct
and indirect brain stimulation techniques) is also deserving of Professor Hickie has led projects for health professionals and
specific evaluation. the community supported by governmental, community agency,
For older people, more emphasis must be placed on evaluating and drug industry partners (Wyeth, Eli Lily, Servier, Pfizer, Astra
the role of informational, psychological, behavioural and other Zeneca) for the identification and management of depression and
social interventions. This is particularly relevant given the large anxiety. He has served on advisory boards convened by the drug
number of role changes that older people encounter, as well as industry in relation to specific antidepressants, including nefazo-
factors such as grief and bereavement, onset of concurrent physical done, duloxetine and desvenlafaxine, and has participated in a
health problems, lack of physical activity, changes in the sleep and multicentre trial of agomelatine. He has participated in Servier-
circadian system, increasing social isolation and the disability sponsored educational programs related to circadian-based
directly attributable to other medical difficulties. Certainly, current therapies.
evidence suggests that psychosocial treatments are likely to have
superior benefits to medication in those with depression and Acknowledgements
dementia (Banerjee et al., 2011). Similarly, in those with cognitive
decline and disability (Alexopoulos et al., 2011; Mowszowski et al., A/Professor Naismith is supported by a Career Development
2010; Naismith et al., 2011a), psychological therapies show Award from the National Health and Medical Research Council of
promise. Australia (NHMRC). Professor Ian Hickie, Dr Louisa Norrie and Ms
Despite the significant advances made in identifying aetiolo- Loren Mowszowski are supported by an NHMRC Australia
gical contributors to LLD, the true challenge facing mental health Fellowship awarded to Professor Hickie.
research is the capacity to translate this basic biomedical
knowledge into enhanced health outcomes (Insel, 2009b). Future References
research must aim to evaluate whether such knowledge can be
successfully applied to early intervention paradigms for the late- Access Economics, 2009. Keeping Dementia Front of Mind: Incidence & Prevalence
onset mood and psychiatric disorders. Knowledge of relevant risk 2009-2050. Alzheimer’s Australia, Canberra.
Adams, K.B., 2001. Depressive symptoms, depletion, or developmental change?
factors should provide pertinent opportunities for early interven- Withdrawal, apathy, and lack of vigor in the geriatric depression scale. Geron-
tion paradigms (Naismith et al., 2009a), particularly since we are tologist 41, 768–777.
now beginning to understand how experience and biology interact Adler, G., Chwalek, K., Jajcevic, A., 2004. Six-month course of mild cognitive
impairment and affective symptoms in late-life depression. European Psychia-
over the life cycle. Emerging neuroimaging, neuropsychological try 19, 502–505.
and other neurobiological markers for illness onset and progres- Aguilera, M., Arias, B., Wichers, M., Barrantes-Vidal, N., Moya, J., Villa, H., van Os, J.,
sion characterize the field; compared with other neuropsychiatric Ibanez, M.I., Ruiperez, M.A., Ortet, G., Fananas, L., 2009. Early adversity and 5-
HTT/BDNF genes: new evidence of gene-environment interactions on depres-
disorders, this is a major advantage in terms of predicting sive symptoms in a general population. Psychological Medicine 39, 1425–1432.
longitudinal course and assessing response to treatments. Fur- Aizenstein, H.J., Andreescu, C., Edelman, K.L., Cochran, J.L., Price, P., Butters, M.A.,
thermore, since this last decade has witnessed major advances in Karp, J., Patel, M., Reynolds III, C.F., 2011. fMRI correlates of white matter
hyperintensities in late-life depression. The American Journal of Psychiatry
the field of neuroplasticity, it now seems apparent that rapid
168, 1075–1082.
reorganization can occur in the brain in response to negative, as Aizenstein, H.J., Butters, M.A., Figurski, J.L., Stenger, V.A., Reynolds 3rd, C.F., Carter,
well as positive stimuli (Mahncke et al., 2006). In this sense, C.S., 2005. Prefrontal and striatal activation during sequence learning in geri-
atric depression. Biological Psychiatry 58, 290–296.
paradigms for LLD need to incorporate prevention of risk factors
Aizenstein, H.J., Butters, M.A., Wu, M., Mazurkewicz, L.M., Stenger, V.A., Gianaros,
that confer common pathways to depression and dementia as well P.J., Becker, J.T., Reynolds 3rd, C.F., Carter, C.S., 2009. Altered functioning of the
as promotion of those factors that lead to healthy brain ageing. executive control circuit in late-life depression: episodic and persistent phe-
Research has not yet addressed how health interventions nomena. The American Journal of Geriatric Psychiatry 17, 30–42.
Alexander, G.E., DeLong, M.R., Strick, P.L., 1986. Parallel organization of functionally
should ideally incorporate the various trajectories to illness onset segregated circuits linking basal ganglia and cortex. Annual Review of Neuro-
including multiple lifestyle factors (e.g. diet, exercise, social science 9, 357–381.
132 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

Alexander, N., Kuepper, Y., Schmitz, A., Osinsky, R., Kozyra, E., Hennig, J., 2009. Antleman, S., Chiodo, L.A., 1981. Dopamine autoreceptor sub-sensitivity: a mecha-
Gene-environment interactions predict cortisol responses after acute stress: nism common to the treatment of depression and the induction of amphet-
implications for the etiology of depression. Psychoneuroendocrinology 34, amine psychosis. Biological Psychiatry 16, 717–727.
1294–1303. Apostolova, L.G., Dutton, R.A., Dinov, I.D., Hayashi, K.M., Toga, A.W., Cummings, J.L.,
Alexopoulos, G.S., 2003. Vascular disease, depression, and dementia. Journal of the Thompson, P.M., 2006. Conversion of mild cognitive impairment to Alzheimer
American Geriatrics Society 51, 1178–1180. disease predicted by hippocampal atrophy maps. Archives of Neurology 63,
Alexopoulos, G.S., 2005. Depression in the elderly. Lancet 365, 1961–1970. 693–699.
Alexopoulos, G.S., 2011. Pharmacotherapy for late-life depression. The Journal of Arean, P.A., Perri, M.G., Nezu, A.M., Schein, R.L., Christopher, F., Joseph, T.X., 1993.
Clinical Psychiatry 72, e04. Comparative effectiveness of social problem-solving therapy and reminiscence
Alexopoulos, G.S., Buckwalter, K., Olin, J., Martinez, R., Wainscott, C., Krishnan, K.R., therapy as treatments for depression in older adults. Journal of Consulting and
2002a. Comorbidity of late life depression: an opportunity for research on Clinical Psychology 61, 1003–1010.
mechanisms and treatment. Biological Psychiatry 52, 543–558. Arean, P.A., Raue, P., Mackin, R.S., Kanellopoulos, D., McCulloch, C., Alexopoulos, G.S.,
Alexopoulos, G.S., Chester, J.G., 1992. Outcomes of geriatric depression. Clinics in 2010. Problem-solving therapy and supportive therapy in older adults with
Geriatric Medicine 8, 363–376. major depression and executive dysfunction. The American Journal of Psychia-
Alexopoulos, G.S., Glatt, C.E., Hoptman, M.J., Kanellopoulos, D., Murphy, C.F., Kelly try 167, 1391–1398.
Jr., R.E., Morimoto, S.S., Lim, K.O., Gunning, F.M., 2010. BDNF Val66met poly- Awad, I.A., Johnson, P.C., Spetzler, R.F., Hodak, J.A., 1986. Incidental subcortical
morphism, white matter abnormalities and remission of geriatric depression. lesions identified on magnetic resonance imaging in the elderly. II Postmortem
Journal of Affective Disorders 125, 262–268. pathological correlations. Stroke 17, 1090–1097.
Alexopoulos, G.S., Kiosses, D.N., Choi, S.J., Murphy, C.F., Lim, K.O., 2002b. Awata, S., Ito, H., Konno, M., Ono, S., Kawashima, R., Fukuda, H., Sato, M., 1998.
Frontal white matter microstructure and treatment response of late-life Regional cerebral blood flow abnormalities in late-life depression: relation to
depression: a preliminary study. The American Journal of Psychiatry 159, refractoriness and chronification. Psychiatry and Clinical Neurosciences 52, 97–
1929–1932. 105.
Alexopoulos, G.S., Kiosses, D.N., Klimstra, S., Kalayam, B., Bruce, M.L., 2002c. Clinical Babyak, M., Blumenthal, J.A., Herman, S., Khatri, P., Doraiswamy, M., Moore, K.,
presentation of the ‘‘depression-executive dysfunction syndrome’’ of late life. Craighead, W.E., Baldewicz, T.T., Krishman, K.R., 2000. Exercise treatment for
The American Journal of Geriatric Psychiatry 10, 98–106. major depression: maintenance of therapeutic benefit at 10 months. Psycho-
Alexopoulos, G.S., Meyers, B.S., Young, R.C., Kakuma, T., Feder, M., Einhorn, A., somatic Medicine 62, 633–638.
Rosendahl, E., 1996. Recovery in geriatric depression. Archives of General Bae, J.N., MacFall, J.R., Krishnan, K.R., Payne, M.E., Steffens, D.C., Taylor, W.D., 2006.
Psychiatry 53, 305–312. Dorsolateral prefrontal cortex and anterior cingulate cortex white matter
Alexopoulos, G.S., Meyers, B.S., Young, R.C., Kakuma, T., Silbersweig, D., Charlson, M., alterations in late-life depression. Biological Psychiatry 60, 1356–1363.
1997. Clinically defined vascular depression. The American Journal of Psychia- Baldwin, R., Jeffries, S., Jackson, A., Sutcliffe, C., Thacker, N., Scott, M., Burns, A., 2004.
try 154, 562–565. Treatment response in late-onset depression: relationship to neuropsychologi-
Alexopoulos, G.S., Meyers, B.S., Young, R.C., Mattis, S., Kakuma, T., 1993. The course cal, neuroradiological and vascular risk factors. Psychological Medicine 34,
of geriatric depression with ‘‘reversible dementia’’: a controlled study. The 125–136.
American Journal of Psychiatry 150, 1693–1699. Baldwin, R., Wild, R., 2004. Management of depression in later life. Advances in
Alexopoulos, G.S., Morimoto, S.S., 2011. The inflammation hypothesis in geriatric Psychiatric Treatment 10, 131–139.
depression. International Journal of Geriatric Psychiatry 26, 1109–1118. Baldwin, R.C., Gallagley, A., Gourlay, M., Jackson, A., Burns, A., 2006. Prognosis of late
Alexopoulos, G.S., Murphy, C.F., Gunning-Dixon, F.M., Glatt, C.E., Latoussakis, V., life depression: a three-year cohort study of outcome and potential predictors.
Kelly Jr., R.E., Kanellopoulos, D., Klimstra, S., Lim, K.O., Young, R.C., Hoptman, International Journal of Geriatric Psychiatry 21, 57–63.
M.J., 2009. Serotonin transporter polymorphisms, microstructural white matter Balleine, B.W., O’Doherty, J.P., 2010. Human and rodent homologies in action
abnormalities and remission of geriatric depression. Journal of Affective Dis- control: corticostriatal determinants of goal-directed and habitual action.
orders 119, 132–141. Neuropsychopharmacology 35, 48–69.
Alexopoulos, G.S., Murphy, C.F., Gunning-Dixon, F.M., Latoussakis, V., Kanellopou- Ballmaier, M., Kumar, A., Thompson, P.M., Narr, K.L., Lavretsky, H., Estanol, L.,
los, D., Klimstra, S., Lim, K.O., Hoptman, M.J., 2008. Microstructural white matter Deluca, H., Toga, A.W., 2004a. Localizing gray matter deficits in late-onset
abnormalities and remission of geriatric depression [see comment]. The Amer- depression using computational cortical pattern matching methods. The Amer-
ican Journal of Psychiatry 165, 238–244. ican Journal of Psychiatry 161, 2091–2099.
Alexopoulos, G.S., Raue, P.J., Kiosses, D.N., Mackin, R.S., Kanellopoulos, D., McCul- Ballmaier, M., Narr, K.L., Toga, A.W., Elderkin-Thompson, V., Thompson, P.M.,
loch, C., Arean, P.A., 2011. Problem-solving therapy and supportive therapy in Hamilton, L., Haroon, E., Pham, D., Heinz, A., Kumar, A., 2008. Hippocampal
older adults with major depression and executive dysfunction: effect on morphology and distinguishing late-onset from early-onset elderly depression.
disability. Archives of General Psychiatry 68, 33–41. The American Journal of Psychiatry 165, 229–237.
Alexopoulos, G.S., Young, R.C., Shindledecker, R.D., 1992. Brain computed tomogra- Ballmaier, M., Sowell, E.R., Thompson, P.M., Kumar, A., Narr, K.L., Lavretsky, H.,
phy findings in geriatric depression and primary degenerative dementia. Bio- Welcome, S.E., DeLuca, H., Toga, A.W., 2004b. Mapping brain size and cortical
logical Psychiatry 31, 591–599. gray matter changes in elderly depression. Biological Psychiatry 55, 382–389.
Allen, N.J., Barres, B.A., 2009. NEUROSCIENCE glia – more than just brain glue. Ballmaier, M., Toga, A.W., Blanton, R.E., Sowell, E.R., Lavretsky, H., Peterson, J., Pham,
Nature 457, 675–677. D., Kumar, A., 2004c. Anterior cingulate, gyrus rectus, and orbitofrontal abnor-
Almeida, O.P., 2008. Vascular depression: myth or reality? International Psycho- malities in elderly depressed patients: an MRI-based parcellation of the pre-
geriatrics 20, 645–652. frontal cortex. The American Journal of Psychiatry 161, 99–108.
Almeida, O.P., Burton, E.J., Ferrier, N., McKeith, I.G., O’Brien, J.T., 2003. Depression Banati, R., Hickie, I.B., 2009. Therapeutic signposts: using biomarkers to guide better
with late onset is associated with right frontal lobe atrophy. Psychological treatment of schizophrenia and other psychotic disorders. The Medical Journal
Medicine 33, 675–681. of Australia 190, S26–S32.
Almeida, O.P., McCaul, K., Hankey, G.J., Norman, P., Jamrozik, K., Flicker, L., 2008. Banati, R.B., Newcombe, J., Gunn, R.N., Cagnin, A., Turkheimer, F., Heppner, F., Price,
Homocysteine and depression in later life. Archives of General Psychiatry 65, G., Wegner, F., Giovannoni, G., Miller, D.H., Perkin, G.D., Smith, T., Hewson, A.K.,
1286–1294. Bydder, G., Kreutzberg, G.W., Jones, T., Cuzner, M.L., Myers, R., 2000. The
Almeida, O.P., Norman, P., Hankey, G.J., Jamrozik, K., Flicker, L., 2007. The association peripheral benzodiazepine binding site in the brain in multiple sclerosis:
between C-reactive protein concentration and depression in later life is due to quantitative in vivo imaging of microglia as a measure of disease activity. Brain
poor physical health: results from the Health in Men Study (HIMS). Psychologi- 123 (Pt 11), 2321–2337.
cal Medicine 37, 1775–1786. Banerjee, S., Hellier, J., Dewey, M., Romeo, R., Ballard, C., Baldwin, R., Bentham, P.,
Almeida, O.P., Norman, P.E., Allcock, R., van Bockxmeer, F., Hankey, G.J., Jamrozik, K., Fox, C., Holmes, C., Katona, C., Knapp, M., Lawton, L., Lindesay, J., Livingston, G.,
Flicker, L., 2009. Polymorphisms of the CRP gene inhibit inflammatory response McCrae, N., Moniz-Cook, E., Murray, J., Nurock, S., Orrell, M., O’Brien, J., Poppe,
and increase susceptibility to depression: the Health in Men Study. Interna- M., Thomas, A., Walwyn, R., Wilson, K., Burns, A., 2011. Sertraline or mirtazapine
tional Journal of Epidemiology 38, 1049–1059. for depression in dementia (HTA-SADD): a randomised, multicentre, double-
American Psychiatric Association, 1994. Diagnostic and Statistical Manual of Men- blind, placebo-controlled trial. Lancet 378, 403–411.
tal Disorders, DSM-IV. American Psychiatric Press, Washington, DC. Banks, W.A., Ortiz, L., Plotkin, S.R., Kastin, A.J., 1991. Human interleukin (IL) 1 alpha,
Anbeek, P., Vincken, K.L., van Osch, M.J.P., Bisschops, R.H.C., van der Grond, J., 2004. murine IL-1 alpha and murine IL-1 beta are transported from blood to brain in
Probabilistic segmentation of white lesions in MR imaging. NeuroImage 21, the mouse by a shared saturable mechanism. The Journal of Pharmacology and
1037–1044. Experimental Therapeutics 259, 988–996.
Andreescu, C., Butters, M.A., Begley, A., Rajji, T., Wu, M., Meltzer, C.C., Reynolds 3rd, Barnes, D.E., Yaffe, K., 2011. The projected effect of risk factor reduction on
C.F., Aizenstein, H., 2008. Gray matter changes in late life depression – a Alzheimer’s disease prevalence. Lancet Neurology 10, 819–828.
structural MRI analysis. Neuropsychopharmacology 33, 2566–2572. Bartolini, M., Coccia, M., Provinciali, L., Gabriella Ceravolo, M., 2005. Motivational
Andreescu, C., Reynolds, C.F., 2011. Late-life depression: evidence-based treatment symptoms of depression mask preclinical Alzheimer’s disease in elderly sub-
and promising new directions for research and clinical practice. The Psychiatric jects. Dementia and Geriatric Cognitive Disorders 19, 31–36.
Clinics of North America 34, 335–355. Basso, M.R., Bornstein, R.A., 1999. Relative memory deficits in recurrent versus first-
Andrews, G., Issakidis, C., Sanderson, K., Corry, J., Lapsley, H., 2004. Utilising Survey episode major depression on a word-list learning task. Neuropsychology 13,
Data to Inform Public Policy: Comparison of the Cost-Effectiveness of Treatment 557–563.
of Ten Mental Disorders. , pp. 526–533. Beekman, A.T.F., Deeg, D.J.H., Braam, A.W., Smit, J.H., Van Tilburg, W., 1997. Con-
Anstey, K., Brodaty, H., 1995. Antidepressants and the elderly: double-blind trials sequences of major and minor depression in later life: a study of disability, well-
1987-1992. International Journal of Geriatric Psychiatry 10, 265–279. being and service utilization. Psychological Medicine 27, 1397–1409.
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 133

Beekman, A.T.F., Deeg, D.J.H., vanTilburg, T., Smit, J.H., Hooijer, C., vanTilburg, W., Bueller, J.A., Aftab, M., Sen, S., Gomez-Hassan, D., Burmeister, M., Zubieta, J.K., 2006.
1995. Major and minor depression in later life: a study of prevalence and risk BDNF Val66Met allele is associated with reduced hippocampal volume in
factors. Journal of Affective Disorders 36, 65–75. healthy subjects. Biological Psychiatry 59, 812–815.
Bell-McGinty, S., Butters, M.A., Meltzer, C.C., Greer, P.J., Reynolds 3rd, C.F., Becker, Bunker, S.J., Colquhoun, D.M., Esler, M.D., Hickie, I.B., Hunt, D., Jelinek, V.M., Old-
J.T., 2002. Brain morphometric abnormalities in geriatric depression: long-term enburg, B.F., Peach, H.G., Ruth, D., Tennant, C.C., Tonkin, A.M., 2003. ‘‘Stress’’ and
neurobiological effects of illness duration. The American Journal of Psychiatry coronary heart disease: psychosocial risk factors. The Medical Journal of
159, 1424–1427. Australia 178, 272–276.
Belmaker, R.H., Agam, G., 2008. Major depressive disorder mechanisms of disease. Butters, M.A., Aizenstein, H.J., Hayashi, K.M., Meltzer, C.C., Seaman, J., Reynolds 3rd,
The New England Journal of Medicine 358, 55–68. C.F., Toga, A.W., Thompson, P.M., Becker, J.T., 2009. Three-dimensional surface
Benjamin, S., McQuoid, D.R., Potter, G.G., Payne, M.E., MacFall, J.R., Steffens, D.C., mapping of the caudate nucleus in late-life depression. The American Journal of
Taylor, W.D., 2010. The brain-derived neurotrophic factor Val66Met polymor- Geriatric Psychiatry 17, 4–12.
phism, hippocampal volume, and cognitive function in geriatric depression. The Butters, M.A., Becker, J.T., Nebes, R.D., Zmuda, M.D., Mulsant, B.H., Pollock, B.G.,
American Journal of Geriatric Psychiatry 18, 323–331. Reynolds 3rd, C.F., 2000. Changes in cognitive functioning following treatment
Bennett, M.R., 2010. Synapse regression in depression: the role of 5-HT receptors in of late-life depression. The American Journal of Psychiatry 157, 1949–1954.
modulating NMDA receptor function and synaptic plasticity. The Australian and Butters, M.A., Klunk, W.E., Mathis, C.A., Price, J.C., Ziolko, S.K., Hoge, J.A., Tsopelas,
New Zealand Journal of Psychiatry 44, 301–308. N.D., Lopresti, B.J., Reynolds, C.F.I., DeKosky, S.T., Meltzer, C.C., 2008. Imaging
Berger, A.K., Fratiglioni, L., Forsell, Y., Winblad, B., Backman, L., 1999. The occurrence Alzheimer pathology in late-life depression with PET and Pittsburgh Com-
of depressive symptoms in the preclinical phase of AD: a population-based pound-B. Alzheimer Disease and Associated Disorders 22, 261–268.
study. Neurology 53, 1998–2002. Byrum, C.E., Ahearn, E.P., Krishnan, K.R., 1999. A neuroanatomic model for depres-
Berman, R.M., Cappiello, A., Anand, A., Oren, D.A., Heninger, G.R., Charney, D.S., sion. Progress in Neuro-psychopharmacology & Biological Psychiatry 23, 175–
Krystal, J.H., 2000. Antidepressant effects of ketamine in depressed patients. 193.
