Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Mar 1.
Published in final edited form as: Mol Psychiatry. 2013 Feb 26;18(9):963–974. doi: 10.1038/mp.2013.20

The Vascular Depression Hypothesis: Mechanisms Linking Vascular Disease with Depression

Warren D Taylor 1, Howard J Aizenstein 2, George S Alexopoulos 3
PMCID: PMC3674224  NIHMSID: NIHMS437868  PMID: 23439482

Abstract

The ‘Vascular Depression’ hypothesis posits that cerebrovascular disease may predispose, precipitate, or perpetuate some geriatric depressive syndromes. This hypothesis stimulated much research that has improved our understanding of the complex relationships between late-life depression (LLD), vascular risk factors, and cognition. Succinctly, there are well-established relationships between late-life depression, vascular risk factors, and cerebral hyperintensities, the radiological hallmark of vascular depression. Cognitive dysfunction is common in late-life depression, particularly executive dysfunction, a finding predictive of poor antidepressant response. Over time, progression of hyperintensities and cognitive deficits predicts a poor course of depression and may reflect underlying worsening of vascular disease. This work laid the foundation for examining the mechanisms by which vascular disease influences brain circuits and influences the development and course of depression. We review data testing the vascular depression hypothesis with a focus on identifying potential underlying vascular mechanisms. We propose a disconnection hypothesis, wherein focal vascular damage and white matter lesion location is a crucial factor influencing neural connectivity that contributes to clinical symptomatology. We also propose inflammatory and hypoperfusion hypotheses, concepts that link underlying vascular processes with adverse effects on brain function that influence the development of depression. Testing such hypotheses will not only inform the relationship between vascular disease and depression but also provide guidance on the potential repurposing of pharmacological agents that may improve late-life depression outcomes.

Keywords: Depression, geriatrics, cerebrovascular, review, neuroimaging, cognition


There is substantial heterogeneity in the biological factors that influence the risk of developing depression across the lifespan. Genetic and epigenetic factors influence gene expression or protein function across a variety of molecular systems, resulting in a vulnerability to developing depression in context of psychosocial adversity.1 This heterogeneity is even more apparent in depression occurring in older adults, or late-life depression (LLD), as aging-related changes across multiple organ systems also contribute to depression. Aging-related changes in the neurologic, immune, and endocrine systems have all been implicated in depression, but one of the best studied in LLD is the role of vascular disease.

The ‘Vascular Depression’ Hypothesis proposed that “cerebrovascular disease may predispose, precipitate, or perpetuate some geriatric depressive syndromes”.2 The hypothesis was accompanied by two proposals on the core characteristics of and how to define a vascular depression subtype. First, Alexopoulos and colleagues proposed a working definition based on the presence of vascular risk factors. The clinical presentation of “vascular depression” was characterized by cognitive deficits, psychomotor retardation, lack of insight, and disability disproportional to the depression severity.2, 3 Investigators subsequently focused on the cognitive dysfunction occurring in LLD and its relationship to response to antidepressants.4 Second, Krishnan and colleagues proposed a MRI-based definition 5 requiring evidence of vascular changes on neuroimaging, referred to as MRI hyperintensities. Both definitions are important as subsequent work demonstrated the internal validity of a vascular depression diagnostic subtype characterized by MRI findings and executive dysfunction.6

White Matter Lesions, White Matter Hyperintensities, and MRI-Defined Vascular Depression

The hallmark of MRI-defined vascular depression is the presence of white matter lesions (WMLs) identified as white matter hyperintensities (WMH) on T2-weighted or fluid attenuated inversion recovery (FLAIR) MRI. Throughout this review we refer to the radiological findings as WMHs, while reserving the term WML when discussing the underlying changes in the cerebral white matter.

WMHs are associated with advanced age 7 and cerebrovascular risk factors including diabetes, cardiac disease, and hypertension.8-12 Vascular dysregulation contributes to WMH development as white matter is sensitive to transient ischemia 13 and many larger WMHs are ischemic in origin.14, 15 Hypertension and blood pressure variability are associated with LLD 16-18 and also contribute to WMH development,19, 20 particularly when accompanied by impaired cerebral vasomotor reactivity and altered autoregulatory processes.21, 22 Such deficits reduce cerebral blood flow (CBF) and may lead to WMHs.23, 24

WMHs in Late-Life Depression

LLD is consistently associated with greater WMH severity 25-28 and greater measured WMH volumes.10, 29-31 These observations guided proposals for MRI-defined vascular depression diagnostic criteria that include a lesion severity criterion.32, 33 In such conceptualizations, age at the initial depressive episode is an important consideration. Compared with early-onset depression, individuals with a later onset (e.g., after 50 years) exhibit greater WMH severity 5, 18, 31, 34-38 and cognitive impairment.39, 40 However, “vascular depression” as a potential diagnostic entity may not be limited to late-onset patients. Individuals with an earlier onset are at increased vascular risk as precedent depression is associated with increased risk of vascular disease and stroke.41-45 Depression in early and mid-life may promote inflammation 46-48 or epigenetic modifications of genes related to vascular homeostasis.49 Thus some individuals with early onset depression may be prone to “vascular depression” later in life.

Localization of WMHs

Although many studies do not specify lesion location, distinctions in WMH location are important in understanding their role in LLD. In nondepressed samples, periventricular WMHs are more closely associated with cognitive impairment than are deep WMHs.50-52 It is possible this is due to anatomical differences, as the periventricular region has a high density of long associating fibers with cortical-subcortical connections, while subcortical deep white matter has a high density of shorter U-fibers connecting adjacent cortical regions.52, 53 However, as WMH severity across regions is highly correlated, distinctions between periventricular and deep WMH may be arbitrary and reflect total WMH disease.54 Nonetheless, others have localized WMHs associated with depression to the frontal 55-58 and temporal lobe.59 More recently several groups reported that LLD is associated with greater WMH severity in specific white matter fiber tracts including the cingulum bundle, uncinate fasciculus, and superior longitudinal fasciculus.60-62

WMHs and Cerebral Function

A small number of studies suggest that greater WMH burden is associated with disrupted function in motor, cognitive, and affective circuits. During a simple motor task, high WMH burden is associated with low BOLD functional signal,63 suggesting that WMH disease interferes with the neural circuit mediating the task. Other studies 64, 65 associated low BOLD activity with poorer performance on executive tasks, supporting the model of altered white matter integrity leading to cognitive network dysfunction. Greater WMH volume is further associated with altered default mode network connectivity at rest between the posterior cingulate cortex and medial prefrontal cortex.66 During affective tasks, in LLD WMHs are associated with altered functional activity with an exaggeration of the functional ‘depression’ fMRI patterns: individuals with high WMH burden showed hyperactivation in limbic regions when presented with fearful faces.67

Neuropathological Studies

Studies utilizing both neuroimaging and neuropathological techniques demonstrate that WMHs represent a wide range of pathological processes, including perivascular demyelination, arteriosclerosis, ischemia, gliosis, or partial loss of myelin and axons.15, 68, 69 Generally, confluent deep WMH but not periventricular WMH appear to be related to ischemic processes.70

There are also regional differences in WMH etiology. Seminal work by Thomas and colleagues showed that deep WMH and punctate lesions of depressed older adults are most likely to have ischemic origins. In LLD ischemic lesions are also more likely to occur in the DLPFC, instead of the ACC or occipital lobe.14 Similarly, depressed elders exhibit increased expression of cellular adhesion molecules (CAMs) in the DLPFC, but not the ACC or occipital lobe.71-73 CAMs are inflammatory markers whose expression is increased by ischemia, supporting a role for ischemia in LLD and highlighting the relationship between vascular and inflammatory processes. Although ischemic pathology is thus consistently localized to the DLPFC, other frontal regions may also be involved. Depressed elders exhibit decreased density of pyramidal neurons in the orbitofrontal cortex, which may be the result of vascular processes 74 in arterioles and medium arteries 75 or alterations in astrocyte-associated immune function.76

Neuropathological studies also demonstrate that LLD can develop in the absence of significant vascular abnormalities. In a study of late-onset depressed elders, depression was not related to either lacunes or microvascular lesions.77 Similarly, in a population-based study where depression was ascertained in a pre-mortem diagnostic interview, depression was associated neither with cerebrovascular nor Alzheimer pathology.78 However, in contrast to prior studies,79 depression was associated with the presence of Lewy bodies.78 Such findings highlight the heterogeneity of pathologies that present as depression.

The Depression-Executive Dysfunction (DED) Syndrome and Cognitive Deficits in LLD

A “depression-executive dysfunction syndrome” 4 was conceptualized as the clinical expression of frontal network impairment caused by vascular and other aging related factors. Accordingly, this syndrome describes depressed patients with vascular disease and evidence of impairment in networks related to mood and executive function.80

Executive function is a frontally mediated domain 81-83 that encompasses cognitive processes including selective attention, response inhibition, and performance monitoring. It is clinically expressed as difficulty with planning, sequencing, organizing, and abstracting. These deficits are common in LLD,84-87 particularly in late-onset depression.40, 86, 88 Efforts to characterize the presentation of the DED Syndrome have dichotomized samples by their performance on executive function tasks. When compared with depressed elders without executive dysfunction, patients with DED exhibit reduced fluency, impaired visual naming, suspiciousness, anhedonia, psychomotor retardation, and significant disability.89-91 As discussed later, studies across eight different samples identified executive dysfunction as a predictor of poor antidepressant response.92-99

Broader Cognitive Deficits in LLD: Relationship with Vascular Risk Factors

Individuals with LLD exhibit executive dysfunction and deficits across other cognitive domains including episodic memory, working memory, visuospatial ability, and processing speed.84, 87, 100-105 Processing speed deficits may influence other cognitive deficits 103-105 and mediate in part executive task performance.106 Although cognitive performance improves with successful antidepressant treatment, such deficits can persist and performance may not be restored to a normal level.100, 107-109

Cognitive deficits are associated with vascular dysfunction. In nondepressed cohorts, there are associations between episodic memory, psychomotor speed, and executive function, and measures of vascular dysfunction, such as hypertension, heart rate variability, and orthostasis.110-112 In LLD, Framingham Vascular Risk scores 113 are associated with deficits in processing speed, executive function, and episodic memory.98, 104 Greater WMH burden is associated with executive dysfunction, perseveration, and slowed processing speed 12, 61, 114-117 but also episodic and visuospatial memory deficits.102, 110, 118, 119 These cross-sectional observations are supported by longitudinal studies demonstrating that progression of WMH severity parallels cognitive decline.120-122

WMHs localized to specific fiber tracts are associated with specific cognitive deficits. Executive function deficits are associated with WMHs in the uncinate fasciculus, superior longitudinal fasciculus, and the cingulum bundle.123 Similarly, episodic memory deficits are associated with WMHs in the cingulum bundle and inferior longitudinal fasciculus.123, 124

Vascular Depression and Course of Depression

Cognitive Dysfunction and Antidepressant Outcomes

In LLD, the relationship between treatment outcome and executive dysfunction is well established. Multiple studies demonstrate that executive dysfunction predicts poor acute response of LLD to antidepressants.92, 94, 96, 97, 99, 125 Executive dysfunction is also associated with high relapse and recurrence rates,126 although not all studies found this association.127 Much of this work used the Mattis Dementia Rating Scale Initiation-Perseveration (I/P) subtest, which yields a composite score of several executive skills.128 Notably, a recent meta-analysis examined the relationship between antidepressant response and performance across multiple executive tests, including the I/P subtest, the Wisconsin Card Sort Test, the Stroop Color-Word test, and others. The authors concluded that the I/P subtest was the only test providing reliable discrimination between antidepressant responders and nonresponders.129 Further analysis of I/P subtest components showed that semantic strategy (semantic clustering) while performing the I/P verbal fluency task explained most of the variance in predicting remission.130 Effective semantic strategy appears to be associated with treatment response regardless of the probing task; effective semantic strategy during verbal learning task was also associated with high LLD remission rate.131 These observations suggest that preserved semantic organization, rather than fluency or verbal learning, is critical for remission during antidepressant treatment. However, impaired response inhibition is another aspect of executive dysfunction that influences antidepressant response.92, 95, 125, 132

Corroborating the role of the cognitive control network in LLD are reports examining resting state functional connectivity (FC). Low resting FC in the cognitive control network, but not in the default network, predict poor acute antidepressant remission rates along with persistence of depression, apathy and dysexecutive behavior.133

Other cognitive deficits can influence outcomes. Poorer performance on tests of episodic memory, language processing, and processing speed are associated with poorer acute antidepressant response 98 and may predict poorer long-term course of depression.134 Further analysis is needed to identify a superordinate cognitive dysfunction that accounts for the relationship of these tests to antidepressant response.