Biological Psychiatry 47, 351–354. Caetano, S.C., Hatch, J.P., Brambilla, P., Sassi, R.B., Nicoletti, M., Mallinger, A.G., Frank,
Berton, O., McClung, C.A., DiLeone, R.J., Krishnan, V., Renthal, W., Russo, S.J., Graham, E., Kupfer, D.J., Keshavan, M.S., Soares, J.C., 2004. Anatomical MRI study of
D., Tsankova, N.M., Bolanos, C.A., Rios, M., Monteggia, L.M., Self, D.W., Nestler, hippocampus and amygdala in patients with current and remitted major
E.J., 2006. Essential role of BDNF in the mesolimbic dopamine pathway in social depression. Psychiatry Research 132, 141–147.
defeat stress. Science 311, 864–868. Camus, V., Kraehenbuhl, H., Preisig, M., Bula, C.J., Waeber, G., 2004. Geriatric
Bhogal, S.K., Teasell, R., Foley, N., Speechley, M., 2004. Lesion location and poststroke depression and vascular disease: what are the links? Journal of Affective
depression: systematic review of the methodological limitations in the litera- Disorders 81, 1–16.
ture. Stroke 35, 794–802. Caraci, F., Copani, A., Nicoletti, F., Drago, F., 2010. Depression and Alzheimer’s
Bjelland, I., Tell, G.S., Vollset, S.E., Refsum, H., Ueland, P.M., 2003. Folate, vitamin disease: neurobiological links and common pharmacological targets. European
B12, homocysteine, and the MTHFR 677C->T polymorphism in anxiety and Journal of Pharmacology 626, 64–71.
depression: the Hordaland Homocysteine Study. Archives of General Psychiatry Carney, R.M., Freedland, K.E., Veith, R.C., 2005. Depression, the autonomic nervous
60, 618–626. system, and coronary heart disease. Psychosomatic Medicine 67, S29–S33.
Blazer, D.G., 2003. Depression in late life: review and commentary. The Journals of Caspi, A., Hariri, A.R., Holmes, A., Uher, R., Moffitt, T.E., 2010. Genetic sensitivity to
Gerontology. Series A, Biological Sciences and Medical Sciences 58, 249–265. the environment: the case of the serotonin transporter gene and its implications
Bloom, D.E., Cafiero, E.T., Jané-Llopis, E., Abrahams-Gessel, S., Bloom, L.R., Fathima, for studying complex diseases and traits. The American Journal of Psychiatry
S., Feigl, A.B., Gaziano, T., Mowafi, M., Pandya, A., Prettner, K., Rosenberg, L., 167, 509–527.
Seligman, B., Stein, A., Weinstein, C., 2011. The global economic burden of non- Caspi, A., Sugden, K., Moffitt, T.E., Taylor, A., Craig, I.W., Harrington, H., McClay, J.,
communicable diseases. In: World Economic Forum, Geneva. Mill, J., Martin, J., Braithwaite, A., Poulton, R., 2003. Influence of life stress on
Blumenthal, J.A., Babayak, M.A., Moore, K.A., Craighead, W.E., Herman, S., Khatri, P., depression: moderation by a polymorphism in the 5-HTT gene. Science 301,
Waugh, R., Napolitano, M.A., Forman, L.M., Appelbaum, M., Doraiswamy, P.M., 386–389.
Krishnan, K.R., 1999. Effects of exercise training on older patients with major Charles, H.C., Lazeyras, F., Krishnan, K.R., Boyko, O.B., Payne, M., Moore, D., 1994.
depression. Archives of Internal Medicine 159, 2349–2356. Brain choline in depression: in vivo detection of potential pharmacodynamic
Bondareff, W., Alpert, M., Friedhoff, A.J., Richter, E.M., Clary, C.M., Batzar, E., 2000. effects of antidepressant therapy using hydrogen localized spectroscopy. Prog-
Comparison of sertraline and nortriptyline in the treatment of major depressive ress in Neuro-psychopharmacology & Biological Psychiatry 18, 1121–1127.
disorder in late life. The American Journal of Psychiatry 157, 729–736. Chen, B., Dowlatshahi, D., MacQueen, G.M., Wang, J.F., Young, L.T., 2001. Increased
Bots, M.L., Hofman, A., Grobbee, D.E., 1997. Increased common carotid intima- hippocampal BDNF immunoreactivity in subjects treated with antidepressant
media thickness. Adaptive response or a reflection of atherosclerosis? Findings medication. Biological Psychiatry 50, 260–265.
from the Rotterdam Study. Stroke 28, 2442–2447. Chen, C.S., Chen, C.C., Kuo, Y.T., Chiang, I.C., Ko, C.H., Lin, H.F., 2006. Carotid intima-
Bowen, D.M., Najlerahim, A., Procter, A.W., Francis, P.T., Murphy, E., 1989. Circum- media thickness in late-onset major depressive disorder. International Journal
scribed changes of the cerebral cortex in neuropsychiatric disorders of later life. of Geriatric Psychiatry 21, 36–42.
Proceedings of the National Academy of Sciences of the United States of America Chen, C.S., Chiang, I.C., Li, C.W., Lin, W.C., Lu, C.Y., Hsieh, T.J., Liu, G.C., Lin, H.F., Kuo,
86, 9504–9508. Y.T., 2009. Proton magnetic resonance spectroscopy of late-life major depres-
Bowley, M.P., Drevets, W.C., Ongur, D., Price, J.L., 2002. Low glial numbers in the sive disorder. Psychiatry Research 172, 210–214.
amygdala in major depressive disorder. Biological Psychiatry 52, 404–412. Chen, C.S., Tsai, J.C., Tsang, H.Y., Kuo, Y.T., Lin, H.F., Chiang, I.C., Devanand, D.P., 2005.
Brassen, S., Kalisch, R., Weber-Fahr, W., Braus, D.F., Buchel, C., 2008. Ventromedial Homocysteine levels, MTHFR C677T genotype, and MRI hyperintensities in late-
prefrontal cortex processing during emotional evaluation in late-life depres- onset major depressive disorder. The American Journal of Geriatric Psychiatry
sion: a longitudinal functional magnetic resonance imaging study. Biological 13, 869–875.
Psychiatry 64, 349–355. Chen, P., Ganguli, M., Mulsant, B.H., DeKosky, S.T., 1999. The temporal relationship
Bremmer, M.A., Beekman, A.T., Deeg, D.J., Penninx, B.W., Dik, M.G., Hack, C.E., between depressive symptoms and dementia: a community-based prospective
Hoogendijk, W.J., 2008. Inflammatory markers in late-life depression: results study. Archives of General Psychiatry 56, 261–266.
from a population-based study. Journal of Affective Disorders 106, 249–255. Chen, Y., Patel, N.C., Guo, J.J., Zhan, S., 2007. Antidepressant prophylaxis for post-
Bremmer, M.A., Deeg, D.J., Beekman, A.T., Penninx, B.W., Lips, P., Hoogendijk, W.J., stroke depression: a meta-analysis. International Clinical Psychopharmacology
2007. Major depression in late life is associated with both hypo- and hyper- 22, 159–166.
cortisolemia. Biological Psychiatry 62, 479–486. Cho, H.J., Lavretsky, H., Olmstead, R., Levin, M.J., Oxman, M.N., Irwin, M.R., 2008.
Breteler, M.M.B., van Amerongen, N.M., van Swieten, J.C., Claus, J.J., Grobbee, D.E., Sleep disturbance and depression recurrence in community-dwelling older
van Gjin, J., Hofman, A., van Harskamp, F., 1994. Cognitive correlates of ven- adults: a prospective study. The American Journal of Psychiatry 165, 1543–
tricular enlargement and cerebral white matter lesions on MRI. The Rotterdam 1550.
Study. Stroke 25, 1109–1115. Chodzko-Zajko, W.J., Proctor, D.N., Fiatarone Singh, M.A., Minson, C.T., Nigg, C.R.,
Brockmann, H., Zobel, A., Joe, A., Biermann, K., Scheef, L., Schuhmacher, A., von Salem, G.J., Skinner, J.S., 2009. Exercise and physical activity for older adults:
Widdern, O., Metten, M., Biersack, H.J., Maier, W., Boecker, H., 2009. The value of position stand. Medicine and Science in Sports and Exercise 41, 1510–1530.
HMPAO SPECT in predicting treatment response to citalopram in patients with Christensen, H., Aiken, A., Batterham, P.J., Walker, J., Mackinnon, A.J., Fenech, M.,
major depression. Psychiatry Research 173, 107–112. Hickie, I.B., 2011. No clear potentiation of antidepressant medication effects by
Brodaty, H., Harris, L., Peters, K., Wilhelm, K., Hickie, I., Boyce, P., Mitchell, P., Parker, folic acid+vitamin B12 in a large community sample. Journal of Affective
G., Eyers, K., 1993. Prognosis of depression in the elderly. A comparison with Disorders 130, 37–45.
younger patients. The British Journal of Psychiatry 163, 589–596. Clarke, R., Daly, L., Robinson, K., Naughten, E., Cahalane, S., Fowler, B., Graham, I.,
Brodaty, H., Luscombe, G., Parker, G., Wilhelm, K., Hickie, I., Austin, M.P., Mitchell, P., 1991. Hyperhomocysteinemia – an independent risk factor for vascular-dis-
1997. Increased rate of psychosis and psychomotor change in depression with ease. The New England Journal of Medicine 324, 1149–1155.
age. Psychological Medicine 27, 1205–1213. Cockayne, N.L., Glozier, N., Naismith, S.L., Christensen, H., Neal, B., Hickie, I.B., 2011.
Brodaty, H., Luscombe, G., Parker, G., Wilhelm, K., Hickie, I., Austin, M.P., Mitchell, P., Internet-based treatment for older adults with depression and co-morbid
2001. Early and late onset depression in old age: different aetiologies, same cardiovascular disease: protocol for a randomised, double-blind, placebo con-
phenomenology. Journal of Affective Disorders 66, 225–236. trolled trial. BMC Psychiatry 11, 10.
Brown, A.S., Gershon, S., 1993. Dopamine and depression. Journal of Neural Trans- Coffey, C.E., Wilkinson, W.E., Weiner, R.D., Parashos, I.A., Djang, W.T., Webb, M.C.,
mission 91, 75–109. Figiel, G.S., Spritzer, C.E., 1993. Quantitative cerebral anatomy in depression. A
Bruce, E.C., Musselman, D.L., 2005. Depression, alterations in platelet function, and controlled magnetic resonance imaging study. Archives of General Psychiatry
ischemic heart disease. Psychosomatic Medicine 67, S34–S36. 50, 7–16.
134 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

Colloby, S.J., Firbank, M.J., Thomas, A.J., Vasudev, A., Parry, S.W., O’Brien, J.T., 2011. Devanand, D.P., Pelton, G.H., Marston, K., Camacho, Y., Roose, S.P., Stern, Y., Sackeim,
White matter changes in late-life depression: a diffusion tensor imaging study. H.A., 2003. Sertraline treatment of elderly patients with depression and cogni-
Journal of Affective Disorders 135, 216–220. tive impairment. International Journal of Geriatric Psychiatry 18, 123–130.
Comings, D.E., MacMurray, J.P., Gonzalez, N., Ferry, L., Peters, W.R., 1999. Associa- Dew, M.A., Reynolds 3rd, C.F., Houck, P.R., Hall, M., Buysse, D.J., Frank, E., Kupfer, D.J.,
tion of the serotonin transporter gene with serum cholesterol levels and heart 1997. Temporal profiles of the course of depression during treatment. Pre-
disease. Molecular Genetics and Metabolism 67, 248–253. dictors of pathways toward recovery in the elderly. Archives of General Psy-
Conwell, Y., Nelson, J.C., Kim, K.M., Mazure, C.M., 1989. Depression in late life: age of chiatry 54, 1016–1024.
onset as marker of a subtype. Journal of Affective Disorders 17, 189–195. Dewar, D., Underhill, S.M., Goldberg, M.P., 2003. Oligodendrocytes and ischemic
Cooper, C., Katona, C., Lyketsos, K., Blazer, D., Brodaty, H., Rabins, P., Lima, C.A., brain injury. Journal of Cerebral Blood Flow and Metabolism 23, 263–274.
Livingston, G., 2011. A systematic review of treatments for refractory depres- Diaconescu, A.O., Kramer, E., Hermann, C., Ma, Y., Dhawan, V., Chaly, T., Eidelberg, D.,
sion in older people. The American Journal of Psychiatry 168, 681–688. McIntosh, A.R., Smith, G.S., 2010. Distinct functional networks associated with
Corsentino, E.A., Sawyer, K., Sachs-Ericsson, N., Blazer, D.G., 2009. Depressive improvement of affective symptoms and cognitive function during citalopram
symptoms moderate the influence of the apolipoproteine epsilon4 allele on treatment in geriatric depression. Human Brain Mapping 32, 1677–1691.
cognitive decline in a sample of community dwelling older adults. The Ameri- Diniz, B.S., Teixeira, A.L., Talib, L., Gattaz, W.F., Forlenza, O.V., 2010a. Interleukin-1
can Journal of Geriatric Psychiatry 17, 155–165. beta serum levels is increased in antidepressant-free elderly depressed
Cotter, D., Hudson, L., Landau, S., 2005. Evidence for orbitofrontal pathology in patients. The American Journal of Geriatric Psychiatry 18, 172–176.
bipolar disorder and major depression, but not in schizophrenia. Bipolar Dis- Diniz, B.S., Teixeira, A.L., Talib, L.L., Mendonca, V.A., Gattaz, W.F., Forlenza, O.V.,
orders 7, 358–369. 2010b. Serum brain-derived neurotrophic factor level is reduced in antidepres-
Coupland, C., Dhiman, P., Morriss, R., Arthur, A., Barton, G., Hippisley-Cox, J., 2011. sant-free patients with late-life depression. The World Journal of Biological
Antidepressant use and risk of adverse outcomes in older people: population Psychiatry 11, 550–555.
based cohort study. British Medical Journal 343, d4551. Djernes, J.K., Gulmann, N.C., Foldager, L., Olesen, F., Munk-Jørgensen, P., 2011. 13
Culang, M.E., Sneed, J.R., Keilp, J.G., Rutherford, B.R., Pelton, G.H., Devanand, D.P., year follow up of morbidity, mortality and use of health services among elderly
Roose, S.P., 2009. Change in cognitive functioning following acute antidepres- depressed patients and general elderly populations. The Australian and New
sant treatment in late-life depression. The American Journal of Geriatric Psy- Zealand Journal of Psychiatry 45, 654–662.
chiatry 17, 881–888. Doraiswamy, P.M., Krishnan, K.R., Oxman, T., Jenkyn, L.R., Coffey, D.J., Burt, T., Clary,
Cummings, J.L., 1985. Treatable dementias: differential diagnosis and obstacles to C.M., 2003. Does antidepressant therapy improve cognition in elderly depressed
recognition. Clinical Therapeutics 7, 480–486. patients? The Journals of Gerontology. Series A, Biological Sciences and Medical
Cummings, J.L., 1993. Frontal-subcortical circuits and human behavior. Archives of Sciences 58, 1137–1144.
Neurology 50, 873–880. Dotson, V.M., Beydoun, M.A., Zonderman, A.B., 2010. Recurrent depressive symp-
Curran, S.M., Murray, C.M., Van Beck, M., Dougall, N., O’Carroll, R.E., Austin, M.P., toms and the incidence of dementia and mild cognitive impairment. Neurology
Ebmeier, K.P., Goodwin, G.M., 1993. A single photon emission computerised 75, 27–34.
tomography study of regional brain function in elderly patients with major Dotson, V.M., Davatzikos, C., Kraut, M.A., Resnick, S.M., 2009. Depressive symptoms
depression and with Alzheimer-type dementia. The British Journal of Psychiatry and brain volumes in older adults: a longitudinal magnetic resonance imaging
163, 155–165. study. Journal of Psychiatry & Neuroscience 34, 367–375.
D’Amelio, F., Eng, L.F., Gibbs, M.A., 1990. Glutamine synthetase immunoreactivity is Drevets, W.C., 1998. Functional neuroimaging studies of depression: the anatomy of
present in oligodendroglia of various regions of the central nervous system. Glia melancholia. Annual Review of Medicine 49, 341–361.
3, 335–341. Drevets, W.C., 2000. Functional anatomical abnormalities in limbic and prefrontal
D’haenen, H., 2001. Imaging the serotonergic system in depression. European cortical structures in major depression. Progress in Brain Research 126, 413–
Archives of Psychiatry and Clinical Neuroscience 251, II/76–II/80. 431.
Dahabra, S., Ashton, C.H., Bahrainian, M., Britton, P.G., Ferrier, I.N., McAllister, V.A., Drevets, W.C., Price, J.L., Furey, M.L., 2008a. Brain structural and functional abnor-
Marsh, V.R., Moore, P.B., 1998. Structural and functional abnormalities in malities in mood disorders: implications for neurocircuitry models of depres-
elderly patients clinically recovered from early- and late-onset depression. sion. Brain Structure & Function 213, 93–118.
Biological Psychiatry 44, 34–46. Drevets, W.C., Savitz, J., Trimble, M., 2008b. The subgenual anterior cingulate cortex
Dalby, R.B., Chakravarty, M.M., Ahdidan, J., Sorensen, L., Frandsen, J., Jonsdottir, K.Y., in mood disorders. CNS Spectrums 13, 663–681.
Tehrani, E., Rosenberg, R., Ostergaard, L., Videbech, P., 2010. Localization of Driscoll, H.C., Basinski, J., Mulsant, B.H., Butters, M.A., Dew, M.A., Houck, P.R.,
white-matter lesions and effect of vascular risk factors in late-onset major Mazumdar, S., Miller, M.D., Pollock, B.G., Stack, J.A., Schlernitzauer, M.A., Rey-
depression. Psychological Medicine 40, 1389–1399. nolds 3rd, C.F., 2005. Late-onset major depression: clinical and treatment-
Damelio, F., Eng, L.F., Gibbs, M.A., 1990. Glutamine-synthetase immunoreactivity is response variability. International Journal of Geriatric Psychiatry 20, 661–667.
present in oligodendroglia of various regions of the central-nervous-system. Duman, R.S., Monteggia, L.M., Duman, R.S., Monteggia, L.M., 2006. A neurotrophic
Glia 3, 335–341. model for stress-related mood disorders. Biological Psychiatry 59, 1116–1127.
Dantzer, R., O’Connor, J.C., Freund, G.G., Johnson, R.W., Kelley, K.W., 2008. From Ebmeier, K., Rose, E., Steele, D., 2006. Cognitive impairment and fMRI in major
inflammation to sickness and depression: when the immune system subjugates depression. Neurotoxicity Research 10, 87–92.
the brain. Nature Reviews. Neuroscience 9, 46–57. Ebmeier, K.P., Glabus, M.F., Prentice, N., Ryman, A., Goodwin, G.M., 1998. A voxel-
Davignon, J., Ganz, P., 2004. Role of endothelial dysfunction in atherosclerosis. based analysis of cerebral perfusion in dementia and depression of old age.
Circulation 109, III27–III32. NeuroImage 7, 199–208.