WMHs as a Predictor of Antidepressant Outcomes

Although greater WMH severity is often associated with poorer acute antidepressant response,36, 98, 99, 135-138 results are inconsistent.97, 139-142 Importantly, these studies assess total cerebral WMH severity, while the effect of WMLs on the antidepressant response may depend on lesion location.138 Unfortunately, methodological differences in assessing WMH severity make it difficult to reconcile discrepant findings across studies.143, 144

Most studies used cross-sectional WMH measures to predict outcome. However, WMHs are a progressive rather than a static process 11, 145 and rate of change may be a more important predictor. The few studies in LLD cohorts examining this question found that greater increases in WMH volume over two- and four-year intervals are associated with nonremission or relapse.146, 147

Some have examined both WMH severity and cognitive dysfunction as predictors of outcome. In these studies cognitive measures are generally a better predictor of antidepressant response than are WMH measures. The largest published study examining a 12-week response to sertraline found that poorer response was independently associated with greater WMH severity and poorer performance in cognitive domains of episodic memory, language processing, processing speed and executive function. However, in models controlling for baseline depression severity, WMH severity was no longer associated with response.98 Smaller studies have found similar patterns, wherein initial associations between WMH severity and response were not statistically significant in models incorporating cognitive measures.97, 99

Other measures of white matter integrity are associated with antidepressant response. In voxel-wide analysis, microstructural white matter abnormalities (low fractional anisotropy, FA) in multiple frontolimbic brain areas, including the rostral and dorsal anterior cingulate, dorsolateral prefrontal cortex, genu of the corpus callosum, hippocampus, and posterior cingulate, were associated with non-remission.148 In region of interest analyses, low FA in the corpus callosum, left superior corona radiata, and right inferior longitudinal fasciculum was associated with lower remission rates.149 In contrast, others found that higher prefrontal white matter FA was associated with a failure to remit to antidepressants,150 a finding that parallels reports in depression of increased functional connectivity to a dorsal nexus.151 Notably, depressed elderly 5-HTTLPR polymorphism short allele carriers had lower FA than long allele homozygotes in frontolimbic areas, including the anterior cingulate, posterior cingulate, dorsolateral and medial prefrontal regions.152 Such differences in FA can be caused by several pathologies, not all of which are vascular in nature.

Vascular Risk Genes in Depression

There is a wide literature examining genetic influences in LLD, including reports examining polymorphisms in BDNF, APOE, and serotonin transporter (5-HTTLPR) genes, amongst others (see recent reviews by Lotrich 153 and Benjamin and Taylor 154). Within this field, there are an increasing number of reports associating LLD with genetic polymorphisms increasing vascular risk.

One example of this literature includes studies examining genetic variation of the renin-angiotensin system (RAS). The RAS regulates fluid homeostasis and blood pressure and is a target for two classes of antihypertensive drugs. Beyond influencing vascular function, RAS polymorphisms may also increase the risk of depression via modulation of monoamines 155 or contributing to hypothalamic-pituitary-adrenal (HPA) axis dysregulation.156 Results from studies examining the common angiotensin converting enzyme (ACE) insertion/deletion (I/D) polymorphism are mixed in adult depression 157-162 and suicidality,162-164 although other ACE polymorphisms may be important.161, 165, 166 Despite its key role in the RAS, the gene encoding for the angiotensin II type 1 receptor (AGTR1) is less well studied. The CC genotype of the A1166C (rs5186) polymorphism is associated with increased responsiveness to Ang-II.167 However, despite one study associating the CC genotype with increased risk of MDD,159 this was not replicated in a elderly sample.168

RAS variation is further related to differences in brain structure and function. Several studies in older adults have now associated variants in ACE and AGTR1 with altered cerebral frontotemporal structure 169-172 and abnormal default mode network activity.173AGTR1 variants are associated with greater WMH progression, particularly in men,168 and this effect may be localized to specific tracts.60

RAS polymorphisms may also influence antidepressant outcomes. In a general adult population, the ACE D variant and the AGTR1 C1166 allele are associated with better acute antidepressant response.174, 175 However, this contrasts with a study of LLD, wherein AGTR1 C1166 allele homozygous individuals exhibited a poorer antidepressant response over a longer period.176 Such age differences could be important, reflecting stress reactivity in earlier life and allostatic effects of an overly active RAS later in life.

Mechanistic Hypotheses

According to a model by Alexopoulos,1 the clinical expression of LLD is mediated by altered brain activity in cognitive and affective circuits, characterized as hypometabolism of dorsal cortical regions and hypermetabolism of ventral limbic structures. Age-, disease-, and stress-related neurobiological changes serve as etiological factors contributing to alterations in circuits regulating mood and cognition. The impact of these neurobiological contributors can be increased by vulnerability factors originating from preexisting differences in mood circuitry.

Arguably, such etiological factors may lead to depression only after crossing a certain threshold above which they acquire a dose effect relationship (Figure 1). In this conceptualization, single or multiple potential “etiological factors” may reach an initial severity threshold and contribute to LLD “vulnerability,” due to frontolimbic compromise evident through imaging or abnormal performance on cognitive or affective tasks. When etiological factors further increase in severity and cross a second threshold, they may have a greater and direct effect on mood circuit function, leading to affective and cognitive symptomatology. Of course, instead of threshold effects such relationships could be cumulative following linear or curvilinear patterns

Figure 1.

Figure 1

Proposed progression of etiological processes affecting frontolimbic function. After crossing an initial threshold, vascular or other processes may serve as one of many factors that increase vulnerability to depression through compromise of affective and cognitive brain circuits. As the underlying disease process progresses, its adverse effects on the affective and cognitive circuits would likewise increase, resulting in the circuit dysfunctions that characterize depression.