Davis, S., Thomas, A., Perry, R., Oakley, A., Kalaria, R.N., O’Brien, J.T., 2002. Glial Edison, P., Archer, H.A., Gerhard, A., Hinz, R., Pavese, N., Turkheimer, F.E., Hammers,
fibrillary acidic protein in late life major depressive disorder: an immunocyto- A., Tai, Y.F., Fox, N., Kennedy, A., Rossor, M., Brooks, D.J., 2008. Microglia,
chemical study. Journal of Neurology, Neurosurgery, and Psychiatry 73, 556– amyloid, and cognition in Alzheimer’s disease: an 11C (R)PK11195-PET and
560. 11C PIB-PET study. Neurobiology of Disease 32, 412–419.
de Asis, J.M., Stern, E., Alexopoulos, G.S., Pan, H., Van Gorp, W., Blumberg, H., Egan, M.F., Kojima, M., Callicott, J.H., Goldberg, G.E., Kolachana, B.S., Bertolino, A.,
Kalayam, B., Eidelberg, D., Kiosses, D., Silbersweig, D.A., 2001. Hippocampal Zaitsev, E., Gold, B., Goldman, D., Dean, M., Lu, B., Weinberger, D.R., 2003. The
and anterior cingulate activation deficits in patients with geriatric depression. BDNF val66met polymorphism affects activity-dependent secretion of BDNF
The American Journal of Psychiatry 158, 1321–1323. and human memory and hippocampal function. Cell 112, 257–269.
de Groot, J.C., de Leeuw, F., Oudkerk, M., Hofman, A., Jolles, J., Breteler, M.M.B., 2000. Egger, K., Schocke, M., Weiss, E., Auffinger, S., Esterhammer, R., Goebel, G., Walch, T.,
Cerebral white matter lesions and depressive symptoms in elderly adults. Mechtcheriakov, S., Marksteiner, J., 2008. Pattern of brain atrophy in elderly
Archives of General Psychiatry 57, 1071–1076. patients with depression revealed by voxel-based morphometry. Psychiatry
de Vries, H.E., Blom-Roosemalen, M.C.M., van Oosten, M., de Boer, A.G., van Berkel, Research 164, 237–244.
T.J.C., Breimer, D.D., Kuiper, J., 1996. The influence of cytokines on the Elderkin-Thompson, V., Hellemann, G., Pham, D., Kumar, A., 2009. Prefrontal brain
integrity of the blood-brain barrier in vitro. Journal of Neuroimmunology morphology and executive function in healthy and depressed elderly. Interna-
64, 37–43. tional Journal of Geriatric Psychiatry 24, 459–468.
Delaloye, C., Baudois, S., de Bilbao, F., Dubois Remund, C., Hofer, F., Lamon, M., Ragno Elderkin-Thompson, V., Kumar, A., Bilker, W.B., Dunkin, J.J., Mintz, J., Moberg, P.J.,
Paquier, C., Weber, K., Herrmann, F.R., Giardini, U., Giannakopoulos, P., 2008. Mesholam, R.I., Gur, R.E., 2003. Neuropsychological deficits among patients
Cognitive impairment in late-onset depression. Limited to a decrement in with late-onset minor and major depression. Archives of Clinical Neuropsy-
information processing resources? European Neurology 60, 149–154. chology 18, 529–549.
DeLuca, A.K., Lenze, E.J., Mulsant, B.H., Butters, M.A., Karp, J.F., Dew, M.A., Pollock, Empana, J.P., Sykes, D.H., Luc, G., Juhan-Vague, I., Arveiler, D., Ferrieres, J., Amouyel,
B.G., Shear, M.K., Houck, P.R., Reynolds 3rd, C.F., 2005. Comorbid anxiety P., Bingham, A., Montaye, M., Ruidavets, J.B., Haas, B., Evans, A., Jouven, X.,
disorder in late life depression: association with memory decline over four Ducimetiere, P., Grp, P.S., 2005. Contributions of depressive mood and circulat-
years. International Journal of Geriatric Psychiatry 20, 848–854. ing inflammatory markers to coronary heart disease in healthy European men –
den Heijer, T., Tiemeier, H., Luijendijk, H.J., van der Lijn, F., Koudstaal, P.J., Hofman, the prospective epidemiological study of myocardial infarction (PRIME). Circu-
A., Breteler, M.M.B., 2011. A study of the bidirectional association between lation 111, 2299–2305.
hippocampal volume on magnetic resonance imaging and depression in the Exner, C., Koschack, J., Irle, E., 2002. The differential role of premotor frontal cortex
elderly. Biological Psychiatry 70, 191–197. and basal ganglia in motor sequence learning: evidence from focal basal ganglia
Dentino, A.N., Pieper, C.F., Rao, K.M.K., Currie, M.S., Harris, T., Blazer, D.G., Cohen, lesions. Learning & Memory 9, 376–386.
H.J., 1999. Association of interleukin-6 and other biologic variables with de- Exner, C., Lange, C., Irle, E., 2009. Impaired implicit learning and reduced pre-
pression in older people living in the community. Journal of the American supplementary motor cortex size in early-onset major depression with melan-
Geriatrics Society 47, 6–11. cholic features. Journal of Affective Disorders 119, 156–162.
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 135

Farrer, L.A., Cupples, L.A., Haines, J.L., Hyman, B., Kukull, W.A., Mayeux, R., Myers, Gillespie, N.A., Kirk, K.M., Evans, D.M., Heath, A.C., Hickie, I.B., Martin, N.G., 2004. Do
R.H., PericakVance, M.A., Risch, N., vanDuijn, C.M., 1997. Effects of age, sex, and the genetic or environmental determinants of anxiety and depression change
ethnicity on the association between apolipoprotein E genotype and Alzheimer with age? A longitudinal study of Australian twins. Twin Research 7, 39–53.
disease – a meta-analysis. JAMA: the Journal of the American Medical Associa- Godin, O., Dufouil, C., Maillard, P., Delcroix, N., Mazoyer, B., Crivello, F., Alperovitch,
tion 278, 1349–1356. A., Tzourio, C., 2008. White matter lesions as a predictor of depression in the
Fazekas, F., Chawluk, J.B., Alavi, A., Hurtig, H.I., Zimmerman, R.A., 1987. MR signal elderly: the 3C-Dijon study. Biological Psychiatry 63, 663–669.
abnormalities at 1.5 T in Alzheimer’s dementia and normal aging. AJR. American Goodwin, G.M., 1997. Neuropsychological and neuroimaging evidence for the
Journal of Roentgenology 149, 351–356. involvement of the frontal lobes in depression. Journal of Psychopharmacology
Ferrucci, L., Corsi, A., Lauretani, F., Bandinelli, S., Bartali, B., Taub, D.D., Guralnik, J.M., 11, 115–122.
Longo, D.L., 2005. The origins of age-related proinflammatory state. Blood 105, Gouin, J.P., Hantsoo, L., Kiecolt-Glaser, J.K., 2008. Immune dysregulation and
2294–2299. chronic stress among older adults: a review. Neuroimmunomodulation 15,
Figiel, G.S., Krishnan, K.R., Doraiswamy, P.M., Nemeroff, C.B., 1991a. Caudate 251–259.
hyperintensities in elderly depressed patients with neuroleptic-induced par- Granowitz, E.V., Santos, A.A., Poutsiaka, D.D., Cannon, J.G., Wilmore, D.W., Wolff,
kinsonism. Journal of Geriatric Psychiatry and Neurology 4, 86–89. S.M., Dinarello, C.A., 1991. Production of interleukin-1-receptor antagonist
Figiel, G.S., Krishnan, K.R., Doraiswamy, P.M., Rao, V.P., Nemeroff, C.B., Boyko, O.B., during experimental endotoxemia. Lancet 338, 1423–1424.
1991b. Subcortical hyperintensities on brain magnetic resonance imaging: a Greenstein, A.S., Paranthaman, R., Burns, A., Jackson, A., Malik, R.A., Baldwin, R.C.,
comparison between late age onset and early onset elderly depressed subjects. Heagerty, A.M., 2010. Cerebrovascular damage in late-life depression is associ-
Neurobiology of Aging 12, 245–247. ated with structural and functional abnormalities of subcutaneous small arter-
Filippi, M., Agosta, F., 2007. Magnetization transfer MRI in multiple sclerosis. Journal ies. Hypertension 56, 734–740.
of Neuroimaging 17, 22S–26S. Greenwald, B.S., Kramer-Ginsberg, E., Bogerts, B., Ashtari, M., Aupperle, P., Wu, H.,
Firbank, M.J., Lloyd, A.J., Ferrier, N., O’Brien, J.T., 2004. A volumetric study of MRI Allen, L., Zeman, D., Patel, M., 1997. Qualitative magnetic resonance imaging
signal hyperintensities in late-life depression. The American Journal of Geriatric findings in geriatric depression. Possible link between later-onset depression
Psychiatry 12, 606–612. and Alzheimer’s disease? Psychological Medicine 27, 421–431.
Firbank, M.J., Narayan, S.K., Saxby, B.K., Ford, G.A., O’Brien, J.T., 2010. Homocysteine Gregory-Roberts, E.M., Naismith, S.L., Cullen, K.M., Hickie, I.B., 2010. Electroconvul-
is associated with hippocampal and white matter atrophy in older subjects with sive therapy-induced persistent retrograde amnesia: could it be minimised by
mild hypertension. International Psychogeriatrics 22, 804–811. ketamine or other pharmacological approaches? Journal of Affective Disorders
Folstein, M., Liu, T., Peter, I., Buel, J., Arsenault, L., Scott, T., Qiu, W.W., 2007. The 126, 39–45.
homocysteine hypothesis of depression. The American Journal of Psychiatry Gunning-Dixon, F.M., Brickman, A.M., Cheng, J.C., Alexopoulos, G.S., 2009. Aging of
164, 861–867. cerebral white matter: a review of MRI findings. International Journal of
Forester, B.P., Harper, D.G., Jensen, J.E., Ravichandran, C., Jordan, B., Renshaw, P.F., Geriatric Psychiatry 24, 109–117.
Cohen, B.M., 2009. 31Phosphorus magnetic resonance spectroscopy study of Gunning-Dixon, F.M., Hoptman, M.J., Lim, K.O., Murphy, C.F., Klimstra, S., Latous-
tissue specific changes in high energy phosphates before and after sertraline sakis, V., Majcher-Tascio, M., Hrabe, J., Ardekani, B.A., Alexopoulos, G.S., 2008.
treatment of geriatric depression. International Journal of Geriatric Psychiatry Macromolecular white matter abnormalities in geriatric depression: a magne-
24, 788–797. tization transfer imaging study. The American Journal of Geriatric Psychiatry 16,
Forkstam, C., Petersson, K.M., 2005. Towards an explicit account of implicit learning. 255–262.
Current Opinion in Neurology 18, 435–441. Gunning-Dixon, F.M., Walton, M., Cheng, J., Acuna, J., Klimstra, S., Zimmerman, M.E.,
Forti, P., Rietti, E., Pisacane, N., Olivelli, V., Mariani, E., Chiappelli, M., Licastro, F., Brickman, A.M., Hoptman, M.J., Young, R.C., Alexopoulos, G.S., 2010. MRI signal
Ravaglia, G., 2010. Blood inflammatory proteins and risk of incident depression hyperintensities and treatment remission of geriatric depression. Journal of
in the elderly. Dementia and Geriatric Cognitive Disorders 29, 11–20. Affective Disorders 126, 395–401.
Frodl, T., Jager, M., Smajstrlova, I., Born, C., Bottlender, R., Palladino, T., Reiser, M., Gunning, F.M., Cheng, J., Murphy, C.F., Kanellopoulos, D., Acuna, J., Hoptman, M.J.,
Moller, H.J., Meisenzahl, E.M., 2008. Effect of hippocampal and amygdala Klimstra, S., Morimoto, S., Weinberg, J., Alexopoulos, G.S., 2009. Anterior
volumes on clinical outcomes in major depression: a 3-year prospective mag- cingulate cortical volumes and treatment remission of geriatric depression.
netic resonance imaging study. Journal of Psychiatry & Neuroscience 33, 423– International Journal of Geriatric Psychiatry 24, 829–836.
430. Hackett, M.L., Anderson, C.S., House, A., Xia, J., 2008. Interventions for treating
Frodl, T., Meisenzahl, E.M., Zetzsche, T., Born, C., Jager, M., Groll, C., Bottlender, R., depression after stroke. Cochrane Database of Systematic Reviews 4 Art. no.:
Leinsinger, G., Moller, H.J., 2003. Larger amygdala volumes in first depressive CD003437.
episode as compared to recurrent major depression and healthy control sub- Hamidi, M., Drevets, W.C., Price, J.L., 2004. Glial reduction in amygdala in major
jects. Biological Psychiatry 53, 338–344. depressive disorder is due to oligodendrocytes. Biological Psychiatry 55, 563–
Frodl, T., Meisenzahl, E.M., Zetzsche, T., Hohne, T., Banac, S., Schorr, C., Jager, M., 569.
Leinsinger, G., Bottlender, R., Reiser, M., Moller, H.J., 2004. Hippocampal and Hamilton, B.A., Naismith, S.L., Scott, E.M., Purcell, S., Hickie, I.B., 2011. Disability is
amygdala changes in patients with major depressive disorder and healthy already pronounced in young people with early stages of affective disorders:
controls during a 1-year follow-up. The Journal of Clinical Psychiatry 65, data from an early intervention service. Journal of Affective Disorders 131, 84–
492–499. 91.
Fujimoto, T., Takeuchi, K., Matsumoto, T., Fujita, S., Honda, K., Higashi, Y., Kato, N., Hamilton, J.P., Siemer, M., Gotlib, I.H., 2008. Amygdala volume in major depressive
2008. Metabolic changes in the brain of patients with late-onset major depres- disorder: a meta-analysis of magnetic resonance imaging studies. Molecular
sion. Psychiatry Research: Neuroimaging 164, 48–57. Psychiatry 13, 993–1000.
Gareri, P., De Fazio, P., De Sarro, G., 2002. Neuropharmacology of depression in aging Hannestad, J., Taylor, W.D., McQuoid, D.R., Payne, M.E., Krishnan, K.R.R., Steffens,
and age-related diseases. Ageing Research Reviews 1, 113–134. D.C., MacFall, J.R., 2006. White matter lesion volumes and caudate volumes in
Gass, P., Hellweg, R., 2010. Peripheral brain-derived neurotrophic factor (BDNF) as a late-life depression. International Journal of Geriatric Psychiatry 21, 1193–
biomarker for affective disorders? The International Journal of Neuropsycho- 1198.
pharmacology 13, 1–4. Hansell, N.K., Wright, M.J., Medland, S.E., Davenport, T.A., Wray, N.R., Martin, N.G.,
Gatt, J.M., Nemeroff, C.B., Dobson-Stone, C., Paul, R.H., Bryant, R.A., Schofield, P.R., Hickie, I.B., 2011. Genetic co-morbidity between neuroticism, anxiety/depres-
Gordon, E., Kemp, A.H., Williams, L.M., 2009. Interactions between BDNF sion and somatic distress in a population sample of adolescent and young adult
Val66Met polymorphism and early life stress predict brain and arousal path- twins. Psychological Medicine 42, 1249–1260.
ways to syndromal depression and anxiety. Molecular Psychiatry 14, 681–695. Hardy, S.E., 2009. Methylphenidate for the treatment of depressive symptoms,
Gerhard, A., Pavese, N., Hotton, G., Turkheimer, F., Es, M., Hammers, A., Eggert, K., including fatigue and apathy, in medically ill older adults and terminally ill
Oertel, W., Banati, R.B., Brooks, D.J., 2006a. In vivo imaging of microglial adults. The American Journal of Geriatric Pharmacotherapy 7, 34–59.
activation with C-11 (R)-PK11195 PET in idiopathic Parkinson’s disease. Neu- Harris, M., Glozier, N., Ratnavadivel, R., Grunstein, R.R., 2009. Obstructive sleep
robiology of Disease 21, 404–412. apnea and depression. Sleep Medicine Reviews 13, 437–444.
Gerhard, A., Trender-Gerhard, I., Turkheimer, F., Quinn, N.P., Bhatia, K.P., Brooks, D.J., Hedayati, S.S., Yalamanchili, V., Finkelstein, F.O., 2012. A practical approach to the
2006b. In vivo imaging of microglial activation with C-11 (R)-PK11195 PET in treatment of depression in patients with chronic kidney disease and end-stage
progressive supranuclear palsy. Movement Disorders 21, 89–93. renal disease. Kidney International 81, 247–255.
Gerretsen, P., Pollock, B.G., 2008. Pharmacogenetics and the serotonin transporter in Heiden, A., Kettenbach, J., Fischer, P., Schein, B., Ba-Ssalamah, A., Frey, R., Naderi,
late-life depression. Expert Opinion on Drug Metabolism 4, 1465–1478. M.M., Gulesserian, T., Schmid, D., Trattnig, S., Imhof, H., Kasper, S., 2005. White
Gerritsen, L., Comijs, H.C., van der Graaf, Y., Knoops, A.J.G., Penninx, B.W.J.H., matter hyperintensities and chronicity of depression. Journal of Psychiatric
Geerlings, M.I., 2011. Depression hypothalamic pituitary adrenal axis, and Research 39, 285–293.
hippocampal and entorhinal cortex volumes: the SMART Medea Study. Biolog- Heils, A., Teufel, A., Petri, S., Seemann, M., Bengel, D., Balling, U., Riederer, P., Lesch,
ical Psychiatry 70, 373–380. K.P., 1995. Functional promoter and polyadenylation site mapping of the
Gerstner, J.R., Yin, J.C., 2010. Circadian rhythms and memory formation. Nature human serotonin (5-HT) transporter gene. Journal of Neural Transmission -
Reviews. Neuroscience 11, 577–588. General Section 102, 247–254.
Ghiadoni, L., Donald, A.E., Cropley, M., Mullen, M.J., Oakley, G., Taylor, M., O’Connor, Hepple, J., 2004. Psychotherapies with older people: an overview. Advances in
G., Betteridge, J., Klein, N., Steptoe, A., Deanfield, J.E., 2000. Mental stress induces Psychiatric Treatment 10, 371–377.
transient endothelial dysfunction in humans. Circulation 102, 2473–2478. Hermens, D.F., Hodge, M.A.R., Naismith, S.L., Kaur, M., Scott, E., Hickie, I.B., 2011.
Giedd, J.N., Blumenthal, J., Jeffries, N.O., Castellanos, F.X., Liu, H., Zijdenbos, A., Paus, Neuropsychological clustering highlights cognitive differences in young people
T., Evans, A.C., Rapoport, J.L., 1999. Brain development during childhood and presenting with depressive symptoms. Journal of International Neuropsycho-
adolescence: a longitudinal MRI study. Nature Neuroscience 2, 861–863. logical Society 17, 267–276.
136 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

Herrmann, L.L., Goodwin, G.M., Ebmeier, K.P., 2007. The cognitive neuropsychology Hong, E.D., Taylor, W.D., McQuoid, D.R., Potter, G.G., Payne, M.E., Ashley-Koch, A.,
of depression in the elderly. Psychological Medicine 37, 1693–1702. Steffens, D.C., 2009. Influence of the MTHFR C677T polymorphism on magnetic
Herrmann, L.L., Le Masurier, M., Ebmeier, K.P., 2008. White matter hyperintensities resonance imaging hyperintensity volume and cognition in geriatric depres-
in late life depression: a systematic review. Journal of Neurology, Neurosurgery, sion. The American Journal of Geriatric Psychiatry 17, 847–855.
and Psychiatry 79, 619–624. Hoptman, M.J., Gunning-Dixon, F.M., Murphy, C.F., Lim, K.O., Alexopoulos, G.S.,
Hickie, I., Lloyd, A., Dixon, G., Halliday, G., McRitchie, D., Scott, E., Mitchell, P., 2006. Structural neuroimaging research methods in geriatric depression. The
Wakefield, D., 1997a. Utilising molecular biological and histopathological American Journal of Geriatric Psychiatry 14, 812–822.
techniques to study the dopaminergic system in patients with melancholia. Hsieh, M.H., McQuoid, D.R., Levy, R.M., Payne, M.E., MacFall, J.R., Steffens, D.C., 2002.
The Australian and New Zealand Journal of Psychiatry 31, 27–35. Hippocampal volume and antidepressant response in geriatric depression.
Hickie, I., Mason, C., Parker, G., Brodaty, H., 1996. Prediction of ECT response: International Journal of Geriatric Psychiatry 17, 519–525.
validation of a refined sign-based (CORE) system for defining melancholia. The Hwang, J.P., Tsai, S.J., Hong, C.J., Yang, C.H., Lirng, J.F., Yang, Y.M., 2006. The Val66Met
British Journal of Psychiatry 169, 68–74. polymorphism of the brain-derived neurotrophic-factor gene is associated with
Hickie, I., Naismith, S., Ward, P.B., Scott, E., Mitchell, P., Wilhelm, K., Parker, G., geriatric depression. Neurobiology of Aging 27, 1834–1837.
2005a. Vascular risk and low serum B12 predict white matter lesions in patients Hybels, C.F., Blazer, D.G., Pieper, C.F., Landerman, L.R., Steffens, D.C., 2009. Profiles
with major depression. Journal of Affective Disorders 85, 327–332. of depressive symptoms in older adults diagnosed with major depression:
Hickie, I., Naismith, S., Ward, P.B., Turner, K., Scott, E., Mitchell, P., Wilhelm, K., latent cluster analysis. The American Journal of Geriatric Psychiatry 17, 387–
Parker, G., 2005b. Reduced hippocampal volumes and memory loss in patients 396.
with early- and late-onset depression. The British Journal of Psychiatry 186, Ikram, M.K., De Jong, F.J., Van Dijk, E.J., Prins, N.D., Hofman, A., Breteler, M.M., De
197–202. Jong, P.T., 2006. Retinal vessel diameters and cerebral small vessel disease: the
Hickie, I., Parsonage, B., Parker, G., 1990a. Prediction of response to electroconvul- Rotterdam Scan Study. Brain 129, 182–188.
sive therapy. Preliminary validation of a sign-based typology of depression. The Ikram, M.K., de Jong, F.J., Vingerling, J.R., Witteman, J.C., Hofman, A., Breteler, M.M.,
British Journal of Psychiatry 157, 65–71. de Jong, P.T., 2004. Are retinal arteriolar or venular diameters associated with
Hickie, I., Scott, E., 1998. Late-onset depressive disorders: a preventable variant of markers for cardiovascular disorders? The Rotterdam Study. Investigative
cerebrovascular disease? Psychological Medicine 28, 1007–1013. Ophthalmology & Visual Science 45, 2129–2134.