Such a threshold model can account for multifactorial contributions to LLD. It also explains why our best clinical and biological measures predict only a modest part of the variance in distinguishing LLD from controls and in predicting treatment response as any individual measure may itself not cross a crucial threshold. This model allows for mixed syndromes of multiple etiological contributors if each disturbance alone or in combination is strong enough to cross the threshold required to induce neural circuit changes mediating LLD.

Given the central role of vascular disease and WMHs in much of the literature, it is worth examining their role within this model. WMHs can be conceptualized as visible markers of ischemic damage, although vascular compromise may result in insults to white and/or gray matter that are not visible on conventional neuroimaging.177 In this context, greater WMH severity represents a state of vascular compromise and ischemic injury in specific regions. Depending on the severity of the underlying WMLs and how strongly they affect affective and cognitive circuitry, such damage may itself predispose individuals to depressive episodes. However, WMHs may also be an index of the severity of underlying vascular disease affecting neural circuits. In this case, the direct effect of WMHs on neural circuits may be modest compared with that of the underlying vascular process.

The Disconnection Hypothesis

Building on the concept of “disconnection syndromes,” ischemia and WMLs may contribute to depression by disrupting neural connections among regions regulating mood and cognition.178 In this model, global cerebral WMH severity is less germane to LLD than is focal damage to specific fiber tracts and neural circuits. Such focal damage would adversely affect the tract's structural and functional connectivity. In turn this state adversely affects the function of connected regions at rest and during cognitive tasks and contribute to neural circuitry alterations that mediate clinical symptoms and influence antidepressant response.

This view is supported by studies examining WMH location. Recent work demonstrates that LLD is associated with greater WMH severity in specific tracts including the cingulum bundle, uncinate fasciculus, and superior longitudinal fasciculus.60-62 Additionally, greater WMH severity in the uncinate and superior longitudinal fasciculi is associated with executive dysfunction 61, 123, 124 and greater depression severity.179

These macrostructural MR findings are supported by diffusion tensor imaging (DTI) studies examining white matter microstructure. DTI changes reflect various pathologies leading to decreased myelin integrity, including demyelination secondary to cerebrovascular and inflammatory changes. WMLs occurring in fiber tracts are also associated with DTI changes.177, 180 LLD is associated with differences on DTI measures of the uncinate fasciculus, cingulum bundle, anterior thalamic radiation, and superior longitudinal fasciculus.181, 182 The uncinate fasciculus and cingulum bundle are particularly relevant to LLD 183-188 because of their role in cognition and emotion processing,188, 189 although other tracts are also associated with antidepressant response in LLD.149 Importantly, differences in the uncinate fasciculus and cingulum bundle are also observed in younger depressed adults.190, 191 Although those younger adult studies do not necessarily suggest a vascular cause in that population, they support that structural alterations in these fiber tracts are associated with depression.

Structural differences have functional effects. Fiber tract structural connectivity is positively correlated with resting state FC of connected regions.192-196 These data warrant a comprehensive examination of the relationships among ischemic focal WML injury, damage to tract microstructure, regional connectivity, and circuit function. Examination of such a “lesion model” can identify neural circuit deficits contributing to depression and predictive of poor outcomes.

The Inflammation Hypothesis

Aging- and disease-related processes promote proinflammatory states in older individuals.197, 198 Further, immune activation can be a characteristic of depression 48, 199 and precipitate depressive symptoms.200 Alexopoulos and Morimoto recently proposed that immune dysregulation may promote the development of affective and cognitive symptoms in LLD.201 This view is supported by studies in midlife adult populations. Administration of cytokines or induction of peripheral inflammation results in an inflammatory response, which in turn is correlated with fatigue, slowed reaction time,202 and mood reduction.200, 203 Even without medical illness, depressed individuals exhibit increased levels of proinflammatory cytokines 204, 205 and reduced anti-inflammatory cytokine levels.206, 207

Several mechanisms may explain these relationships. Proinflammatory cytokines affect monoamine neurotransmitter pathways,208 including indoleamine 2,3-dioxygenase upregulation and kynurenine pathway activation.209-211 This results in decreased tryptophan and serotonin and increased synthesis of detrimental tryptophan catabolites that promote hippocampal damage and apoptosis.211, 212 Cytokines, including IL-1β, also reduce extracellular serotonin levels by activating the serotonin transporter.213 Additionally, proinflammatory cytokines disrupt glucocorticoid receptor function 214 and reduce neurotrophic support.215

Peripheral inflammation is associated with altered brain function in areas mediating cognition and mood. In healthy individuals, typhoid-induced IL-6 levels correlate with lower mood, increased subgenual anterior cingulate cortex activity during affective processing, and altered subgenual cingulate connectivity.203 Laboratory-administered social stressors increase inflammatory maker levels that in turn are associated with activation of the dorsal anterior cingulate cortex and anterior insula.216 In bereaved women undergoing grief elicitation, inflammatory markers were positively associated with subgenual cingulate and orbitofrontal cortex activation.217

Supporting this work is a growing literature demonstrating that genetic polymorphisms promoting pro-inflammatory processes may contribute to depression 218, 219 and in LLD may be related to age of onset.220 Such relationships are likely moderated through an effect on cytokine levels that in turn are associated with sad mood 221 and altered emotion processing.222

Regarding LLD, the aging process disrupts immune function,223 increasing peripheral immune activity and shifting the CNS into a proinflammatory state.198 Elevated peripheral cytokine levels are associated with depressive symptoms in older adults, with the most consistent finding being for IL-6, but also implicating IL-1β, IL-8 and TNFα.224-228 Proinflammatory states in older adults are associated with cognitive deficits, including poorer executive function, poorer memory performance, worse global cognition, and steeper decline in cognition.229-231 Finally, greater IL-6 and C-reactive protein levels are associated with greater WMH burden.232-235

Successful antidepressant treatment may reduce proinflammatory markers.48 This action may be a direct effect as in vitro studies demonstrate that antidepressants reduce proinflammatory markers while increasing levels of anti-inflammatory cytokines.236-240 Results in clinical populations are inconsistent, but a recent meta-analysis concluded that antidepressants, particularly SSRIs, reduce IL-6, IL-1β, and TNFα.241 Additional evidence supports that some anti-inflammatory drugs and may have antidepressant properties,242-244 particularly in individuals with elevated pre-treatment proinflammatory markers.245

Importantly, pro-inflammatory processes also contribute to neurodegenerative processes. Increased peripheral inflammatory marker levels are associated with increased risk for all-cause dementia 246 and central inflammatory processes significantly contribute to the pathogenesis of Alzheimer's disease.247 However, nonsteroidal anti-inflammatory drugs have not consistently demonstrated clinical benefit in Alzheimer's disease 247 and in a recent consensus statement were considered but not recommended as a priority for further investigation.248

The Hypoperfusion Hypothesis

Vascular dysregulation is common in LLD 249-254 and CBF reductions can impair regional brain function, contributing to affective and cognitive symptoms. Regional cerebral metabolic activity is tightly correlated with CBF, which is regulated by local interactions between neurons, glia, and the vasculature.255, 256 Blood flow to the brain is influenced by systemic hemodynamics and cerebrovascular autoregulation, with cerebral arteries contracting or dilating as arterial pressure changes. These processes interact to maintain stable perfusion. However, these processes are impaired in the context of vascular disease:256 hypertension, diabetes, and atherosclerosis lead to vascular wall hypertrophy, reduced arterial lumen diameter, reduced arterial distensibility, and endothelial cell dysfunction.256-258 Such vascular changes, including increased intima media thickness, increased arterial stiffness, and endothelial dysfunction are pronounced in LLD populations 249-254 and endothelial function may be particularly poor in antidepressant nonresponders.259 Such vascular pathology results in reduced blood flow velocities and decreased vasomotor reactivity,260, 261 adversely affecting CBF.

Perfusion deficits do not need to cause ischemia in order to influence brain function. Reduced CBF impairs protein synthesis 262 crucial for cognitive processing 263, 264 and for maintaining the integrity of cortical functional maps.265 Thus mild CBF reduction may impair cognitive and affective processes, while greater CBF reduction in the context of autoregulatory deficits may cause ischemic injury. The subcortical white matter is particularly sensitive to these changes because it is supplied by terminal arterioles with limited collateral flow 266 and so susceptible to infarction due to impaired autoregulation.267 Greater WMH severity may be a marker of broader deficits in perfusion and autoregulation as individuals with greater WMH severity exhibit reduced CBF in both white matter and gray matter regions.268-270 Depressed elders with greater WMH severity also exhibit decreased perfusion in the cingulate gyrus,271 a region involved in cognitive and affective processing.272

PET, SPECT, and MRI arterial spin labeling (ASL) have identified CBF differences in midlife adult depressed populations.273-276 Advanced age is itself associated with decreased frontotemporal CBF,277 an effect mediated by vascular risk factors.278 In LLD, perfusion deficits are more severe, particularly in the medial and lateral prefrontal cortex (PFC), subcortical, and temporal structures.269, 279-283 Deficits in the dorsolateral PFC may improve with antidepressant response, while other perfusion deficits may not.281 Others report no change in CBF in antidepressant nonresponders,284-288 suggesting that persistently reduced regional CBF may be a biomarker of nonresponse.

Cerebral hypoperfusion is associated with Alzheimer's disease and mild cognitive impairment,289-291 but also with abnormal performance on specific cognitive tasks.291-294 For example, resting hippocampal blood flow is positively correlated with spatial memory performance.292 Although not specifically a measure of perfusion, reduced arterial CBF predicts slower processing speed,295 a domain that may mediate other deficits in LLD.103 Perfusion deficits may thus contribute to the affective and cognitive symptoms observed in LLD. Intriguingly, these data support investigating the utility of agents that may improve perfusion or restore endothelial function.296, 297

Role of Vascular Risk Factors in Late-Life Depression

Fifteen years after the formulation of the “vascular depression” hypothesis,2 a confluence of findings suggests that:

  1. Vascular risk factors and radiological findings are often associated with LLD.

  2. Cognitive deficits across multiple domains are common in LLD and associated with vascular risk factors and greater WMH severity. Such deficits, particularly executive dysfunction, predict poor response to antidepressants.

  3. WMHs, although associated with cognitive dysfunction, do not consistently predict poor antidepressant response. However, progressive WMH disease is associated with poorer depression course.

The link between progressive vascular disease and the development and persistence of LLD invites study of the underlying mechanisms by which vascular disease alters brain function in such a way to mediate the development of clinical symptoms. The ultimate goal of this line of study is to develop novel treatments or repurpose existent agents to improve LLD outcomes.

We propose three mechanistic paths to “vascular depression.” Although the processes of these pathways could proceed independently, they are complementary and interconnected and so may all contribute to LLD (Figure 2). For example, inflammation participates in vascular remodeling, accelerates vascular damage 298 and can contribute to perfusion deficits. In turn, regional perfusion deficits adversely affect gray matter function and contribute to the development of WMLs that disrupt regional structural and functional connectivity.

Figure 2.

Figure 2

This model links the disconnection, inflammation, and hypoperfusion processes proposed in late-life depression. Vascular disease may contribute to altered brain function characteristic of depression (dorsal hypometabolism, ventral hypermetabolism) either through structural damage adversely affecting connectivity, through perfusion deficits altering regional function, or both. Proinflammatory processes increase vascular risk but may also affect brain function through independent processes.

In this model, vascular disease is an important and central contributor to LLD. Not only is vascular disease common and perhaps unavoidable in later life, but it also interacts with other pathological processes related to depression. However, vascular disease is not the only contributor to LLD. Other biological and environmental factors can increase the risk for depression by alterating neural circuit structure and function. Similarly, genetic influences may increase risk of depression by acting at multiple points, such altering function of neural circuits, changing the biological response to stress, adding to vascular risk, or predisposing to proinflammatory states.

We are not proposing a single mechanism underlying LLD. Non-vascular factors clearly contribute to LLD. The same genetic, epigenetic, and environmental factors that contribute to depression in younger adults continue to confer vulnerability to depression in later life. Moreover, although alterations in immune and endocrine regulation affect vascular risk, they may also increase the risk of depression through independent mechanisms.