Hickie, I., Scott, E., Mitchell, P., Wilhelm, K., Austin, M.P., Bennett, B., 1995. Subcor- Ikram, M.K., Luijendijk, H.J., Hofman, A., de Jong, P.T.V.M., Breteler, M.M.B., Vinger-
tical hyperintensities on magnetic resonance imaging: clinical correlates and ling, J.R., Tiemeier, H., 2010. Retinal vascular calibers and risk of late-life
prognostic significance in patients with severe depression. Biological Psychiatry depression: the Rotterdam Study. The American Journal of Geriatric Psychiatry
37, 151–160. 18, 452–456.
Hickie, I., Scott, E., Naismith, S., Ward, P.B., Turner, K., Parker, G., Mitchell, P., Insel, T.R., 2009a. Disruptive insights in psychiatry: transforming a clinical disci-
Wilhelm, K., 2001a. Late-onset depression: genetic, vascular and clinical con- pline. The Journal of Clinical Investigation 119, 700–705.
tributions. Psychological Medicine 13, 1403–1412. Insel, T.R., 2009b. Translating scientific opportunity into public health impact: a
Hickie, I., Scott, E., Wilhelm, K., Brodaty, H., 1997b. Subcortical hyperintensities on strategic plan for research on mental illness. Archives of General Psychiatry 66,
magnetic resonance imaging in patients with severe depression – a longitudinal 128–133.
evaluation. Biological Psychiatry 42, 367–374. Inzitari, D., Simoni, M., Pracucci, G., Poggesi, A., Basile, A.M., Chabriat, H., Erkinjuntti,
Hickie, I., Silove, D., Hickie, C., Wakefield, D., Lloyd, A., 1990b. Is there immune T., Fazekas, F., Ferro, J.M., Hennerici, M., Langhorne, P., O’Brien, J., Barkhof, F.,
dysfunction in depressive disorders? Psychological Medicine 20, 755–761. Visser, M.C., Wahlund, L.O., Waldemar, G., Wallin, A., Pantoni, L., Grp, L.S., 2007.
Hickie, I., Simons, L., Naismith, S., Simons, J., McCallum, J., Pearson, K., 2003. Vascular Risk of rapid global functional decline in elderly patients with severe cerebral
risk to late-life depression: evidence from a longitudinal community study. The age-related white matter changes – The LADIS study. Archives of Internal
Australian and New Zealand Journal of Psychiatry 37, 62–65. Medicine 167, 81–88.
Hickie, I., Ward, P., Scott, E., Haindl, W., Walker, B., Dixon, J., Turner, K., 1999. Neo- Iosifescu, D.V., Nierenberg, A.A., Alpert, J.E., Smith, M., Bitran, S., Dording, C., Fava,
striatal rCBF correlates of psychomotor slowing in patients with major depres- M., 2003. The impact of medical comorbidity on acute treatment in major
sion. Psychiatry Research 92, 75–81. depressive disorder. The American Journal of Psychiatry 160, 2122–2127.
Hickie, I.B., 2004. Reducing the burden of depression: are we making progress in Ishizaki, J., Yamamoto, H., Takahashi, T., Takeda, M., Yano, M., Mimura, M., 2008.
Australia? The Medical Journal of Australia 181, S4–S5. Changes in regional cerebral blood flow following antidepressant treatment
Hickie, I.B., 2011. Antidepressants in elderly people. British Medical Journal 343, in late-life depression. International Journal of Geriatric Psychiatry 23, 805–
d4660. 811.
Hickie, I.B., Davenport, T.A., Luscombe, G.M., 2006. Mental health expenditure in Javitt, D.C., Schoepp, D., Kalivas, P.W., Volkow, N.D., Zarate, C., Merchant, K., Bear,
Australia: time for affirmative action. Australian and New Zealand Journal of M.F., Umbricht, D., Hajos, M., Potter, W.Z., Lee, C.M., 2011. Translating gluta-
Public Health 30, 119–122. mate: from pathophysiology to treatment. Science Translational Medicine 3,
Hickie, I.B., Davenport, T.A., Naismith, S.L., Scott, E.M., Hadzi-Pavlovic, D., Koschera, 102mr2.
A., 2001b. Treatment of common mental disorders in Australian general prac- Joe, A.Y., Tielmann, T., Bucerius, J., Reinhardt, M.J., Palmedo, H., Maier, W., Biersack,
tice. The Medical Journal of Australia 175, 16. H.J., Zobel, A., 2006. Response-dependent differences in regional cerebral blood
Hickie, I.B., Groom, G.L., McGorry, P.D., Davenport, T.A., Luscombe, G.M., 2005c. flow changes with citalopram in treatment of major depression. Journal of
Australian mental health reform: time for real outcomes. The Medical Journal of Nuclear Medicine 47, 1319–1325.
Australia 182, 401–406. Johnson, L.R., Farb, C., Morrison, J.H., McEwen, B.S., LeDoux, J.E., 2005. Localization of
Hickie, I.B., Naismith, S.L., Norrie, L.M., Scott, E.M., 2009. Managing depression glucocorticoid receptors at postsynaptic membranes in the lateral amygdala.
across the life cycle: new strategies for clinicians and their patients. Internal Neuroscience 136, 289–299.
Medicine Journal 39, 720–727. Jones, J.S., Stanley, B., Mann, J.J., Frances, A.J., Guido, J.R., Traskman-Bendz, L.,
Hickie, I.B., Naismith, S.L., Ward, P.B., Little, C.L., Pearson, M., Scott, E.M., Mitchell, P., Winchel, R., Brown, R.P., Stanley, M., 1990. CSF 5-HIAA and HVA concentrations
Wilhelm, K., Parker, G., 2007a. Psychomotor slowing in older patients with in elderly depressed patients who attempted suicide. The American Journal of
major depression: relationships with blood flow in the caudate nucleus and Psychiatry 147, 1225–1227.
white matter lesions. Psychiatry Research: Neuroimaging 155, 211–220. Jorge, R.E., Moser, D.J., Acion, L., Robinson, R.G., 2008. Treatment of vascular
Hickie, I.B., Naismith, S.L., Ward, P.B., Scott, E.M., Mitchell, P.B., Schofield, P.R., depression using repetitive transcranial magnetic stimulation. Archives of
Scimone, A., Wilhelm, K., Parker, G., 2007b. Serotonin transporter gene status General Psychiatry 65, 268–276.
predicts caudate nucleus but not amygdala or hippocampal volumes in older Jorm, A.F., Vanduijn, C.M., Chandra, V., Fratiglioni, L., Graves, A.B., Heyman, A.,
persons with major depression. Journal of Affective Disorders 98, 137–142. Kokmen, E., Kondo, K., Mortimer, J.A., Rocca, W.A., Shalat, S.L., Soininen, H.,
Hickie, I.B., Rogers, N.L., 2011. Novel melatonin-based therapies: potential advances Hofman, A., 1991. Psychiatric history and related exposures as risk-factors for
in the treatment of major depression. Lancet 378, 621–631. Alsheimers-disease – a collaborative reanalysis of case-control studies. Inter-
Hickie, I.B., Scott, E.M., Hermens, D.F., Naismith, S.L., Guastella, A.J., Kaur, M., Sidis, national Journal of Epidemiology 20, S43–S47.
A., Whitwell, B., Glozier, N., Pantelis, C., Wood, S., McGorry, P. Applying a clinical Kamholz, B., Unutzer, J., 2007. Depression after hip fracture. Journal of the American
staging framework in young people who present with admixtures of anxious, Geriatrics Society 55, 126–127.
depressive or psychotic symptoms. Early Intervention in Psychiatry, in press. Kanellopoulos, D., Gunning, F.M., Morimoto, S.S., Hoptman, M.J., Murphy, C.F., Kelly,
Highet, N.J., Hickie, I.B., Davenport, T.A., 2002. Monitoring awareness of and R.E., Glatt, C., Lim, K.O., Alexopoulos, G.S., 2011. Hippocampal volumes and the
attitudes to depression in Australia. The Medical Journal of Australia 176 brain-derived neurotrophic factor val66met polymorphism in geriatric major
(Suppl.), S63–S68. depression. The American Journal of Geriatric Psychiatry 19, 13–22.
Highet, N.J., Luscombe, G.M., Davenport, T.A., Burns, J.M., Hickie, I.B., 2006. Positive Katon, W.J., 2003. Clinical and health services relationships between major depres-
relationships between public awareness activity and recognition of the impacts sion, depressive symptoms, and general medical illness. Biological Psychiatry
of depression in Australia. The Australian and New Zealand Journal of Psychia- 54, 216–226.
try 40, 55–58. Kay, D.W.K., Roth, M., Hopkins, B., 1955. Affective disorders arising in the senium.1.
Holsboer, F., 2000. The corticosteroid receptor hypothesis of depression. Neurop- Their association with organic cerebral degeneration. Journal of Mental Science
sychopharmacology 23, 477–501. 101, 302–316.
Holtzheimer, P., Meeks, T.W., Kelley, M.E., Mufti, M., Young, R., McWhorter, K., Vito, Keller, J., Flores, B., Gomez, R.G., Solvason, H.B., Kenna, H., Williams, G.H., Schatz-
N., Chismar, R., Quinn, S., Dey, S., Byrd, E.H., McDonald, W.M., 2008. A double berg, A.F., 2006. Cortisol circadian rhythm alterations in psychotic major
blind, placebo-controlled pilot study of galantamine augmentation of antide- depression. Biological Psychiatry 60, 275–281.
pressant treatment in older adults with major depression. International Journal Kendler, K.S., Fiske, A., Gardner, C.O., Gatz, M., 2009. Delineation of two genetic
of Geriatric Psychiatry 23, 625–631. pathways to major depression. Biological Psychiatry 65, 808–811.
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 137

Kendler, K.S., Gardner, C.O., Lichtenstein, P., 2008. A developmental twin study of Krishnan, K.R., Hays, J.C., Tupler, L.A., George, L.K., Blazer, D.G., 1995. Clinical and
symptoms of anxiety and depression: evidence for genetic innovation and phenomenological comparisons of late-onset and early-onset depression. The
attenuation. Psychological Medicine 38, 1567–1575. American Journal of Psychiatry 152, 785–788.
Kendler, K.S., Kuhn, J.W., Vittum, J., Prescott, C.A., Riley, B., 2005a. The interaction of Krishnan, K.R., McDonald, W.M., 1995. Arteriosclerotic depression. Medical Hy-
stressful life events and a serotonin transporter polymorphism in the prediction potheses 44, 111–115.
of episodes of major depression – a replication. Archives of General Psychiatry Krishnan, K.R., McDonald, W.M., Doraiswamy, P.M., Tupler, L.A., Husain, M., Boyko,
62, 529–535. O.B., Figiel, G.S., Ellinwood Jr., E.H., 1993. Neuroanatomical substrates of de-
Kendler, K.S., Kuhn, J.W., Vittum, J., Prescott, C.A., Riley, B., 2005b. The interaction of pression in the elderly. European Archives of Psychiatry and Clinical Neurosci-
stressful life events and a serotonin transporter polymorphism in the prediction ence 243, 41–46.
of episodes of major depression: a replication. Archives of General Psychiatry Krishnan, K.R., McDonald, W.M., Escalona, P.R., Doraiswamy, P.M., Na, C., Husain,
62, 529–535. M.M., Figiel, G.S., Boyko, O.B., Ellinwood, E.H., Nemeroff, C.B., 1992. Magnetic
Kennedy, K.M., Raz, N., 2009. Aging white matter and cognition: differential effects resonance imaging of the caudate nuclei in depression. Preliminary observa-
of regional variations in diffusion properties on memory, executive functions, tions. Archives of General Psychiatry 49, 553–557.
and speed. Neuropsychologia 47, 916–927. Krishnan, K.R., Taylor, W.D., McQuoid, D.R., MacFall, J.R., Payne, M.E., Provenzale,
Kenny, E.R., O’Brien, J.T., Cousins, D.A., Richardson, J., Thomas, A.J., Firbank, M.J., J.M., Steffens, D.C., 2004. Clinical characteristics of magnetic resonance
Blamire, A.M., 2010. Functional connectivity in late-life depression using rest- imaging-defined subcortical ischemic depression. Biological Psychiatry 55,
ing-state functional magnetic resonance imaging. The American Journal of 390–397.
Geriatric Psychiatry 18, 643–651. Krishnan, K.R., Tupler, L.A., Ritchie Jr., J.C., McDonald, W.M., Knight, D.L., Nemeroff,
Kessler, R.C., Birnbaum, H., Bromet, E., Hwang, I., Sampson, N., Shahly, V., 2010. Age C.B., Carroll, B.J., 1996. Apolipoprotein E-epsilon 4 frequency in geriatric de-
differences in major depression: results from the National Comorbidity Survey pression. Biological Psychiatry 40, 69–71.
Replication (NCS-R). Psychological Medicine 40, 225–237. Krishnan, V., Nestler, E.J., 2010. Linking molecules to mood: new insight into the
Khundakar, A., Morris, C., Oakley, A., McMeekin, W., Thomas, A.J., 2009. Morpho- biology of depression. The American Journal of Psychiatry 167, 1305–1320.
metric analysis of neuronal and glial cell pathology in the dorsolateral prefron- Kumar, A., Bilker, W., Jin, Z., Udupa, J., 2000a. Atrophy and high intensity lesions:
tal cortex in late-life depression. The British Journal of Psychiatry 195, 163–169. complementary neurobiological mechanisms in late-life major depression.
Khundakar, A., Morris, C., Oakley, A., Thomas, A.J., 2011. Morphometric analysis of Neuropsychopharmacology 22, 264–274.
neuronal and glial cell pathology in the caudate nucleus in late-life depression. Kumar, A., Bilker, W., Jin, Z., Udupa, J., Gottlieb, G., 1999. Age of onset of depression
The American Journal of Geriatric Psychiatry 19, 132–141. and quantitative neuroanatomic measures: absence of specific correlates.
Kiecolt-Glaser, J.K., Preacher, K.J., MacCallum, R.C., Atkinson, C., Malarkey, W.B., Psychiatry Research 91, 101–110.
Glaser, R., 2003. Chronic stress and age-related increases in the proinflamma- Kumar, A., Bilker, W., Lavretsky, H., Gottlieb, G., 2000b. Volumetric asymmetries in
tory cytokine IL-6. Proceedings of the National Academy of Sciences of the late-onset mood disorders: an attenuation of frontal asymmetry with depres-
United States of America 100, 9090–9095. sion severity. Psychiatry Research 100, 41–47.
Kim, D.H., Payne, M.E., Levy, R.M., MacFall, J.R., Steffens, D.C., 2002. APOE genotype Kumar, A., Gupta, R.C., Albert Thomas, M., Alger, J., Wyckoff, N., Hwang, S., 2004.
and hippocampal volume change in geriatric depression. Biological Psychiatry Biophysical changes in normal-appearing white matter and subcortical nuclei
51, 426–429. in late-life major depression detected using magnetization transfer. Psychiatry
Kim, J.-M., Stewart, R., Kim, S.-W., Yang, S.-J., Shin, I.-S., Yoon, J.-S., 2006. Vascular Research 130, 131–140.
risk factors and incident late-life depression in a Korean population. The British Kumar, A., Jin, Z., Bilker, W., Udupa, J., Gottlieb, G., 1998. Late-onset minor and major
Journal of Psychiatry 189, 26–30. depression: early evidence for common neuroanatomical substrates detected
Kim, J.M., Stewart, R., Kim, S.W., Yang, S.J., Shin, I.S., Yoon, J.S., 2009. Modification by by using MRI. Proceedings of the National Academy of Sciences of the United
two genes of associations between general somatic health and incident de- States of America 95, 7654–7658.
pressive syndrome in older people. Psychosomatic Medicine 71, 286–291. Kumar, A., Kepe, V., Barrio, J.R., Siddarth, P., Manoukian, V., Elderkin-Thompson, V.,
Kim, J.M., Stewart, R., Kim, S.W., Yang, S.J., Shina, I.S., Kim, Y.H., Yoon, J.S., 2008. BDNF Small, G.W., 2011. Protein binding in patients with late-life depression.
genotype potentially modifying the association between incident stroke and Archives of General Psychiatry 68, 1143–1150.
depression. Neurobiology of Aging 29, 789–792. Kumar, A., Newberg, A., Alavi, A., Berlin, J., Smith, R., Reivich, M., 1993. Regional
Kiosses, D.N., Alexopoulos, G.S., Murphy, C., 2000. Symptoms of striatofrontal cerebral glucose metabolism in late-life depression and Alzheimer disease: a
dysfunction contribute to disability in geriatric depression. International Jour- preliminary positron emission tomography study. Proceedings of the National
nal of Geriatric Psychiatry 15, 992–999. Academy of Sciences of the United States of America 90, 7019–7023.
Kiosses, D.N., Klimstra, S., Murphy, C., Alexopoulos, G.S., 2001. Executive dysfunc- Kumar, A., Thomas, A., Lavretsky, H., Yue, K., Huda, A., Curran, J., Venkatraman, T.,
tion and disability in elderly patients with major depression. The American Estanol, L., Mintz, J., Mega, M., Toga, A., 2002. Frontal white matter biochemi-
Journal of Geriatric Psychiatry 9, 269–274. cal abnormalities in late-life major depression detected with proton magnetic
Kizilbash, A.H., Vanderploeg, R.D., Curtiss, G., 2002. The effects of depression and resonance spectroscopy. The American Journal of Psychiatry 159, 630–
anxiety on memory performance. Archives of Clinical Neuropsychology 17, 57– 636.
67. Kumral, E., Evyapan, D., Balkir, K., 1999. Acute caudate vascular lesions. Stroke 30,
Knol, M.J., Twisk, J.W.R., Beekman, A.T.F., Heine, R.J., Snoek, F.J., Pouwer, F., 2006. 100–108.
Depression as a risk factor for the onset of type 2 diabetes mellitus. A meta- Kuzma, E., Sattler, C., Toro, P., Schönknecht, P., Schröder, J., 2011. Premorbid
analysis. Diabetologia 49, 837–845. personality traits and their course in mild cognitive impairment: results from
Kohler, S., Thomas, A.J., Barnett, N.A., O’Brien, J.T., 2010a. The pattern and course of a prospective population-based study in Germany. Dementia and Geriatric
cognitive impairment in late-life depression. Psychological Medicine 40, 591– Cognitive Disorders 32, 171–177.
602. Lai, T., Payne, M.E., Byrum, C.E., Steffens, D.C., Krishnan, K.R., 2000. Reduction of
Kohler, S., Thomas, A.J., Lloyd, A., Barber, R., Almeida, O.P., O’Brien, J.T., 2010b. White orbital frontal cortex volume in geriatric depression. Biological Psychiatry 48,
matter hyperintensities, cortisol levels, brain atrophy and continuing cognitive 971–975.
deficits in late-life depression. The British Journal of Psychiatry 196, 143–149. Lange, C., Irle, E., 2004. Enlarged amygdala volume and reduced hippocampal
Kohler, S., van Boxtel, M., Jolles, J., Verhey, F., 2009. Depressive symptoms and risk volume in young women with major depression. Psychological Medicine 34,
for dementia: a 9-year follow-up of the Maastricht Aging Study. The American 1059–1064.
Journal of Geriatric Psychiatry 19, 902–905. Laudisio, A., Marzetti, E., Pagano, F., Pozzi, G., Bernabei, R., Zuccala, G., 2009.
Kondo, D.G., Speer, M.C., Krishnan, K.R., McQuoid, D.R., Slifer, S.H., Pieper, C.F., Depressive symptoms and metabolic syndrome: selective association in older
Billups, A.V., Steffens, D.C., 2007. Association of AGTR1 with 18-month treat- women. Journal of Geriatric Psychiatry and Neurology 22, 215–222.
ment outcome in late-life depression. The American Journal of Geriatric Psy- Lautenschlager, N.T., Almeida, O.P., Flicker, L., Janca, A., 2004. Can physical activity
chiatry 15, 564–572. improve the mental health of older adults? Annals of General Hospital
Kop, W.J., Gottdiener, J.S., Tangen, C.M., Fried, L.P., McBurnie, M.A., Walston, J., Psychiatry 3, 1–5.
Newman, A., Hirsch, C., Tracy, R.P., 2002. Inflammation and coagulation factors Lavretsky, H., Ballmaier, M., Pham, D., Toga, A., Kumar, A., 2007. Neuroanatomical
in persons > 65 years of age with symptoms of depression but without evidence characteristics of geriatric apathy and depression: a magnetic resonance imag-
of myocardial ischemia. The American Journal of Cardiology 89, 419–424. ing study. The American Journal of Geriatric Psychiatry 15, 386–394.
Krabbe, K.S., Pedersen, M., Bruunsgaard, H., 2004. Inflammatory mediators in the Lavretsky, H., Kurbanyan, K., Ballmaier, M., Mintz, J., Toga, A., Kumar, A., 2004. Sex
elderly. Experimental Gerontology 39, 687–699. differences in brain structure in geriatric depression. The American Journal of
Kral, V.A., Emery, O.B., 1989. Long-term follow-up of depressive pseudodementia of Geriatric Psychiatry 12, 653–657.
the aged. Canadian Journal of Psychiatry 34, 445–446. Lavretsky, H., Lesser, I.M., Wohl, M., Miller, B.L., 1998. Relationship of age, age at
Kramer-Ginsberg, E., Greenwald, B.S., Krishnan, K.R., Christiansen, B., Hu, J., Ashtari, onset, and sex to depression in older adults. The American Journal of Geriatric
M., Patel, M., Pollack, S., 1999. Neuropsychological functioning and MRI signal Psychiatry 6, 248–256.
hyperintensities in geriatric depression. The American Journal of Psychiatry Lavretsky, H., Roybal, D.J., Ballmaier, M., Toga, A.W., Kumar, A., 2005. Antidepressant
156, 438–444. exposure may protect against decrement in frontal gray matter volumes in
Kreutzberg, G.W., 1996. Microglia: a sensor for pathological events in the CNS. geriatric depression. The Journal of Clinical Psychiatry 66, 964–967.
Trends in Neurosciences 19, 312–318. Lavretsky, H., Zheng, L., Weiner, M.W., Mungas, D., Reed, B., Kramer, J.H., Jagust, W.,
Krishnan, K.R., Figiel, G.S., 1989. Neurobehavioral changes with caudate lesions. Chui, H., Mack, W.J., 2008. The MRI brain correlates of depressed mood,
Neurology 39, 1410–1411. anhedonia, apathy, and anergia in older adults with and without cognitive
Krishnan, K.R., Hays, J.C., Blazer, D.G., 1997. MRI-defined vascular depression. The impairment or dementia. International Journal of Geriatric Psychiatry 23, 1040–
American Journal of Psychiatry 154, 497–501. 1050.
138 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

Lee, J.S., Potter, G.G., Wagner, H.R., Welsh-Bohmer, K.A., Steffens, D.C., 2007. MacQueen, G.M., Campbell, S., McEwen, B.S., Macdonald, K., Amano, S., Joffe, R.T.,
Persistent mild cognitive impairment in geriatric depression. International Nahmias, C., Young, L.T., 2003. Course of illness, hippocampal function, and
Psychogeriatrics 19, 125–135. hippocampal volume in major depression. Proceedings of the National Acade-
Lee, S.H., Payne, M.E., Steffens, D.C., McQuoid, D.R., Lai, T.J., Provenzale, J.M., my of Sciences of the United States of America 100, 1387–1392.
Krishnan, K.R., 2003. Subcortical lesion severity and orbitofrontal cortex volume Maes, M., 1995. Evidence for an immune-response in major depression – a review
in geriatric depression. Biological Psychiatry 54, 529–533. and hypothesis. Progress in Neuro-psychopharmacology & Biological Psychia-
Lenze, E.J., Munin, M.C., Dew, M.A., Rogers, J.C., Seligman, K., Mulsant, B.H., Reynolds try 19, 11–38.
3rd, C.F., 2004. Adverse effects of depression and cognitive impairment on Magri, F., Cravello, L., Barili, L., Sarra, S., Cinchetti, W., Salmoiraghi, F., Micale, G.,
rehabilitation participation and recovery from hip fracture. International Jour- Ferrari, E., 2006. Stress and dementia: the role of the hypothalamic-pituitary-
nal of Geriatric Psychiatry 19, 472–478. adrenal axis. Aging Clinical and Experimental Research 18, 167–170.
Lerer, B., Gillon, D., Lichtenberg, P., Gorfine, M., Gelfin, Y., Shapira, B., 1996. Mahley, R.W., Weisgraber, K.H., Huang, Y.D., 2006. Apolipoprotein E4: a causative
Interrelationship of age, depression, and central serotonergic function: evi- factor and therapeutic target in neuropathology including Alzheimer’s disease.
dence from fenfluramine challenge studies. International Psychogeriatrics 8, Proceedings of the National Academy of Sciences of the United States of America
83–102. 103, 5644–5651.
Lesser, I.M., Boone, K.B., Mehringer, C.M., Wohl, M.A., Miller, B.L., Berman, N.G., Mahncke, H.W., Bronstone, A., Merzenich, M.M., Mahncke, H.W., Bronstone, A.,
1996. Cognition and white matter hyperintensities in older depressed patients. Merzenich, M.M., 2006. Brain plasticity and functional losses in the aged:
The American Journal of Psychiatry 153, 1280–1287. scientific bases for a novel intervention. Progress in Brain Research 157, 81–109.
Lesser, I.M., Mena, I., Boone, K.B., Miller, B.L., Mehringer, C.M., Wohl, M., 1994. Maier, S.F., Watkins, L.R., 1998. Cytokines for psychologists: implications of bidi-
Reduction of cerebral blood flow in older depressed patients. Archives of rectional immune-to-brain communication for understanding behavior, mood,
General Psychiatry 51, 677–686. and cognition. Psychological Review 105, 83–107.
Lesser, I.M., Miller, B.L., Boone, K.B., Hill-Gutierrez, E., Mehringer, C.M., Wong, K., Maisonpierre, P.C., Le Beau, M.M., Espinosa III, R., Ip, N.Y., Belluscio, L., De La Monte,
Mena, I., 1991. Brain injury and cognitive function in late-onset psychotic S.M., Squinto, S., Furth, M.E., Yancopoulos, G.D., 1991. Human and rat brain-
depression. The Journal of Neuropsychiatry and Clinical Neurosciences 3, derived neurotrophic factor and neurotrophin-3: gene structures, distributions,
33–40. and chromosomal localizations. Genomics 10, 558–568.
Liao, D., Cooper, L., Cai, J., Toole, J., Bryan, N., Burke, G., Shahar, E., Nieto, J., Mosley, T., Maj, M., Veltro, F., Pirozzi, R., Lobrace, S., Magliano, L., 1992. Pattern of recurrence of
Heiss, G., 1997. The prevalence and severity of white matter lesions, their illness after recovery from an episode of major depression: a prospective study.
relationship with age, ethnicity, gender, and cardiovascular disease risk factors: The American Journal of Psychiatry 149, 795–800.
the ARIC Study. Neuroepidemiology 16, 149–162. Malberg, J.E., Eisch, A.J., Nestler, E.J., Duman, R.S., 2000. Chronic antidepressant
Lichtman, J.H., Bigger, J.T., Blumenthal, J.A., Frasure-Smith, N., Kaufmann, P.G., treatment increases neurogenesis in adult rat hippocampus. The Journal of
Lesperance, F., Mark, D.B., Sheps, D.S., Taylor, C.B., Froelicher, E.S., 2008. De- Neuroscience 20, 9104–9110.
pression and coronary heart disease: recommendations for screening, referral, Manes, F., Jorge, R., Morcuende, M., Yamada, T., Paradiso, S., Robinson, R.G., 2001. A
and treatment: a science advisory from the American Heart Association Pre- controlled study of repetitive transcranial magnetic stimulation as a treatment
vention Committee of the Council on Cardiovascular Nursing, Council on of depression in the elderly. International Psychogeriatrics 13, 225–231.
Clinical Cardiology, Council on Epidemiology and Prevention, and Interdisci- Marin, M.-F., Lord, C., Andrews, J., Juster, R.-P., Sindi, S., Arsenault-Lapierre, G.,
plinary Council on Quality of Care and Outcomes Research Endorsed by the Fiocco, A.J., Lupien, S.J., 2011. Chronic stress, cognitive functioning and mental
American Psychiatric Association. Circulation 118, 1768–1775. health. Neurobiology of Learning and Memory 96, 583–595.
Licinio, J., Wong, M.L., 2011. Pharmacogenomics of antidepressant treatment Mast, B.T., Miles, T., Penninx, B.W., Yaffe, K., Rosano, C., Satterfield, S., Ayonayon,
effects. Dialogues in Clinical Neuroscience 13, 63–71. H.N., Harris, T., Simonsick, E.M., 2008. Vascular disease and future risk of
Lieverse, R., Van Someren, E.J.W., Nielen, M.M.A., Uitdehaag, B.M.J., Smit, J.H., depressive symptomatology in older adults: findings from the health, aging
Hoogendijk, W.J.G., 2011. Bright light treatment in elderly patients with non- and body composition study. Biological Psychiatry 64, 320–326.
seasonal Major Depressive Disorder: a randomized placebo-controlled trial. Mayberg, H.S., 1997. Limbic-cortical dysregulation: a proposed model of depres-
Archives of General Psychiatry 68, 61–70. sion. The Journal of Neuropsychiatry and Clinical Neurosciences 9, 471–481.
Lloyd, A.J., Ferrier, I.N., Barber, R., Gholkar, A., Young, A.H., O’Brien, J.T., 2004. Mayberg, H.S., 2003. Modulating dysfunctional limbic-cortical circuits in depres-
Hippocampal volume change in depression: late- and early-onset illness com- sion: towards development of brain-based algorithms for diagnosis and opti-
pared. The British Journal of Psychiatry 184, 488–495. mised treatment. British Medical Bulletin 65, 193–207.
Lobo, A., Launer, L.J., Fratiglioni, L., Andersen, K., Di Carlo, A., Breteler, M.M., 2000. Mayberg, H.S., 2007. Defining the neural circuitry of depression: toward a new
Prevalence of dementia and major subtypes in Europe: a collaborative study of nosology with therapeutic implications. Biological Psychiatry 61, 729–730.
population-based cohorts. Neurologic diseases in the Elderly Research Group. Mayberg, H.S., Brannan, S.K., Tekell, J.L., Arturo Silva, J., Mahurin, R.K., McGinnis, S.,
Neurology 54, S4–S9. Jerabek, P.A., 2000. Regional metabolic effects of fluoxetine in major depression:
Lockwood, K.A., Alexopoulos, G.S., Kakuma, T., Van Gorp, W.G., 2000. Subtypes of serial changes and relationship to clinical response. Biological Psychiatry 48,
cognitive impairment in depressed older adults. The American Journal of 830–843.
Geriatric Psychiatry 8, 201–208. Mayberg, H.S., Lozano, A.M., Voon, V., McNeely, H.E., Seminowicz, D., Hamani, C.,
Longstreth Jr., W.T., Diehr, P., Manolio, T.A., Beauchamp, N.J., Jungreis, C.A., Lefko- Schwalb, J.M., Kennedy, S.H., 2005. Deep brain stimulation for treatment-
witz, D., Cardiovascular Health Study Collaborative Research, G., 2001. Cluster resistant depression. Neuron 45, 651–660.
analysis and patterns of findings on cranial magnetic resonance imaging of the McCaffery, J.M., Frasure-Smith, N., Dube, M.P., Theroux, P., Rouleau, G.A., Duan,
elderly: the Cardiovascular Health Study. Archives of Neurology 58, 635–640. Q.L., Lesperance, F., 2006. Common genetic vulnerability to depressive symp-
Loo, C.K., Mitchell, P.B., 2005. A review of the efficacy of transcranial magnetic toms and coronary artery disease: a review and development of candidate
stimulation (TMS) treatment for depression, and current and future strategies genes related to inflammation and serotonin. Psychosomatic Medicine 68,
to optimize efficacy. Journal of Affective Disorders 88, 255–267. 187–200.
Lorenzetti, V., Allen, N.B., Fornito, A., Yucel, M., 2009. Structural brain abnormalities McCusker, J., Cole, M., Keller, E., Bellavance, F., Berard, A., 1998. Effectiveness of
in major depressive disorder: a selective review of recent MRI studies. Journal of treatments of depression in older ambulatory patients. Archives of Internal
Affective Disorders 117, 1–17. Medicine 158, 705–712.
Lotrich, F.E., 2011. Gene-environment interactions in geriatric depression. The McEwen, B.S., Olie, J.P., 2005. Neurobiology of mood, anxiety, and emotions as
Psychiatric Clinics of North America 34, 357–376. revealed by studies of a unique antidepressant: tianeptine. Molecular Psychia-
Lotrich, F.E., Loftis, J.M., Ferrell, R.E., Rabinovitz, M., Hauser, P., 2011. IL28B poly- try 10, 525–537.
morphism is associated with both side effects and clearance of hepatitis C McGorry, P.D., Hickie, I.B., Yung, A.R., Pantelis, C., Jackson, H.J., 2006. Clinical staging
during interferon-alpha therapy. Journal of Interferon & Cytokine Research 31, of psychiatric disorders: a heuristic framework for choosing earlier, safer and
331–336. more effective interventions. The Australian and New Zealand Journal of
Luber, M.P., Meyers, B.S., Williams-Russo, P.G., Hollenberg, J.P., DiDomenico, T.N., Psychiatry 40, 616–622.
Charlson, M.E., Alexopoulos, G.S., 2001. Depression and service utilization in McGorry, P.D., Tanti, C., Stokes, R., Hickie, I.B., Carnell, K., Littlefield, L.K., Moran, J.,
elderly primary care patients. The American Journal of Geriatric Psychiatry 9, 2007. Headspace: Australia’s National Youth Mental Health Foundation – where
169–176. young minds come first. The Medical Journal of Australia 187, S68–S70.
Luijendijk, H.J., van den Berg, J.F., Dekker, M.J., van Tuijl, H.R., Otte, W., Smit, F., McNally, L., Bhagwagar, Z., Hannestad, J., 2008. Inflammation, glutamate, and glia in
Hofman, A., Stricker, B.H., Tiemeier, H., 2008. Incidence and recurrence of late- depression: a literature review. CNS Spectrums 13, 501–510.
life depression. Archives of General Psychiatry 65, 1394–1401. Meltzer, C.C., Price, J.C., Mathis, C.A., Butters, M.A., Ziolko, S.K., Moses-Kolko, E.,
Lyness, J.M., King, D.A., Conwell, Y., Cox, C., Caine, E.D., 2000. Cerebrovascular risk Mazumdar, S., Mulsant, B.H., Houck, P.R., Lopresti, B.J., Weissfeld, L.A., Reynolds,
factors and 1-year depression outcome in older primary care patients. The C.F., 2004. Serotonin 1A receptor binding and treatment response in late-life
American Journal of Psychiatry 157, 1499–1501. depression. Neuropsychopharmacology 29, 2258–2265.
MacFall, J.R., Payne, M.E., Provenzale, J.E., Krishnan, K.R., 2001. Medial orbital frontal Meltzer, C.C., Price, J.C., Mathis, C.A., Greer, P.J., Cantwell, M.N., Houck, P.R., Mulsant,
lesions in late-onset depression. Biological Psychiatry 49, 803–806. B.H., Ben-Eliezer, D., Lopresti, B., DeKosky, S.T., Reynolds 3rd, C.F., 1999. PET
Mackin, R.S., Arean, P.A., 2009. Impaired financial capacity in late life depression is imaging of serotonin type 2A receptors in late-life neuropsychiatric disorders.
associated with cognitive performance on measures of executive functioning The American Journal of Psychiatry 156, 1871–1878.
and attention. Journal of International Neuropsychological Society 15, 793–798. Meltzer, C.C., Smith, G., DeKosky, S.T., Pollock, B.G., Mathis, C.A., Moore, R.Y., Kupfer,
MacQueen, G., Frodl, T., 2011. The hippocampus in major depression: evidence for D.J., Reynolds 3rd, C.F., 1998. Serotonin in aging, late-life depression, and
the convergence of the bench and bedside in psychiatric research? Molecular Alzheimer’s disease: the emerging role of functional imaging. Neuropsycho-
Psychiatry 16, 252–264. pharmacology 18, 407–430.
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 139

Mendez, M.F., Adams, N.L., Lewandowski, K.S., 1989. Neurobehavioral changes Naismith, S.L., Longley, W.A., Scott, E.M., Hickie, I.B., 2007. Disability in major
associated with caudate lesions. Neurology 39, 349–354. depression related to self-rated and objectively-measured cognitive deficits:
Milaneschi, Y., Corsi, A.M., Penninx, B.W., Bandinelli, S., Guralnik, J.M., Ferrucci, L., a preliminary study. BMC Psychiatry 7, 32.
2009. Interleukin-1 receptor antagonist and incident depressive symptoms over 6 Naismith, S.L., Norrie, L., Lewis, S.J., Rogers, N.L., Scott, E.M., Hickie, I.B., 2009b. Does
years in older persons: The InCHIANTI Study. Biological Psychiatry 65, 973–978. sleep disturbance mediate neuropsychological functioning in older people with
Milev, R., Abraham, G., Hasey, G., 2009. Repetitive transcranial magnetic stimulation depression? Journal of Affective Disorders 116, 139–143.
for treatment of medication-resistant depression in older adults: a case series. Naismith, S.L., Redoblado-Hodge, M.A., Lewis, S.J., Scott, E.M., Hickie, I.B., 2009c.
The Journal of ECT 25, 44–49. Cognitive training in affective disorders improves memory: a preliminary study
Miller, B.L., 1991. A review of chemical issues in 1H NMR spectroscopy: N-acetyl-L- using the NEAR approach. Journal of Affective Disorders 121, 258–262.
aspartate, creatine and choline. NMR in Biomedicine 4, 47–52. Naismith, S.L., Rogers, N.L., Lewis, S.J., Terpening, Z., Ip, T., Diamond, K., Norrie, L.,
Miyajima, F., Ollier, W., Mayes, A., Jackson, A., Thacker, N., Rabbitt, P., Pendleton, N., Hickie, I.B., 2011b. Sleep disturbance relates to neuropsychological functioning
Horan, M., Payton, A., 2008. Brain-derived neurotrophic factor polymorphism in late-life depression. Journal of Affective Disorders 132, 139–145.
Val66Met influences cognitive abilities in the elderly. Genes, Brain, and Behav- Nakatani, D., Sato, H., Sakata, Y., Shiotani, I., Kinjo, K., Mizuno, H., Shimizu, M., Ito, H.,
ior 7, 411–417. Koretsune, Y., Hirayama, A., Hori, M., 2005. Influence of serotonin transporter
Modrego, P.J., Ferrandez, J., 2004. Depression in patients with mild cognitive gene polymorphism on depressive symptoms and new cardiac events after
impairment increases the risk of developing dementia of Alzheimer type: a acute myocardial infarction. American Heart Journal 150, 652–658.
prospective cohort study. Archives of Neurology 61, 1290–1293. National Institute on Aging, 1999. Progress Report on Alzheimer’s Disease, 1999.
Mongeau, R., Blier, P., de Montigny, C., 1997. The serotonergic and noradrenergic Bethesda MD.
systems of the hippocampus: their interactions and the effects of antidepres- Navarro, V., Gasto, C., Lomena, F., Mateos, J.J., Marcos, T., 2001. Frontal cerebral
sant treatments. Brain Research. Brain Research Reviews 23, 145–195. perfusion dysfunction in elderly late-onset major depression assessed by
99M
Morrison, M.F., Redei, E., TenHave, T., Parmelee, P., Boyce, A.A., Sinha, P.S., Katz, I.R., TC-HMPAO SPECT. NeuroImage 14, 202–205.
2000. Dehydroepiandrosterone sulfate and psychiatric measures in a frail, Navarro, V., Gasto, C., Lomena, F., Mateos, J.J., Marcos, T., Portella, M.J., 2002.
elderly residential care population. Biological Psychiatry 47, 144–150. Normalization of frontal cerebral perfusion in remitted elderly major depres-
Mosimann, U.P., Schmitt, W., Greenberg, B.D., Kosel, M., Müri, R.M., Berkhoff, M., sion: a 12 month follow-up SPECT study. NeuroImage 16, 781–787.
Hess, C.W., Fisch, H.U., Schlaepfer, T.E., 2004. Repetitive transcranial magnetic Nebes, R.D., Pollock, B.G., Houck, P.R., Butters, M.A., Mulsant, B.H., Zmuda, M.D.,
stimulation: a putative add-on treatment for major depression in elderly Reynolds 3rd, C.F., 2003. Persistence of cognitive impairment in geriatric
patients. Psychiatry Research 126, 123–133. patients following antidepressant treatment: a randomized, double-blind clin-
Mottram, P.G., Wilson, K., Strobl, J.J., 2006. Antidepressants for depressed elderly. ical trial with nortriptyline and paroxetine. Journal of Psychiatric Research 37,
Cochrane Database of Systematic Reviews 1, CD003491. 99–108.