Just as there are non-vascular factors contributing to LLD, there may be additional mechanisms linking vascular disease with LLD. Altered homocysteine metabolism is associated with LLD,299 with hyperhomocysteinuria potentially contributing to vascular risk by increasing atherosclerosis and endothelial dysfunction.300 Another example is altered sympathoadrenomedullary system and hypothalamic-pituitary-adnenal (HPA) axis function 301 that, among other activities, affects inflammation.302 These and other mechanisms may contribute to the development or perpetuation of LLD both through vascular and non-vascular pathways and require further study. Such studies would benefit from the incorporation of genomic approaches along with measures of systemic function, thus linking molecular mechanisms with clinically salient measures and outcomes.

Demonstrating that such mechanisms affect neural circuit function opens the door for intervention studies examining whether modulation of these mechanisms can improve depression outcomes. There is already preliminary support for using anti-inflammatory drugs in depression. Etanercept, a soluble TNFα receptor, and celecoxib, a cyclo-oxygenase-2 inhibitor, may reduce depressive symptoms in patients with inflammatory diseases 242, 243 and infliximab may improve depression in patients with greater pre-treatment inflammation.245 Similarly, omega-3 fatty acids are anti-inflammatory and also affect monoamine neurotransmission,303 suggesting an antidepressant role for omega-3 supplementation. However, recent meta-analyses have not provided strong support for omega-3 supplementation for depression, although larger well designed trials may be warranted.304, 305 Finally, drugs that modulate the renin-angiotensin system improve cerebral perfusion 297 and may improve cognition,306-308 although they have not been well studied in depression. We suggest that study of these agents in LLD may produce a novel, targeted pharmacology for the large group of LLD patients who are unresponsive to conventional antidepressants.

Acknowledgments

Financial Support: This project was supported by NIH grants R01 MH077745, R01 MH076079, R01 MH079414, and P30 MH085943

Footnotes

CONFLICTS OF INTEREST

Dr. Taylor and Dr. Aizenstein report no conflicts of interest. Dr. Alexopoulos reports research grant support from Forest, serving as a consultant for Lilly, and serving on the speakers’ bureau for Astra Zeneca, Forest, Merck, Avanir, and Lundbeck. Dr. Alexopoulos also is a stockholder of Johnson & Johnson.