Mowszowski, L., Batchelor, J., Naismith, S.L., 2010. Early intervention for cognitive Nebes, R.D., Pollock, B.G., Mulsant, B.H., Kirshner, M.A., Halligan, E., Zmuda, M.,
decline. Can cognitive training be used as a selective prevention technique? Reynolds 3rd, C.F., 1997. Low-level serum anticholinergicity as a source of
International Psychogeriatrics 22, 537–548. baseline cognitive heterogeneity in geriatric depressed patients. Psychophar-
Mulsant, B.H., Ganguli, M., 1999. Epidemiology and diagnosis of depression in late macology Bulletin 33, 715–720.
life. The Journal of Clinical Psychiatry 20, 9–15. Nebes, R.D., Reynolds 3rd, C.F., Boada, F., Meltzer, C.C., Fukui, M.B., Saxton, J.,
Munafo, M.R., Brown, S.M., Hariri, A.R., 2008. Serotonin transporter (5-HTTLPR) Halligan, E.M., DeKosky, S.T., 2002. Longitudinal increase in the volume of
genotype and amygdala activation: a meta-analysis. Biological Psychiatry 63, white matter hyperintensities in late-onset depression. International Journal
852–857. of Geriatric Psychiatry 17, 526–530.
Murata, T., Kimura, H., Omori, M., Kado, H., Kosaka, H., Iidaka, T., Itoh, H., Wada, Y., Nebes, R.D., Vora, I.J., Meltzer, C.C., Fukui, M.B., Williams, R.L., Kamboh, M.I., Saxton,
2001. MRI white matter hyperintensities, (1)H-MR spectroscopy and cognitive J., Houck, P.R., DeKosky, S.T., Reynolds 3rd, C.F., 2001. Relationship of deep white
function in geriatric depression: a comparison of early- and late-onset cases. matter hyperintensities and apolipoprotein E genotype to depressive symp-
International Journal of Geriatric Psychiatry 16, 1129–1135. toms in older adults without clinical depression. The American Journal of
Murphy, C.F., Alexopoulos, G.S., 2004. Longitudinal association of initiation/persev- Psychiatry 158, 878–884.
eration and severity of geriatric depression. The American Journal of Geriatric Nelson, J.C., Delucchi, K., Schneider, L.S., 2008. Efficacy of second generation anti-
Psychiatry 12, 50–56. depressants in late-life depression: a meta-analysis of the evidence. The Amer-
Murphy, C.F., Gunning-Dixon, F.M., Hoptman, M.J., Lim, K.O., Ardekani, B., Shields, ican Journal of Geriatric Psychiatry 16, 558–567.
J.K., Hrabe, J., Kanellopoulos, D., Shanmugham, B.R., Alexopoulos, G.S., 2007. Nestler, E.J., Barrot, M., DiLeone, R.J., Eisch, A.J., Gold, S.J., Monteggia, L.M., 2002.
White-matter integrity predicts stroop performance in patients with geriatric Neurobiology of depression. Neuron 34, 13–25.
depression. Biological Psychiatry 61, 1007–1010. Nguyen, T.T., Wong, T.Y., Islam, F.M.A., Hubbard, L., Miller, J., Haroon, E., Darwin, C.,
Murphy Jr., G.M., Hollander, S.B., Rodrigues, H.E., Kremer, C., Schatzberg, A.F., 2004. Esser, B., Kumar, A., 2008. Is depression associated with microvascular disease
Effects of the serotonin transporter gene promoter polymorphism on mirtaza- in patients with type 2 diabetes? Depression and Anxiety 25, E158–E162.
pine and paroxetine efficacy and adverse events in geriatric major depression. Nicholson, A., Kuper, H., Hemingway, H., 2006. Depression as an aetiologic and
Archives of General Psychiatry 61, 1163–1169. prognostic factor in coronary heart disease: a meta-analysis of 6362 events
Murphy, G.M.J., Kremer, C., Rodrigues, H., Schatzberg, A.F., Mirtazapine versus among 146 538 participants in 54 observational studies. European Heart
Paroxetine Study Group, 2003. The Apolipoprotein E e4 allele and antidepres- Journal 27, 2763–2774.
sant efficacy in cognitively intact elderly depressed patients. Biological Psychi- NIH Consensus Development Panel on Depression in Late Life, 1992. Diagnosis and
atry 54, 665–673. treatment of depression in late life. JAMA: the Journal of the American Medical
Musselman, D.L., Evans, D.L., Nemeroff, C.B., 1998. The relationship of depression to Association 268, 1018–1024.
cardiovascular disease – epidemiology, biology, and treatment. Archives of Nikonenko, A.G., Radenovic, L., Andjus, P.R., Skibo, G.G., 2009. Structural features of
General Psychiatry 55, 580–592. ischemic damage in the hippocampus. Anatomical Record 292, 1914–1921.
Naismith, S., Hickie, I., Ward, P.B., Turner, K., Scott, E., Little, C., Mitchell, P., Wilhelm, Niti, M., Yap, K.B., Kua, E.H., Ng, T.P., 2009. APOE-epsilon 4, depressive symptoms,
K., Parker, G., 2002. Caudate nucleus volumes and genetic determinants of and cognitive decline in Chinese older adults: Singapore Longitudinal Aging
homocysteine metabolism in the prediction of psychomotor speed in older Studies. The Journals of Gerontology. Series A, Biological Sciences and Medical
persons with depression. The American Journal of Psychiatry 159, 2096–2098. Sciences 64, 306–311.
Naismith, S.L., Diamond, K., Carter, P.E., Norrie, L.M., Redoblado-Hodge, M.A., Lewis, Nobler, M.S., Mann, J.J., Sackeim, H.A., 1999a. Serotonin, cerebral blood flow, and
S.J., Hickie, I.B., 2011a. Enhancing memory in late-life depression: the effects of a cerebral metabolic rate in geriatric major depression and normal aging. Brain
combined psychoeducation and cognitive training program. The American Research Reviews 30, 250–263.
Journal of Geriatric Psychiatry 19, 240–248. Nobler, M.S., Pelton, G.H., Sackeim, H.A., 1999b. Cerebral blood flow and metabolism
Naismith, S.L., Glozier, N., Burke, D., Carter, P., Scott, E., Hickie, I., 2009a. Early in late-life depression and dementia. Journal of Geriatric Psychiatry and Neu-
intervention for cognitive decline: is there a role for multiple medical or rology 12, 118–127.
behavioural interventions? Early Intervention in Psychiatry 3, 19–27. Nobler, M.S., Roose, S.P., Prohovnik, I., Moeller, J.R., Louie, J., Van Heertum, R.L.,
Naismith, S.L., Hickie, I.B., Lewis, S.J., 2010a. The role of mild depression in sleep Sackeim, H.A., 2000. Regional cerebral blood flow in mood disorders, V. Effects
disturbance and quality of life in Parkinson’s disease. The Journal of Neuropsy- of antidepressant medication in late-life depression. The American Journal of
chiatry and Clinical Neurosciences 22, 384–389. Geriatric Psychiatry 8, 289–296.
Naismith, S.L., Hickie, I.B., Turner, K., Little, C.L., Winter, V., Ward, P.B., Wilhelm, K., Nobuhara, K., Okugawa, G., Sugimoto, T., Minami, T., Tamagaki, C., Takase, K., Saito, Y.,
Mitchell, P., Parker, G., 2003. Neuropsychological performance in patients with Sawada, S., Kinoshita, T., 2006. Frontal white matter anisotropy and symptom
depression is associated with clinical, etiological and genetic risk factors. severity of late-life depression: a magnetic resonance diffusion tensor imaging
Journal of Clinical and Experimental Neuropsychology 25, 866–877. study. Journal of Neurology, Neurosurgery, and Psychiatry 77, 120–122.
Naismith, S.L., Hickie, I.B., Ward, P.B., Scott, E., Little, C., 2006. Impaired implicit Norrie, L.M., Diamond, K., Hickie, I.B., Rogers, N.L., Fearns, S., Naismith, S.L., 2011.
sequence learning in depression: a probe for frontostriatal dysfunction? Psy- Can older ‘‘at risk’’ adults benefit from psychoeducation targeting healthy brain
chological Medicine 36, 313–323. aging? International Psychogeriatrics 23, 413–424.
Naismith, S.L., Lagopoulos, J., Ward, P.B., Davey, C.G., Little, C., Hickie, I.B., 2010b. O’Brien, J., Ames, D., Chiu, E., Schweitzer, I., Desmond, P., Tress, B., 1998. Severe deep
Fronto-striatal correlates of impaired implicit sequence learning in major white matter lesions and outcome in elderly patients with major depressive
depression: an fMRI study. Journal of Affective Disorders 125, 256–261. disorder: follow up study. British Medical Journal 317, 982–984.
Naismith, S.L., Lewis, S.J., 2011. DASH’’ symptoms in patients with Parkinson’s O’Brien, J., Desmond, P., Ames, D., Schweitzer, I., Harrigan, S., Tress, B., 1996. A
disease: red flags for early cognitive decline. Journal of Clinical Neuroscience 18, magnetic resonance imaging study of white matter lesions in depression and
352–355. Alzheimer’s disease. The British Journal of Psychiatry 168, 477–485.
140 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

O’Brien, J.T., Lloyd, A., McKeith, I., Gholkar, A., Ferrier, N., 2004. A longitudinal study Portella, M.J., Marcos, T., Rami, L., Navarro, V., Gasto, C., Salamero, M., 2003. Residual
of hippocampal volume, cortisol levels, and cognition in older depressed cognitive impairment in late-life depression after a 12-month period follow-up.
subjects. The American Journal of Psychiatry 161, 2081–2090. International Journal of Geriatric Psychiatry 18, 571–576.
O’Doherty, J., Dayan, P., Schultz, J., Deichmann, R., Friston, K., Dolan, R.J., 2004. Potter, G.G., Blackwell, A.D., McQuoid, D.R., Payne, M.E., Steffens, D.C., Sahakian, B.J.,
Dissociable roles of ventral and dorsal striatum in instrumental conditioning. Welsh-Bohmer, K.A., Krishnan, K.R., 2007. Prefrontal white matter lesions and
Science 304, 452–454. prefrontal task impersistence in depressed and nondepressed elders. Neurop-
O’Hara, R., Hallmayer, J.F., 2007. Serotonin transporter polymorphism and stress: a sychopharmacology 32, 2135–2142.
view across the lifespan. Current Psychiatry Reports 9, 173–175. Potter, G.G., Steffens, D.C., 2007. Contribution of depression to cognitive im-
Okello, A., Edison, P., Archer, H.A., Turkheimer, F.E., Kennedy, J., Bullock, R., Walker, pairment and dementia in older adults. Neurologist 13, 105–117.
Z., Kennedy, A., Fox, N., Rossor, M., Brooks, D.J., 2009. Microglial activation and Potter, G.G., Wagner, H.R., Burke, J.R., Plassman, B.L., Welsh-Bohmer, K.A., Steffens,
amyloid deposition in mild cognitive impairment A PET study. Neurology 72, D.C., 2012. Neuropsychological predictors of dementia in late-life Major De-
56–62. pressive Disorder. The American Journal of Geriatric Psychiatry epub ahead of
Oldehinkel, A.J., van den Berg, M.D., Flentge, F., Bouhuys, A.L., ter Horst, G.J., Ormel, print.
J., 2001. Urinary free cortisol excretion in elderly persons with minor and major Qiu, A., Taylor, W.D., Zhao, Z., MacFall, J.R., Miller, M.I., Key, C.R., Payne, M.E.,
depression. Psychiatry Research 104, 39–47. Steffens, D.C., Krishnan, K.R., 2009. APOE related hippocampal shape alteration
Owen, A.M., 2000. The role of the lateral frontal cortex in mnemonic processing: the in geriatric depression. NeuroImage 44, 620–626.
contribution of functional neuroimaging. Experimental Brain Research 133, 33– Rajagopalan, S., Brook, R., Rubenfire, M., Pitt, E., Young, E., Pitt, B., 2001. Abnormal
43. bronchial artery flow-mediated vasodilation in young adults with major de-
Ownby, R.L., Crocco, E., Acevedo, A., John, V., Loewenstein, D., 2006. Depression and pression. The American Journal of Cardiology 88, 196–198.
risk for Alzheimer disease: systematic review, meta-analysis, and metaregres- Rajkowska, G., 2000. Postmortem studies in mood disorders indicate altered
sion analysis. Archives of General Psychiatry 63, 530–538. numbers of neurons and glial cells. Biological Psychiatry 48, 766–777.
Pagani, M., Salmaso, D., Nardo, D., Jonsson, C., Jacobsson, H., Larsson, S.A., Gardner, Rajkowska, G., Miguel-Hidalgo, J.J., 2007. Gliogenesis and glial pathology in depres-
A., 2007. Imaging the neurobiological substrate of atypical depression by SPECT. sion. CNS & Neurological Disorders-Drug Targets 6, 219–233.
European Journal of Nuclear Medicine and Molecular Imaging 34, 110–120. Rajkowska, G., Miguel-Hidalgo, J.J., Dubey, P., Stockmeier, C.A., Krishnan, K.R.,
Pan, A., Lucas, M., Sun, Q., van Dam, R.M., Franco, O.H., Willett, W.C., Manson, J.E., 2005a. Prominent reduction in pyramidal neurons density in the orbitofrontal
Rexrode, K.M., Ascherio, A., Hu, F.B., 2011. Increased mortality risk in women cortex of elderly depressed patients. Biological Psychiatry 58, 297–306.
with depression and diabetes mellitus. Archives of General Psychiatry 68, 42– Rajkowska, G., Miguel-Hidalgo, J.J., Dubey, P., Stockmeier, C.A., Krishnan, K.R.R.,
50. 2005b. Prominent reduction in pyramidal neurons density in the orbitofrontal
Pan, A., Ye, X.W., Franco, O.H., Li, H.X., Yu, Z.J., Wang, J., Qi, Q.B., Gu, W.J., Pang, X.H., cortex of elderly depressed patients. Biological Psychiatry 58, 297–306.
Liu, H., Lin, X., 2008. The association of depressive symptoms with inflamma- Rajkowska, G., Miguel-Hidalgo, J.J., Wei, J., Dilley, G., Pittman, S.D., Meltzer, H.Y.,
tory factors and adipokines in middle-aged and older Chinese. PLoS One 3, 1–6. Overholser, J.C., Roth, B.L., Stockmeier, C.A., 1999. Morphometric evidence for
Pantel, J., Schroder, J., Essig, M., Popp, D., Dech, H., Knopp, M.V., Schad, L.R., neuronal and glial prefrontal cell pathology in major depression. Biological
Eysenbach, K., Backenstrass, M., Friedlinger, M., 1997. Quantitative magnetic Psychiatry 45, 1085–1098.
resonance imaging in geriatric depression and primary degenerative dementia. Ramasubbu, R., 2011. Therapy for prevention of post-stroke depression. Expert
Journal of Affective Disorders 42, 69–83. Opinion on Pharmacotherapy 12, 2177–2187.
Panza, F., Frisardi, V., Capurso, C., D’Introno, A., Colacicco, A.M., Imbimbo, B.P., Ramasubbu, R., Patten, S.B., 2003. Effect of depression on stroke morbidity and
Santamato, A., Vendemiale, G., Seripa, D., Pilotto, A., Capurso, A., Solfrizzi, V., mortality. Canadian Journal of Psychiatry 48, 250–257.
2010. Late-life depression, mild cognitive impairment, and dementia: possible Rapp, M.A., Dahlman, K., Sano, M., Grossman, H.T., Haroutunian, V., Gorman, J.M.,
continuum? The American Journal of Geriatric Psychiatry 18, 98–116. 2005. Neuropsychological differences between late-onset and recurrent geri-
Paradise, M., Naismith, S.L., Norrie, L.M., Graeber, M.B., Hickie, I.B., in press. The role atric major depression. The American Journal of Psychiatry 162, 691–698.
of glia in late-life depression. International Psychogeriatrics. Raskin, J., Wiltse, C.G., Siegal, A., Sheikh, J., Xu, J., Dinkel, J.J., Rotz, B.T., Mohs, R.C.,
Patankar, T.F., Baldwin, R., Mitra, D., Jeffries, S., Sutcliffe, C., Bums, A., Jackson, A., 2007. Efficacy of duloxetine on cognition, depression, and pain in elderly
2007. Virchow-Robin space dilatation may predict resistance to antidepressant patients with major depressive disorder: an 8-week, double-blind, placebo-
monotherapy in elderly patients with depression. Journal of Affective Disorders controlled trial. The American Journal of Psychiatry 164, 900–909.
97, 265–270. Rasmuson, S., Andrew, R., Nasman, B., Seckl, J.R., Walker, B.R., Olsson, T., 2001.
Penninx, B.W., Beekman, A.T., Bandinelli, S., Corsi, A.M., Bremmer, M., Hoogendijk, Increased glucocorticoid production and altered cortisol metabolism in women
W.J., Guralnik, J.M., Ferrucci, L., 2007. Late-life depressive symptoms are asso- with mild to moderate Alzheimer’s disease. Biological Psychiatry 49, 547–552.
ciated with both hyperactivity and hypoactivity of the hypothalamo-pituitary- Reynolds 3rd, C.F., Frank, E., Perel, J.M., Imber, S.D., Cornes, C., Miller, M.D., Mazumdar,
adrenal axis. The American Journal of Geriatric Psychiatry 15, 522–529. S., Houck, P.R., Dew, M.A., Stack, J.A., Pollock, B.G., Kupfer, D.J., 1999. Nortriptyline
Penninx, B.W., Geerlings, S.W., Deeg, D.J., van Eijk, J.T., van Tilburg, W., Beekman, and interpersonal psychotherapy as maintenance therapies for recurrent major
A.T., 1999. Minor and major depression and the risk of death in older persons. depression: a randomized controlled trial in patients older than 59 years. JAMA:
Archives of General Psychiatry 56, 889–895. the Journal of the American Medical Association 281, 39–45.
Penninx, B.W., Kritchevsky, S.B., Yaffe, K., Newman, A.B., Simonsick, E.M., Rubin, S., Reynolds 3rd, C.F., Dew, M.A., Frank, E., Begley, A.E., Miller, M.D., Cornes, C.,
Ferrucci, L., Harris, T., Pahor, M., 2003. Inflammatory markers and depressed Mazumdar, S., Perel, J.M., Kupfer, D.J., 1998. Effects of age at onset of first
mood in older persons: results from the Health, Aging and Body Composition lifetime episode of recurrent major depression on treatment response and
study. Biological Psychiatry 54, 566–572. illness course in elderly patients. The American Journal of Psychiatry 155,
Perera, T.D., Coplan, J.D., Lisanby, S.H., Lipira, C.M., Arif, M., Carpio, C., Spitzer, G., 795–799.
Santarelli, L., Scharf, B., Hen, R., Rosoklija, G., Sackeim, H.A., Dwork, A.J., 2007. Reynolds, C.F.r., Smith, G.S., Dew, M.A., Mulsant, B.H., Miller, M.D., Schlernitzauer,
Antidepressant-induced neurogenesis in the hippocampus of adult nonhuman M., Stack, J.A., Houck, P.R., Pollock, B.G., 2005. Accelerating symptom-reduction
primates. The Journal of Neuroscience 27, 4894–4901. in late-life depression: a double-blind, randomized, placebo-controlled trial of
Pezawas, L., Meyer-Lindenberg, A., Drabant, E.M., Verchinski, B.A., Munoz, K.E., sleep deprivation. The American Journal of Geriatric Psychiatry 13, 353–358.
Kolachana, B.S., Egan, M.F., Mattay, V.S., Hariri, A.R., Weinberger, D.R., 2005. 5- Reynolds, S.L., Haley, W.E., Kozlenko, N., 2008. The impact of depressive symptoms
HTTLPR polymorphism impacts human cingulate-amygdala interactions: a and chronic diseases on active life expectancy in older Americans. The American
genetic susceptibility mechanism for depression. Nature Neuroscience 8, Journal of Geriatric Psychiatry 16, 425–432.
828–834. Risch, N., Herrell, R., Lehner, T., Liang, K.Y., Eaves, L., Hoh, J., Griem, A., Kovacs, M.,
Pezawas, L., Verchinski, B.A., Mattay, V.S., Callicott, J.H., Kolachana, B.S., Straub, R.E., Ott, J., Merikangas, K.R., 2009. Interaction between the Serotonin Transporter
Egan, M.F., Meyer-Lindenberg, A., Weinberger, D.R., 2004. The brain-derived Gene (5-HTTLPR), stressful life events, and risk of depression: a meta-analysis.
neurotrophic factor val66met polymorphism and variation in human cortical JAMA: the Journal of the American Medical Association 301, 2462–2471.
morphology. The Journal of Neuroscience 24, 10099–10102. Ritchie, K., Carriere, I., Ritchie, C.W., Berr, C., Artero, S., Ancelin, M.L., 2010. Designing
Phillips-Bute, B., Mathew, J.P., Blumenthal, J.A., Morris, R.W., Podgoreanu, M.V., prevention programmes to reduce incidence of dementia: prospective cohort
Smith, M., Stafford-Smith, M., Grocott, H.P., Schwinn, D.A., Newman, M.F., Team, study of modifiable risk factors. British Medical Journal 341, c3885.