REFERENCES

  • 1.Alexopoulos GS. Depression in the elderly. Lancet. 2005;365:1961–1970. doi: 10.1016/S0140-6736(05)66665-2. [DOI] [PubMed] [Google Scholar]
  • 2.Alexopoulos GS, Meyers BS, Young RC, Campbell S, Silbersweig D, Charlson M. ‘Vascular depression’ hypothesis. Arch Gen Psychiatry. 1997;54(10):915–922. doi: 10.1001/archpsyc.1997.01830220033006. [DOI] [PubMed] [Google Scholar]
  • 3.Alexopoulos GS, Meyers BS, Young RC, Kakuma T, Silbersweig D, Charlson M. Clinically defined vascular depression. Am J Psychiatry. 1997;154(4):562–565. doi: 10.1176/ajp.154.4.562. [DOI] [PubMed] [Google Scholar]
  • 4.Alexopoulos GS. “The Depression-Executive Dysfunction Syndrome of Late Life”: a specific target for D3 agonists? Am J Geriatr Psychiatry. 2001;9:22–29. [PubMed] [Google Scholar]
  • 5.Krishnan KRR, Hays JC, Blazer DG. MRI-defined vascular depression. Am J Psychiatry. 1997;154(4):497–501. doi: 10.1176/ajp.154.4.497. [DOI] [PubMed] [Google Scholar]
  • 6.Sneed JR, Rindskopf D, Steffens DC, Krishnan KR, Roose SP. The vascular depression subtype: evidence of internal validity. Biol Psychiatry. 2008 Sep 15;64(6):491–497. doi: 10.1016/j.biopsych.2008.03.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Awad IA, Spetzler RF, Hodak JA, Awad CA, Carey R. Incidental subcortical lesions identified on magnetic resonance imaging in the elderly. I. Correlation with age and cerebrovascular risk factors. Stroke. 1986;17:1084–1089. doi: 10.1161/01.str.17.6.1084. [DOI] [PubMed] [Google Scholar]
  • 8.Dufouil C, de Kersaint-Gilly A, Besancon V, Levy C, Auffray E, Brunnereau L, et al. Longitudinal study of blood pressure and white matter hyperintensities. The EVA MRI cohort. Neurology. 2001;56:921–926. doi: 10.1212/wnl.56.7.921. [DOI] [PubMed] [Google Scholar]
  • 9.Longstreth WTJ, Manolio TA, Arnold A, Burke GL, Bryan N, Jungreis CA, et al. Clinical correlates of white matter findings on cranial magnetic resonance imaging of 3301 elderly people: the cardiovascular health study. Stroke. 1996;27:1274–1282. doi: 10.1161/01.str.27.8.1274. [DOI] [PubMed] [Google Scholar]
  • 10.Taylor WD, MacFall JR, Payne ME, McQuoid DR, Steffens DC, Provenzale JM, et al. Greater MRI lesion volumes in elderly depressed subjects than in control subjects. Psychiatry Res. 2005;139:1–7. doi: 10.1016/j.pscychresns.2004.08.004. [DOI] [PubMed] [Google Scholar]
  • 11.Taylor WD, MacFall JR, Provenzale JM, Payne ME, McQuoid DR, Steffens DC, et al. Serial MR imaging of hyperintense white matter lesion volumes in elderly subjects: correlation with vascular risk factors. Am J Roentgenol. 2003;181:571–576. doi: 10.2214/ajr.181.2.1810571. [DOI] [PubMed] [Google Scholar]
  • 12.Jokinen H, Kalska H, Ylikoski R, Madureira S, Verdelho A, Gouw A, et al. MRI-defined subcortical ischemic vascular disease: baseline clinical and neuropsychological findings. The LADIS Study. Cerebrovasc Dis. 2009;27(4):336–344. doi: 10.1159/000202010. [DOI] [PubMed] [Google Scholar]
  • 13.Pantoni L, Garcia JH, Gutierrez JA. Cerebral white matter is highly vulnerable to ischemia. Stroke. 1996 Sep;27(9):1641–1646. doi: 10.1161/01.str.27.9.1641. [DOI] [PubMed] [Google Scholar]
  • 14.Thomas AJ, O'Brien JT, Davis S, Ballard C, Barber R, Kalaria RN, et al. Ischemic basis for deep white matter hyperintensities in major depression. Arch Gen Psychiatry. 2002;59:785–792. doi: 10.1001/archpsyc.59.9.785. [DOI] [PubMed] [Google Scholar]
  • 15.Thomas AJ, Perry R, Barber R, Kalaria RN, O'Brien JT. Pathologies and pathological mechanisms for white matter hyperintensities in depression. Ann N Y Acad Sci. 2002;977:333–339. doi: 10.1111/j.1749-6632.2002.tb04835.x. [DOI] [PubMed] [Google Scholar]
  • 16.Taylor WD, McQuoid DR, Krishnan KR. Medical comorbidity in late-life depression. Int J Geriatr Psychiatry. 2004;19:935–943. doi: 10.1002/gps.1186. [DOI] [PubMed] [Google Scholar]
  • 17.Vasudev A, O'Brien JT, Tan MP, Parry SW, Thomas AJ. A study of orthostatic hypotension, heart rate variability and baroreflex sensitivity in late-life depression. J Affect Disord. 2011 Jun;131(1-3):374–378. doi: 10.1016/j.jad.2010.11.001. [DOI] [PubMed] [Google Scholar]
  • 18.Lavretsky H, Lesser IM, Wohl M, Miller BL. Relationship of age, age at onset, and sex to depression in older adults. Am J Geriatr Psychiatry. 1998;6:248–256. [PubMed] [Google Scholar]
  • 19.Matsubayashi K, Okumiya K, Wada T, Osaki Y, Fujisawa M, Doi Y, et al. Postural dysregulation in systolic blood pressure is associated with worsened scoring on neurobehavioral function tests and leukoaraiosis in the older elderly living in a community. Stroke. 1997 Nov;28(11):2169–2173. doi: 10.1161/01.str.28.11.2169. [DOI] [PubMed] [Google Scholar]
  • 20.Puisieux F, Monaca P, Deplanque D, Delmaire C, di Pompeo C, Monaca C, et al. Relationship between leuko-araiosis and blood pressure variability in the elderly. Eur Neurol. 2001;46(3):115–120. doi: 10.1159/000050783. [DOI] [PubMed] [Google Scholar]
  • 21.Isaka Y, Okamoto M, Ashida K, Imaizumi M. Decreased cerebrovascular dilatory capacity in subjects with asymptomatic periventricular hyperintensities. Stroke. 1994 Feb;25(2):375–381. doi: 10.1161/01.str.25.2.375. [DOI] [PubMed] [Google Scholar]
  • 22.Bakker SL, de Leeuw FE, de Groot JC, Hofman A, Koudstaal PJ, Breteler MM. Cerebral vasomotor reactivity and cerebral white matter lesions in the elderly. Neurology. 1999 Feb;52(3):578–583. doi: 10.1212/wnl.52.3.578. [DOI] [PubMed] [Google Scholar]
  • 23.Marstrand JR, Garde E, Rostrup E, Ring P, Rosenbaum S, Mortensen EL, et al. Cerebral perfusion and cerebrovascular reactivity are reduced in white matter hyperintensities. Stroke. 2002;33:972–976. doi: 10.1161/01.str.0000012808.81667.4b. [DOI] [PubMed] [Google Scholar]
  • 24.Oishi M, Mochizuki Y. Regional cerebral blood flow and cerebrospinal fluid glutamate in leukoaraiosis. J Neurol. 1998 Dec;245(12):777–780. doi: 10.1007/s004150050286. [DOI] [PubMed] [Google Scholar]
  • 25.Coffey CE, Figiel GS, Djang WT, Cress M, Saunders WB, Weiner RD. Leukoencephalopathy in elderly depressed patients referred for ECT. Biol Psychiatry. 1988;24(2):143–161. doi: 10.1016/0006-3223(88)90270-3. [DOI] [PubMed] [Google Scholar]
  • 26.Coffey CE, Figiel GS, Djang WT, Saunders WB, Weiner RD. White matter hyperintensities on magnetic resonance imaging: clinical and anatomic correlates in the depressed elderly. Journal of Neuropsychiatry & Clinical Neurosciences. 1989;1(2):135–144. doi: 10.1176/jnp.1.2.135. [DOI] [PubMed] [Google Scholar]