P.I., 2008. Relationship of genetic variability and depressive symptoms to Ritchie, K., Gilham, C., Ledesert, B., Touchon, J., Kotzki, P.O., 1999. Depressive illness,
adverse events after coronary artery bypass graft surgery. Psychosomatic depressive symptomatology and regional cerebral blood flow in elderly people
Medicine 70, 953–959. with sub-clinical cognitive impairment. Age and Ageing 28, 385–391.
Phillips, M.L., Drevets, W.C., Rauch, S.L., Lane, R., 2003. Neurobiology of emotion Robinson, R.G., Spalleta, G., 2010. Poststroke depression: a review. Canadian Journal
perception I: the neural basis of normal emotion perception. Biological Psychi- of Psychiatry 55, 341–349.
atry 54, 504–514. Roose, S.P., Schatzberg, A.F., 2005. The efficacy of antidepressants in the treatment
Pinquart, M., Duberstein, P.R., Lyness, J.M., 2006. Treatments for later-life depres- of late-life depression. Journal of Clinical Psychopharmacology 25, S1–S7.
sive conditions: a meta-analytic comparison of pharmacotherapy and psycho- Ross, B.D., 1991. Biochemical considerations in 1H spectroscopy. Glutamate and
therapy. The American Journal of Psychiatry 163, 1493–1501. glutamine; myo-inositol and related metabolites. NMR in Biomedicine 4, 59–
Pisljar, M., Pirtosek, Z., Repovs, G., Grgic, M., 2008. Executive dysfunction in late- 63.
onset depression. Psychiatria Danubina 20, 231–235. Rostami, M., Hosseini, S.M., Takahashi, M., Sugiura, M., Kawashima, R., 2009. Neural
Pittenger, C., Duman, R.S., 2008. Stress, depression, and neuroplasticity: a conver- bases of goal-directed implicit learning. NeuroImage 48, 303–310.
gence of mechanisms. Neuropsychopharmacology Reviews 33, 88–109. Royall, D.R., Lauterbach, E.C., Cummings, J.L., Reeve, A., Rummans, T.A., Kaufer, D.I.,
Popeo, D., Kellner, C.H., 2009. ECT for Parkinson’s disease. Medical Hypotheses 73, LaFrance Jr., W.C., Coffey, C.E., 2002. Executive control function: a review of its
468–469. promise and challenges for clinical research. A report from the Committee on
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 141

Research of the American Neuropsychiatric Association. The Journal of Neuro- Sheline, Y.I., Barch, D.M., Garcia, K., Gersing, K., Pieper, C., Welsh-Bohmer, K.,
psychiatry and Clinical Neurosciences 14, 377–405. Steffens, D.C., Doraiswamy, P.M., Sheline, Y.I., Barch, D.M., Garcia, K., Gersing,
Rubanyi, G.M., 1993. The role of endothelium in cardiovascular homeostasis and K., Pieper, C., Welsh-Bohmer, K., Steffens, D.C., Doraiswamy, P.M., 2006. Cogni-
diseases. Journal of Cardiovascular Pharmacology 22, S1–S14. tive function in late life depression: relationships to depression severity,
Rugulies, R., 2002. Depression as a predictor for coronary heart disease – a review cerebrovascular risk factors and processing speed. Biological Psychiatry 60,
and meta-analysis. American Journal of Preventive Medicine 23, 51–61. 58–65.
Rutherford, B., Sneed, J., Miyazaki, M., Eisenstadt, R., Devanand, D., Sackeim, H., Sheline, Y.I., Gado, M.H., Kraemer, H.C., 2003. Untreated depression and hippocam-
Roose, S., 2007. An open trial of aripiprazole augmentation for SSRI non- pal volume loss. The American Journal of Psychiatry 160, 1516–1518.
remitters with late-life depression. International Journal of Geriatric Psychiatry Sheline, Y.I., Gado, M.H., Price, J.L., 1998. Amygdala core nuclei volumes are
22, 986–991. decreased in recurrent major depression. Neuroreport 9, 2023–2028.
Sachdev, P., Parslow, R., Salonikas, C., Lux, O., Wen, W., Kumar, R., Naidoo, D., Sheline, Y.I., Mintun, M.A., Barch, D.M., Wilkins, C., Snyder, A.Z., Moerlein, S.M.,
Christensen, H., Jorm, A., 2004. Homocysteine and the brain in midadult life: 2004. Decreased hippocampal 5-HT2A receptor binding in older depressed
evidence for an increased risk of leukoaraiosis in men. Archives of Neurology 61, patients using [18F]altanserin positron emission tomography. Neuropsycho-
1369–1376. pharmacology 29, 2235–2241.
Sachdev, P.S., Brodaty, H., Valenzuela, M.J., Lorentz, L., Looi, J.C.L., Berman, K., Ross, Sheline, Y.I., Pieper, C.F., Barch, D.M., Welsh-Boehmer, K., McKinstry, R.C., MacFall,
A., Wen, W., Zagami, A.S., 2006. Clinical determinants of dementia and mild J.R., D’Angelo, G., Garcia, K.S., Gersing, K., Wilkins, C., Taylor, W., Steffens, D.C.,
cognitive impairment following ischaemic stroke: The Sydney Stroke Study. Krishnan, R.R., Doraiswamy, P.M., 2010. Support for the vascular depression
Dementia and Geriatric Cognitive Disorders 21, 275–283. hypothesis in late-life depression: results of a 2-site, prospective, antidepres-
Sachs-Ericsson, N., Sawyer, K., Corsentino, E., Collins, N., Steffens, D.C., 2011. The sant treatment trial. Archives of General Psychiatry 67, 277–285.
moderating effect of the APOE epsilon 4 allele on the relationship between Sheline, Y.I., Price, J.L., Vaishnavi, S.N., Mintun, M.A., Barch, D.M., Epstein, A.A.,
hippocampal volume and cognitive decline in older depressed patients. The Wilkins, C.H., Snyder, A.Z., Couture, L., Schechtman, K., McKinstry, R.C., 2008.
American Journal of Geriatric Psychiatry 19, 23–32. Regional white matter hyperintensity burden in automated segmentation
Saczynski, J.S., Beiser, A., Seshadri, S., Auerbach, S., Wolf, P.A., Au, R., 2010. Depres- distinguishes late-life depressed subjects from comparison subjects matched
sive symptoms and risk of dementia: the Framingham Heart Study. Neurology for vascular risk factors. The American Journal of Psychiatry 165, 524–532.
75, 35–41. Sheline, Y.I., Sanghavi, M., Mintun, M.A., Gado, M.H., 1999. Depression duration but
Saint-Cyr, J.A., 2003. Frontal-striatal circuit functions: context, sequence, and not age predicts hippocampal volume loss in medically healthy women with
consequence. Journal of International Neuropsychological Society 9, 103–127. recurrent major depression. The Journal of Neuroscience 19, 5034–5043.
Salloway, S., Correia, S., Boyle, P., Malloy, P., Schneider, L., Lavretsky, H., Sackheim, Sheline, Y.I., Wang, P.W., Gado, M.H., Csernansky, J.G., Vannier, M.W., 1996. Hippo-
H., Roose, S., Krishnan, K.R., 2002. MRI subcortical hyperintensities in old and campal atrophy in recurrent major depression. Proceedings of the National
very old depressed outpatients: the important role of age in late-life depression. Academy of Sciences of the United States of America 93, 3908–3913.
Journal of the Neurological Sciences 204, 227–233. Shelton, R.C., Claiborne, J., Sidoryk-Wegrzynowicz, M., Reddy, R., Aschner, M., Lewis,
Salloway, S., Malloy, P., Kohn, R., Gillard, E., Duffy, J., Rogg, J., Tung, G., Richardson, E., D.A., Mirnics, K., 2011. Altered expression of genes involved in inflammation
Thomas, C., Westlake, R., 1996. MRI and neuropsychological differences in and apoptosis in frontal cortex in major depression. Molecular Psychiatry 16,
early- and late-life-onset geriatric depression. Neurology 46, 1567–1574. 751–762.
Santos, M., Kovari, E., Hof, P.R., Gold, G., Bouras, C., Giannakopoulos, P., 2009. The Shi, Y.Y., You, J.Y., Yuan, Y.G., Zhang, X.R., Li, H.L., Hou, G., 2010. Plasma BDNF and tPA
impact of vascular burden on late-life depression. Brain Research Reviews 62, are associated with late-onset geriatric depression. Psychiatry and Clinical
19–32. Neurosciences 64, 249–254.
Sapolsky, R.M., Krey, L.C., McEwen, B.S., 1986. The neuroendocrinology of stress and Shima, S., Shikano, T., Kitamura, T., Masuda, Y., Tsukumo, T., Kanba, S., Asai, M., 1984.
aging – the glucocorticoid cascade hypothesis. Endocrine Reviews 7, 284–301. Depression and ventricular enlargement. Acta Psychiatric Scandinavica 70,
Sapolsky, R.M., Romero, L.M., Munck, A.U., 2000. How do glucocorticoids influence 275–277.
stress responses? Integrating permissive, suppressive, stimulatory, and prepar- Shimony, J.S., Sheline, Y.I., D’Angelo, G., Epstein, A.A., Benzinger, T.L., Mintun, M.A.,
ative actions. Endocrine Reviews 21, 55–89. McKinstry, R.C., Snyder, A.Z., 2009. Diffuse microstructural abnormalities of
Sarginson, J.E., Lazzeroni, L.C., Ryan, H.S., Ershoff, B.D., Schatzberg, A.F., Murphy, normal-appearing white matter in late life depression: a diffusion tensor
G.M., 2010. ABCB1 (MDR1) polymorphisms and antidepressant response in imaging study. Biological Psychiatry 66, 245–252.
geriatric depression. Pharmacogenetics and Genomics 20, 467–475. Si, X., Miguel-Hidalgo, J.J., O’Dwyer, G., Stockmeier, C.A., Rajkowska, G., 2004. Age-
Sarginson, J.E., Lazzeroni, L.C., Ryan, H.S., Schatzberg, A.F., Murphy, G.M.J., 2009. dependent reductions in the level of glial fibrillary acidic protein in the
FKBP5 polymorphisms and antidepressant response in geriatric depression. prefrontal cortex in major depression. Neuropsychopharmacology 29, 2088–
American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics 153B, 2096.
554–560. Simpson, J.E., Ince, P.G., Higham, C.E., Gelsthorpe, C.H., Fernando, M.S., Matthews, F.,
Savitz, J., Drevets, W.C., 2009. Bipolar and major depressive disorder: neuroimaging Forster, G., O’Brien, J.T., Barber, R., Kalaria, R.N., Brayne, C., Shaw, P.J., Stoeber, K.,
the developmental-degenerative divide. Neuroscience and Biobehavioral Williams, G.H., Lewis, C.E., Wharton, S.B., 2007. Microglial activation in white
Reviews 33, 699–771. matter lesions and nonlesional white matter of ageing brains. Neuropathology
Schatzberg, A.F., Kremer, C., Rodrigues, H.E., Murphy, G.M., 2002. Double-blind, and Applied Neurobiology 33, 670–683.
randomized comparison of mirtazapine and paroxetine in elderly depressed Simpson, S., Baldwin, R.C., Jackson, A., Burns, A., 1999. The differentiation of DSM-
patients. The American Journal of Geriatric Psychiatry 10, 541–550. III-R psychotic depression in later life from nonpsychotic depression: compar-
Scheltens, P., Barkhof, F., Leys, D., Pruvo, J.P., Nauta, J.J.P., Vermersch, P., Steinling, M., isons of brain changes measured by multispectral analysis of magnetic reso-
Valk, J., 1993. A semiquantitative rating-scale for the assessment of signal nance brain images, neuropsychological findings, and clinical features.
hyperintensities on magnetic-resonance-imaging. Journal of the Neurological Biological Psychiatry 45, 193–204.
Sciences 114, 7–12. Simpson, S.W., Jackson, A., Baldwin, R.C., Burns, A., 1997. Subcortical hyperinten-
Schoevers, R.A., Geerlings, M.I., Deeg, D.J.H., Holwerda, T.J., Jonker, C., Beekman, sities in late-life depression: acute response to treatment and neuropsycholog-
A.T.F., 2009. Depression and excess mortality: evidence for a dose response ical impairment. International Psychogeriatrics 9, 257–275.
relation in community living elderly. International Journal of Geriatric Psychi- Singh, N.A., Clements, K.M., Fiatarone Singh, M.A., 2001. The efficacy of exercise as a
atry 24, 169–176. long-term antidepressant in elderly subjects: a randomized, controlled trial.
Schrader, G., Cheok, F., Hordacre, A.L., Marker, J., Wade, V., 2005. Effect of psychiatry The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences
liaison with general practitioners on depression severity in recently hospita- 56A, M497–M504.
lised cardiac patients: a randomised controlled trial. The Medical Journal of Slaughter, J.R., Slaughter, K.A., Nichols, D., Holmes, S.E., Martens, M.P., 2001.
Australia 182, 272–276. Prevalence, clinical manifestations, etiology, and treatment of depression in
Schroeter, M.L., Steiner, J., Mueller, K., 2011. Glial pathology is modified by age in Parkinson’s disease. The Journal of Neuropsychiatry and Clinical Neurosciences
mood disorders – a systematic meta-analysis of serum S100B in vivo studies. 13, 187–196.
Journal of Affective Disorders 134, 32–38. Smith, G.S., Kramer, E., Ma, Y., Kingsley, P., Dhawan, V., Chaly, T., Eidelberg, D.,
Schweitzer, I., Tuckwell, V., Ames, D., O’Brien, J., 2001. Structural neuroimaging 2009a. The functional neuroanatomy of geriatric depression. International
studies in late-life depression: a review. World Journal of Biological Psychiatry Journal of Geriatric Psychiatry 24, 798–808.
2, 83–88. Smith, G.S., Kramer, E., Reynolds, C.F., Hermann, C.R., Ma, Y., Greenwald, B., Pollock,
Schweitzer, I., Tuckwell, V., O’Brien, J., Ames, D., 2002. Is late onset depression a B.G., Eidelberg, D., 2004. Positron emission tomography studies of the func-
prodrome to dementia? International Journal of Geriatric Psychiatry 17, 997– tional neuroanatomy of geriatric depression. Biological Psychiatry 55, 181S.
1005. Smith, G.S., Reynolds, C.F.r., Houck, P.R., Dew, M.A., Ginsberg, J., Ma, Y., Mulsant, B.H.,
Serfaty, M.A., Haworth, D., Blanchard, M., Buszewicz, M., Murad, S., King, M., 2009. Pollock, B.G., 2009b. The cerebral glucose metabolic response to combined total
Clinical effectiveness of individual cognitive behavioral therapy for depressed sleep deprivation and antidepressant treatment in geriatric depression: a
older people in primary care: a randomized controlled trial. Archives of General randomized, placebo controlled study. Psychiatry Research 171, 1–9.
Psychiatry 66, 1332–1340. Smith, G.S., Reynolds, C.F.r., Houck, P.R., Dew, M.A., Ma, Y., Mulsant, B.H., Pollock,
Shah, A.J., Veledar, E., Hong, Y., Bremner, J.D., Vaccarino, V., 2011. Depression and B.G., 2002. Glucose metabolic response to total sleep deprivation, recovery
history of attempted suicide as risk factors for heart disease mortality in young sleep, and acute anti-depressant treatment as functional neuroanatomic cor-
individuals. Archives of General Psychiatry 68, 1135–1142. relates of treatment outcome in geriatric depression. The American Journal of
Shallice, T., Fletcher, P., Frith, C.D., Grasby, P., Frackowiak, R.S.J., Dolan, R.J., 1994. Geriatric Psychiatry 10, 561–567.
Brain regions associated with acquisition and retrieval of verbal episodic Smith, G.S., Reynolds, C.F.r., Pollock, B., Derbyshire, S., Nozfinger, E., Dew, M.A.,
memory. Nature 368, 633–663. Houck, P.R., Milko, D., Cidis Meltzer, C., Kupfer, D.J., 1999. Cerebral glucose
142 S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143

metabolic response to combined total sleep deprivation and antidepressant postmortem hippocampus in major depression. Biological Psychiatry 56, 640–
treatment in geriatric depression. The American Journal of Psychiatry 156, 683– 650.
689. Story, T.J., Potter, G.G., Attix, D.K., Welsh-Bohmer, K.A., Steffens, D.C., 2008. Neu-
Smith, K., 2011. Trillion-dollar brain drain. Nature 478, 15. rocognitive correlates of response to treatment in late-life depression. The
Smith, P.J., Blumenthal, J.A., Babyak, M.A., Doraiswamy, P.M., Hinderliter, A., Hoff- American Journal of Geriatric Psychiatry 16, 752–759.
man, B.M., Waugh, R., Sherwood, A., 2009c. Intima-media thickness and age of Subramaniam, H., Mitchell, A.J., 2005. The prognosis of depression in late life versus
first depressive episode. Biological Psychology 80, 361–364. mid-life: implications for the treatment of older adults. International Psycho-
Sneed, J.R., Rindskopf, D., Steffens, D.C., Krishnan, K.R.R., Roose, S.P., 2008. The geriatrics 17, 533–537.
vascular depression subtype: evidence of internal validity. Biological Psychiatry Sun, C., Tikellis, G., Klein, R., Steffens, D.C., Marino Larsen, E.K., Siscovick, D.S., Klein,
64, 491–497. B.E.K., Wong, T.Y., 2007. Are microvascular abnormalities in the retina associ-
Sneed, J.R., Roose, S.P., Keilp, J.G., Krishnan, K.R., Alexopoulos, G.S., Sackeim, H.A., ated with depression symptoms? The Cardiovascular Health study. The Amer-
2007. Response inhibition predicts poor antidepressant treatment response in ican Journal of Geriatric Psychiatry 15, 335–343.
very old depressed patients. The American Journal of Geriatric Psychiatry 15, Surtees, P.G., Wainwright, N.W.J., Willis-Owen, S.A.G., Luben, R., Day, N.E., Flint, J.,
553–563. 2006. Social adversity, the serotonin transporter (5-HTTLPR) polymorphism
Soares, J.C., Mann, J.J., 1997. The functional neuroanatomy of mood disorders. and major depressive disorder. Biological Psychiatry 59, 224–229.
Journal of Psychiatric Research 31, 393–432. Sweet, R.A., Hamilton, R.L., Butters, M.A., Mulsant, B.H., Pollock, B.G., Lewis, D.A.,
Spalletta, G., Bossu, P., Ciaramella, A., Bria, P., Caltagirone, C., Robinson, R.G., 2006. Lopez, O.L., DeKosky, S.T., Reynolds 3rd, C.F., 2004. Neuropathologic correlates
The etiology of poststroke depression: a review of the literature and a new of late-onset major depression. Neuropsychopharmacology 29, 2242–2250.
hypothesis involving inflammatory cytokines. Molecular Psychiatry 11, 984– Taki, Y., Kinomura, S., Awata, S., Inoue, K., Sato, K., Ito, H., Goto, R., Uchida, S., Tsuji, I.,
991. Arai, H., Kawashima, R., Fukuda, H., 2005. Male elderly subthreshold depression
Sparkman, N.L., Johnson, R.W., 2008. Neuroinflammation associated with aging patients have smaller volume of medial part of prefrontal cortex and precentral
sensitizes the brain to the effects of infection or stress. Neuroimmunomodula- gyrus compared with age-matched normal subjects: a voxel-based morphom-
tion 15, 323–330. etry. Journal of Affective Disorders 88, 313–320.
Squire, L.R., 1992. Memory and the hippocampus: a synthesis from findings with Tamburo, R.J., Siegle, G.J., Stetten, G.D., Cois, C.A., Butters, M.A., Reynolds 3rd, C.F.,
rats, monkeys, and humans. Psychological Review 99, 195–231. Aizenstein, H.J., 2009. Amygdalae morphometry in late-life depression. Inter-
Starkstein, S.E., Robinson, R.G., Price, T.R., 1987. Comparison of cortical and sub- national Journal of Geriatric Psychiatry 24, 837–846.
cortical lesions in the production of poststroke mood disorders. Brain 110 (Pt 4), Taragano, F.E., Bagnatti, P., Allegri, R.F., 2005. A double-blind, randomized clinical
1045–1059. trial to assess the augmentation with nimodipine of antidepressant therapy in
Steffens, D.C., 2008. Separating mood disturbance from mild cognitive impairment the treatment of ‘‘vascular depression’’. International Psychogeriatrics 17, 487–
in geriatric depression. International Review of Psychiatry 20, 374–381. 498.
Steffens, D.C., Bosworth, H.B., Provenzale, J.M., MacFall, J.R., 2002a. Subcortical Taylor, W.D., Doraiswamy, P.M., 2004. A systematic review of antidepressant
white matter lesions and functional impairment in geriatric depression. De- placebo-controlled trials for geriatric depression: limitations of current data
pression and Anxiety 15, 23–28. and directions for the future. Neuropsychopharmacology 29, 2285–2299.
Steffens, D.C., Byrum, C.E., McQuoid, D.R., Greenberg, D.L., Payne, M.E., Blitchington, Taylor, W.D., Kuchibhatla, M., Payne, M.E., Macfall, J.R., Sheline, Y.I., Krishnan, K.R.,
T.F., MacFall, J.R., Krishnan, K.R., 2000. Hippocampal volume in geriatric de- Doraiswamy, P.M., 2008a. Frontal white matter anisotropy and antidepressant
pression. Biological Psychiatry 48, 301–309. remission in late-life depression. PLoS One 3, e3267.
Steffens, D.C., Conway, C.R., Dombeck, C.B., Wagner, H.R., Tupler, L.A., Weiner, R.D., Taylor, W.D., MacFall, J.R., Payne, M.E., McQuoid, D.R., Provenzale, J.M., Steffens, D.C.,
2001. Severity of subcortical gray matter hyperintensity predicts ECT response Krishnan, K.R., 2004. Late-life depression and microstructural abnormalities in
in geriatric depression. The Journal of ECT 17, 45–49. dorsolateral prefrontal cortex white matter. The American Journal of Psychiatry
Steffens, D.C., Hays, J.C., Krishnan, K.R., 1999a. Disability in geriatric depression. The 161, 1293–1296.
American Journal of Geriatric Psychiatry 7, 34–40. Taylor, W.D., MacFall, J.R., Payne, M.E., McQuoid, D.R., Steffens, D.C., Provenzale, J.M.,
Steffens, D.C., Helms, M.J., Krishnan, K.R., Burke, G.L., 1999b. Cerebrovascular Krishnan, R.R., 2005a. Greater MRI lesion volumes in elderly depressed subjects
disease and depression symptoms in the cardiovascular health study. Stroke than in control subjects. Psychiatry Research Neuroimaging 139, 1–7.
30, 2159–2166. Taylor, W.D., MacFall, J.R., Steffens, D.C., Payne, M.E., Provenzale, J.M., Krishnan, K.R.,
Steffens, D.C., Krishnan, K.R., 1998. Structural neuroimaging and mood disorders: 2003a. Localization of age-associated white matter hyperintensities in late-life
recent findings, implications for classification, and future directions. Biological depression. Progress in Neuro-psychopharmacology & Biological Psychiatry 27,
Psychiatry 43, 705–712. 539–544.
Steffens, D.C., McQuoid, D.R., 2005. Impact of symptoms of generalized anxiety Taylor, W.D., McQuoid, D.R., Ashley-Koch, A., MacFall, J.R., Bridgers, J., Krishnan, R.R.,
disorder on the course of late-life depression. The American Journal of Geriatric Steffens, D.C., 2010a. BDNF Val66Met genotype and 6-month remission rates in
Psychiatry 13, 40–47. late-life depression. The Pharmacogenomics Journal 11, 146–154.
Steffens, D.C., McQuoid, D.R., Payne, M.E., Potter, G.G., 2011. Change in hippocampal Taylor, W.D., Steffens, D.C., Ashley-Koch, A., Payne, M.E., MacFall, J.R., Potocky, C.F.,
volume on magnetic resonance imaging and cognitive decline among older Krishnan, R.R., 2010b. Angiotensin receptor gene polymorphisms and 2-year
depressed and nondepressed subjects in the neurocognitive outcomes of de- change in hyperintense lesion volume in men. Molecular Psychiatry 15, 816–
pression in the elderly study. The American Journal of Geriatric Psychiatry 19, 822.
4–12. Taylor, W.D., Steffens, D.C., McQuoid, D.R., Payne, M.E., Lee, S.H., Lai, T.J., Krishnan,
Steffens, D.C., McQuoid, D.R., Potter, G.G., 2009. Outcomes of older cognitively K.R., 2003b. Smaller orbital frontal cortex volumes associated with functional
impaired individuals with current and past depression in the NCODE study. disability in depressed elders. Biological Psychiatry 53, 144–149.
Journal of Geriatric Psychiatry and Neurology 22, 52–61. Taylor, W.D., Steffens, D.C., Payne, M.E., MacFall, J.R., Marchuk, D.A., Svenson, I.K.,
Steffens, D.C., Norton, M.C., Hart, A.D., Skoog, I., Corcoran, C., Breitner, J.C., Cache Krishnan, K.R., 2005b. Influence of serotonin transporter promoter region
County Study, G., 2003. Apolipoprotein E genotype and major depression in a polymorphisms on hippocampal volumes in late-life depression. Archives of
community of older adults. The Cache County Study. Psychological Medicine 33, General Psychiatry 62, 537–544.
541–547. Taylor, W.D., Zuchner, S., McQuoid, D.R., Payne, M.E., MacFall, J.R., Steffens, D.C.,
Steffens, D.C., Payne, M.E., Greenberg, D.L., Byrum, C.E., Welsh-Bohmer, K.A., Speer, M.C., Krishnan, K.R., 2008b. The brain-derived neurotrophic factor
Wagner, H.R., MacFall, J.R., 2002b. Hippocampal volume and incident dementia VAL66MET polymorphism and cerebral white matter hyperintensities in
in geriatric depression. The American Journal of Geriatric Psychiatry 10, 62–71. late-life depression. The American Journal of Geriatric Psychiatry 16, 263–
Steffens, D.C., Potter, G.G., McQuoid, D.R., MacFall, J.R., Payne, M.E., Burke, J.R., 271.
Plassman, B.L., Welsh-Bohmer, K.A., 2007. Longitudinal magnetic resonance Taylor, W.D., Zuchner, S., McQuoid, D.R., Steffens, D.C., Speer, M.C., Krishnan, K.R.,
imaging vascular changes, apolipoprotein E genotype, and development of 2007. Allelic differences in the brain-derived neurotrophic factor Val66Met
dementia in the neurocognitive outcomes of depression in the elderly study. polymorphism in late-life depression. The American Journal of Geriatric Psy-
The American Journal of Geriatric Psychiatry 15, 839–849. chiatry 15, 850–857.
Steffens, D.C., Svenson, I., Marchuk, D.A., Levy, R.M., Hays, J.C., Flint, E.P., Krishnan, Tekin, S., Cummings, J.L., 2002. Frontal-subcortical neuronal circuits and clinical
K.R., Siegler, I.C., 2002c. Allelic differences in the serotonin transporter-linked neuropsychiatry – an update. Journal of Psychosomatic Research 53, 647–654.
polymorphic region in geriatric depression. The American Journal of Geriatric Teodorczuk, A., Firbank, M.J., Pantoni, L., Poggesi, A., Erkinjuntti, T., Wallin, A.,
Psychiatry 10, 185–191. Wahlund, L.O., Scheltens, P., Waldemar, G., Schrotter, G., Ferro, J.M., Chabriat, H.,
Steffens, D.C., Taylor, W.D., McQuoid, D.R., Krishnan, K.R., 2008. Short/long hetero- Bazner, H., Visser, M., Inzitari, D., O’Brien, J.T., Grp, L., 2010. Relationship
zygotes at 5HTTLPR and white matter lesions in geriatric depression. Interna- between baseline white-matter changes and development of late-life depres-
tional Journal of Geriatric Psychiatry 23, 244–248. sive symptoms: 3-year results from the LADIS study. Psychological Medicine 40,
Steptoe, A., Kunz-Ebrecht, S.R., Owen, N., 2003. Lack of association between de- 603–610.
pressive symptoms and markers of immune and vascular inflammation in Teodorczuk, A., O’Brien, J.T., Firbank, M.J., Pantoni, L., Poggesi, A., Erkinjuntti, T.,
middle-aged men and women. Psychological Medicine 33, 667–674. Wallin, A., Wahlund, L.O., Gouw, A., Waldemar, G., Schmidt, R., Ferro, J.M.,
Stewart, R., Prince, M., Mann, A., Richards, M., Brayne, C., 2001. Stroke, vascular risk Chabriat, H., Bazner, H., Inzitari, D., 2007. White matter changes and late-life
factors and depression: cross-sectional study in a UK Caribbean-born popula- depressive symptoms: longitudinal study. The British Journal of Psychiatry 191,
tion. The British Journal of Psychiatry 178, 23–28. 212–217.
Stockmeier, C.A., Mahajan, G.J., Konick, L.C., Overholser, J.C., Jurjus, G.J., Meltzer, Teper, E., O’Brien, J.T., 2008. Vascular factors and depression. International Journal of
H.Y., Uylings, H.B., Friedman, L., Rajkowska, G., 2004. Cellular changes in the Geriatric Psychiatry 23, 993–1000.
S.L. Naismith et al. / Progress in Neurobiology 98 (2012) 99–143 143

Thomas, A.J., Davis, S., Morris, C., Jackson, E., Harrison, R., O’Brien, J.T., 2005. Increase von Gunten, A., Fox, N.C., Cipolotti, L., Ron, M.A., 2000. A volumetric study of
in interleukin-1beta in late-life depression. The American Journal of Psychiatry hippocampus and amygdala in depressed patients with subjective memory
162, 175–177. problems. The Journal of Neuropsychiatry and Clinical Neurosciences 12, 493–
Thomas, A.J., Ferrier, I.N., Kalaria, R.N., Woodward, S.A., Ballard, C., Oakley, A., Perry, 498.
R.H., O’Brien, J.T., 2000. Elevation in late-life depression of intercellular adhe- Wahlund, L.O., Barkhof, F., Fazekas, F., Bronge, L., Augustin, M., Sjogren, M., Wallin,
sion molecule-1 expression in the dorsolateral prefrontal cortex. The American A., Ader, H., Leys, D., Pantoni, L., Pasquier, F., Erkinjuntti, T., Scheltens, P.,
Journal of Psychiatry 157, 1682–1684. European Task Force Age-Related, W., 2001. A new rating scale for age-related
Thomas, A.J., Gallagher, P., Robinson, L.J., Porter, R.J., Young, A.H., Ferrier, I.N., white matter changes applicable to MRI and CT. Stroke 32, 1318–1322.
O’Brien, J.T., 2009. A comparison of neurocognitive impairment in younger Walker, J.G., Mackinnon, A.J., Batterham, P., Jorm, A.F., Hickie, I., McCarthy, A.,
and older adults with major depression. Psychological Medicine 39, 725– Fenech, M., Christensen, H., 2010. Mental health literacy, folic acid and vitamin
733. B12, and physical activity for the prevention of depression in older adults:
Thomas, A.J., Kalaria, R.N., O’Brien, J.T., 2004. Depression and vascular disease: what randomised controlled trial. The British Journal of Psychiatry 197, 45–54.
is the relationship? Journal of Affective Disorders 79, 81–95. Wang, J.W., David, D.J., Monckton, J.E., Battaglia, F., Hen, R., 2008a. Chronic fluoxe-
Thomas, A.J., O’Brien, J.T., Davis, S., Ballard, C., Barber, R., Kalaria, R.N., Perry, R.H., tine stimulates maturation and synaptic plasticity of adult-born hippocampal
2002. Ischemic basis for deep white matter hyperintensities in major depres- granule cells. The Journal of Neuroscience 28, 1374–1384.
sion: a neuropathological study. Archives of General Psychiatry 59, 785–792. Wang, L., Krishnan, K.R., Steffens, D.C., Potter, G.G., Dolcos, F., McCarthy, G., 2008b.
Thomas, A.J., Perry, R., Kalaria, R.N., Oakley, A., McMeekin, W., O’Brien, J.T., 2003. Depressive state- and disease-related alterations in neural responses to affec-
Neuropathological evidence for ischemia in the white matter of the dorsolateral tive and executive challenges in geriatric depression. The American Journal of
prefrontal cortex in late-life depression. International Journal of Geriatric Psychiatry 165, 863–871.
Psychiatry 18, 7–13. Wang, L., Swank, J.S., Glick, I.E., Gado, M.H., Miller, M.I., Morris, J.C., Csernansky, J.G.,
Thompson, L.W., Coon, D.W., Gallagher-Thompson, D., Sommer, B.R., Koin, D., 2001. 2003. Changes in hippocampal volume and shape across time distinguish
Comparison of desipramine and cognitive/behavioral therapy in the treatment dementia of the Alzheimer type from healthy aging. NeuroImage 20, 667–682.
of elderly outpatients with mild-to-moderate depression. The American Journal Welch, G.N., Loscalzo, J., 1998. Homocysteine and atherothrombosis. The New
of Geriatric Psychiatry 9, 225–240. England Journal of Medicine 338, 1042–1050.
Tiemeier, H., Hofman, A., van Tuijl, H.R., Kiliaan, A.J., Meijer, J., Breteler, M.M., 2003. Wennstrom, M., Hellsten, J., Tingstrom, A., 2004. Electroconvulsive seizures induce
Inflammatory proteins and depression in the elderly. Epidemiology 14, 103– proliferation of NG2-expressing glial cells in adult rat amygdala. Biological
107. Psychiatry 55, 464–471.
Traykov, L., Bayle, A.C., Latour, F., Lenoir, H., Seux, M.L., Hanon, O., Pequignot, R., Bert, Whyte, E.M., Dew, M.A., Gildengers, A., Lenze, E.J., Bharucha, A., Mulsant, B.H.,
P., Moulin, F., Cantegreil, I., Wenisch, E., Batouche, F., Mehrabian, S., de Rotrou, J., Reynolds, C.F., 2004. Time course of response to antidepressants in late-life
Rigaud, A.S., 2007. Apolipoprotein E epsilon 4 allele frequency in elderly major depression: therapeutic implications. Drugs & Aging 21, 531–554.
depressed patients with and without cerebrovascular disease. Journal of the Whyte, E.M., Pollock, B.G., Wagner, W.R., Mulsant, B.H., Ferrell, R.E., Mazumdar, S.,
Neurological Sciences 257, 280–283. Reynolds 3rd, C.F., 2001. Influence of serotonin-transporter-linked promoter
Trichard, C., Martinot, J.L., Alagille, M., Masure, M.C., Hardy, P., Ginestet, D., Feline, A., region polymorphism on platelet activation in geriatric depression. The Ameri-
1995. Time-course of prefrontal lobe dysfunction in severely depressed inpa- can Journal of Psychiatry 158, 2074–2076.
tients – a longitudinal neuropsychological study. Psychological Medicine 25, Wilhelm, K., Mitchell, P., Sengoz, A., Hickie, I., Brodaty, H., Boyce, B., 1994. Treatment
79–85. resistant depression in an Australian context II: outcome of a series of patients.
Tsopelas, C., Stewart, R., Savva, G.M., Brayne, C., Ince, P., Thomas, A., Matthews, F.E., The Australian and New Zealand Journal of Psychiatry 28, 23–33.
2011. Neuropathological correlates of late-life depression in older people. The Wilson, K., Mottram, P.G., Sivananthan, A., Nightingale, A., 2001. Antidepressants
British Journal of Psychiatry 198, 109–114. versus placebo for the depressed elderly. Cochrane Database of Systematic
Tulving, E., Markowitsch, H.J., 1997. Memory beyond the hippocampus. Current Reviews 2, CD000561.
Opinion in Neurobiology 7, 209–216. Wolkowitz, O.M., Reus, V.I., Mellon, S.H., 2011. Of sound mind and body: depression,
Tupler, L.A., Krishnan, K.R., McDonald, W.M., Dombeck, C.B., D’Souza, S., Steffens, disease, and accelerated aging. Dialogues in Clinical Neuroscience 13, 25–39.
D.C., 2002. Anatomic location and laterality of MRI signal hyperintensities in Wong, A., Mok, V., Fan, Y.H., Lam, W.W.M., Liang, K.S., Wong, K.S., 2006. Hyperho-
late-life depression. Journal of Psychosomatic Research 53, 665–676. mocysteinemia is associated with volumetric white matter change in patients
Tyrka, A.R., Mello, A.F., Mello, M.F., Gagne, G.G., Grover, K.E., Anderson, G.M., Price, with small vessel disease. Journal of Neurology 253, 441–447.
L.H., Carpenter, L.L., 2006. Temperament and hypothalamic–pituitary–adrenal Woo, S.L., Prince, S.E., Petrella, J.R., Hellegers, C., Doraiswamy, P.M., 2009. Modula-
axis function in healthy adults. Psychoneuroendocrinology 31, 1036–1045. tion of a human memory circuit by subsyndromal depression in late life: a
Upadhyaya, A.K., Abou-Saleh, M.T., Wilson, K., Grime, S.J., Critchley, M., 1990. A functional magnetic resonance imaging study. The American Journal of Geriat-
study of depression in old age using single-photon emission computerised ric Psychiatry 17, 24–29.
tomography. The British Journal of Psychiatry 9, 76–81. Wu, J.C., Gillin, J.C., Buchsbaum, M.S., Schachat, C., Darnall, L.A., Keator, D.B., Fallon,
Ustun, T.B., Ayuso-Mateos, J.L., Chatterji, S., Mathers, C., Murray, C.J., 2004. Global J.H., Bunney, W.E., 2008. Sleep deprivation PET correlations of Hamilton symp-
burden of depressive disorders in the year 2000.[see comment] The British tom improvement ratings with changes in relative glucose metabolism in
Journal of Psychiatry 184, 386–392. patients with depression. Journal of Affective Disorders 107, 181–186.
Vaishnavi, S., Taylor, W.D., 2006. Neuroimaging in late-life depression. International Wu, Q., Magnus, J., Liu, J., Bencaz, A., Hentz, J., 2009. Depression and low bone
Review of Psychiatry 18, 443–451. mineral density: a meta-analysis of epidemiologic studies. Osteoporosis Inter-
van den Biggelaar, A.H., Gussekloo, J., de Craen, A.J., Frolich, M., Stek, M.L., van der national 20, 1309–1320.
Mast, R.C., Westendorp, R.G., 2007. Inflammation and interleukin-1 signaling Wyckoff, N., Kumar, A., Gupta, R.C., Alger, J., Hwang, S., Thomas, M.A., 2003.
network contribute to depressive symptoms but not cognitive decline in old Magnetization transfer imaging and magnetic resonance spectroscopy of nor-
age. Experimental Gerontology 42, 693–701. mal-appearing white matter in late-life major depression. Journal of Magnetic
van den Heuval, M.P., Hulshoff Pol, H.E., 2010. Exploring the brain network: a review Resonance Imaging 18, 537–543.
on resting-state fMRI functional connectivity. European Neuropsychopharma- Yang, Q., Huang, X., Hong, N., Yu, X., 2007. White matter microstructural abnor-
cology 20, 519–534. malities in late-life depression. International Psychogeriatrics 19, 757–766.
Van Otterloo, E., O’Dwyer, G., Stockmeier, C.A., Steffens, D.C., Krishnan, R.R., Raj- Yuan, Y., Zhang, Z., Bai, F., Yu, H., Shi, Y., Qian, Y., Liu, W., You, J., Zhang, X., Liu, Z.,
kowska, G., 2009. Reductions in neuronal density in elderly depressed are 2008. Abnormal neural activity in the patients with remitted geriatric depres-
region specific. International Journal of Geriatric Psychiatry 24, 856–864. sion: a resting-state functional magnetic resonance imaging study. Journal of
van Straaten, E.C.W., Fazekas, F., Rostrup, E., Scheltens, P., Schmidt, R., Pantoni, L., Affective Disorders 111, 145–152.
Inzitari, D., Waldemar, G., Erkinjuntti, T., Mantyla, R., Wahlund, L.O., Barkhof, F., Yuan, Y., Zhang, Z., Bai, F., Yu, H., Shi, Y., Qian, Y., Zang, Y., Zhu, C., Liu, W., You, J.,
Grp, L., 2006. Impact of white matter hyperintensities scoring method on 2007. White matter integrity of the whole brain is disrupted in first-episode
correlations with clinical data – The LADIS study. Stroke 37, 836–840. remitted geriatric depression. Neuroreport 18, 1845–1849.
Venkatraman, T.N., Krishnan, R.R., Steffens, D.C., Song, A.W., Taylor, W.D., 2009. Yuan, Y.G., Zhang, Z.J., Bai, F., You, J.Y., Yui, H., Shi, Y.M., Liu, W., 2010. Genetic
Biochemical abnormalities of the medial temporal lobe and medial prefrontal variation in apolipoprotein E alters regional gray matter volumes in remitted
cortex in late-life depression. Psychiatry Research 172, 49–54. late-onset depression. Journal of Affective Disorders 121, 273–277.
Versluis, C.E., van der Mast, R.C., van Buchem, M.A., Bollen, E.L., Blauw, G.J., Eekhof, Zarate, C.A., Payne, J.L., Quiroz, J., Sporn, J., Denicoff, K.K., Luckenbaugh, D., Charney,
J.A., van der Wee, N.J., de Craen, A.J., 2006. Progression of cerebral white matter D.S., Manji, H.K., 2004. An open-label trial of riluzole in patients with treatment-
lesions is not associated with development of depressive symptoms in elderly resistant major depression. The American Journal of Psychiatry 161, 171–174.
subjects at risk of cardiovascular disease: The PROSPER Study. International Zhao, Z., Taylor, W.D., Styner, M., Steffens, D.C., Krishnan, K.R., MacFall, J.R., 2008.
Journal of Geriatric Psychiatry 21, 375–381. Hippocampus shape analysis and late-life depression. PLoS One 3, e1837.

You might also like