  • 27.Dolan RJ, Poynton AM, Bridges PK, Trimble MR. Altered magnetic resonance white-matter T1 values in patients with affective disorder. Br J Psychiatry. 1990;157:107–110. doi: 10.1192/bjp.157.1.107. [DOI] [PubMed] [Google Scholar]
  • 28.Fujikawa T, Yamawaki S, Touhouda Y. Incidence of silent cerebral infarction in patients with major depression. Stroke. 1993;24:1631–1634. doi: 10.1161/01.str.24.11.1631. [DOI] [PubMed] [Google Scholar]
  • 29.Greenwald BS, Kramer-Ginsberg E, Krishnan KRR, Ashtari M, Aupperle PM, Patel M. MRI signal hyperintensities in geriatric depression. Am J Psychiatry. 1996;153:1212–1215. doi: 10.1176/ajp.153.9.1212. [DOI] [PubMed] [Google Scholar]
  • 30.Kumar A, Bilker W, Jin Z, Udupa J. Atrophy and high intensity lesions: complementary neurobiological mechanisms in late-life depression. Neuropsychopharmacology. 2000;22:264–274. doi: 10.1016/S0893-133X(99)00124-4. [DOI] [PubMed] [Google Scholar]
  • 31.Herrmann LL, Le Masurier M, Ebmeier KP. White matter hyperintensities in late life depression: a systematic review. J Neurol Neurosurg Psychiatry. 2008 Jun;79(6):619–624. doi: 10.1136/jnnp.2007.124651. [DOI] [PubMed] [Google Scholar]
  • 32.Steffens DC, Krishnan KRR. Structural neuroimaging and mood disorders: recent findings, implications for classification, and future directions. Biol Psychiatry. 1998;43:705–712. doi: 10.1016/s0006-3223(98)00084-5. [DOI] [PubMed] [Google Scholar]
  • 33.Krishnan KRR, Tayor WD, McQuoid DR, MacFall JR, Payne ME, Provenzale JM, et al. Clinical characteristics of magnetic resonance imaging-defined subcortical ischemic depression. Biol Psychiatry. 2004;55:390–397. doi: 10.1016/j.biopsych.2003.08.014. [DOI] [PubMed] [Google Scholar]
  • 34.de Groot JC, de Leeuw F, Oudkerk M, Hofman A, Jolles J, Breteler MMB. Cerebral white matter lesions and depressive symptoms in elderly adults. Arch Gen Psychiatry. 2000;57:1071–1076. doi: 10.1001/archpsyc.57.11.1071. [DOI] [PubMed] [Google Scholar]
  • 35.Figiel GS, Krishnan KRR, Doraiswamy PM, Rao VP, Nemeroff CB, Boyko OB. Subcortical hyperintensities on brain magnetic resonance imaging: a comparison between late age onset and early onset elderly depressed subjects. Neurobiol Aging. 1991;12(3):245–247. doi: 10.1016/0197-4580(91)90104-r. [DOI] [PubMed] [Google Scholar]
  • 36.Hickie I, Scott E, Mitchell P, Wilhelm K, Austin MP, Bennett B. Subcortical hyperintensities on magnetic resonance imaging: clinical correlates and prognostic significance in patients with severe depression. Biol Psychiatry. 1995;37(3):151–160. doi: 10.1016/0006-3223(94)00174-2. [DOI] [PubMed] [Google Scholar]
  • 37.Salloway S, Malloy P, Kohn R, Gillard E, Duffy J, Rogg J, et al. MRI and neuropsychological differences in early- and late-life-onset geriatric depression. Neurology. 1996;46(6):1567–1574. doi: 10.1212/wnl.46.6.1567. [DOI] [PubMed] [Google Scholar]
  • 38.Kumar A, Bilker W, Jin Z, Udupa J, Gottlieb G. Age of onset of depression and quantitative neuroanatomic measures: absence of specific correlates. Psychiatry Res. 1999;91:101–110. doi: 10.1016/s0925-4927(99)00021-9. [DOI] [PubMed] [Google Scholar]
  • 39.Dillon C, Allegri RF, Serrano CM, Iturry M, Salgado P, Glaser FB, et al. Late- versus early-onset geriatric depression in a memory research center. Neuropsychiatr Dis Treat. 2009;5:517–526. doi: 10.2147/ndt.s7320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rapp MA, Dahlman K, Sano M, Grossman HT, Haroutunian V, Gorman JM. Neuropsychological differences between late-onset and recurrent geriatric major depression. Am J Psychiatry. 2005 Apr;162(4):691–698. doi: 10.1176/appi.ajp.162.4.691. [DOI] [PubMed] [Google Scholar]
  • 41.Wassertheil-Smoller S, Applegate WB, Berge K, Chang CJ, Davis BR, Grimm R, Jr, et al. Change in depression as a precursor of cardiovascular events. SHEP Cooperative Research Group (Systoloc Hypertension in the elderly). Arch Intern Med. 1996 Mar 11;156(5):553–561. [PubMed] [Google Scholar]
  • 42.Krishnan M, Mast BT, Ficker LJ, Lawhorne L, Lichtenberg PA. The effects of preexisting depression on cerebrovascular health outcomes in geriatric continuing care. J Gerontol A Biol Sci Med Sci. 2005 Jul;60(7):915–919. doi: 10.1093/gerona/60.7.915. [DOI] [PubMed] [Google Scholar]
  • 43.Frasure-Smith N, Lesperance F, Talajic M. Depression and 18-month prognosis after myocardial infarction. Circulation. 1995;91:999–1005. doi: 10.1161/01.cir.91.4.999. [DOI] [PubMed] [Google Scholar]
  • 44.Musselman DL, Evans DL, Nemeroff CB. The relationship of depression to cardiovascular disease: Epidemiology, biology, and treatment. Arch Gen Psychiatry. 1998;55:580–592. doi: 10.1001/archpsyc.55.7.580. [DOI] [PubMed] [Google Scholar]
  • 45.Glassman AH, Bigger JT, Gaffney M, Shapiro PA, Swenson JR. Onset of major depression associated with acute coronary syndromes: relationship of onset, major depressive disorder history, and episode severity to sertraline benefit. Arch Gen Psychiatry. 2006 Mar;63(3):283–288. doi: 10.1001/archpsyc.63.3.283. [DOI] [PubMed] [Google Scholar]
  • 46.Frasure-Smith N, Lesperance F, Irwin MR, Sauve C, Lesperance J, Theroux P. Depression, C-reactive protein and two-year major adverse cardiac events in men after acute coronary syndromes. Biol Psychiatry. 2007 Aug 15;62(4):302–308. doi: 10.1016/j.biopsych.2006.09.029. [DOI] [PubMed] [Google Scholar]
  • 47.Surtees PG, Wainwright NW, Boekholdt SM, Luben RN, Wareham NJ, Khaw KT. Major depression, C-reactive protein, and incident ischemic heart disease in healthy men and women. Psychosom Med. 2008 Oct;70(8):850–855. doi: 10.1097/PSY.0b013e318183acd5. [DOI] [PubMed] [Google Scholar]
  • 48.Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol Psychiatry. 2009 May 1;65(9):732–741. doi: 10.1016/j.biopsych.2008.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zill P, Baghai TC, Schule C, Born C, Frustuck C, Buttner A, et al. DNA Methylation Analysis of the Angiotensin Converting Enzyme (ACE) Gene in Major Depression. PLoS ONE. 2012;7(7):e40479. doi: 10.1371/journal.pone.0040479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.van den Heuvel DM, ten Dam VH, de Craen AJ, Admiraal-Behloul F, Olofsen H, Bollen EL, et al. Increase in periventricular white matter hyperintensities parallels decline in mental processing speed in a non-demented elderly population. J Neurol Neurosurg Psychiatry. 2006;77:149–153. doi: 10.1136/jnnp.2005.070193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Debette S, Bombois S, Bruandet A, Delbeuck X, Lepoittevin S, Delmaire C, et al. Subcortical hyperintensities are associated with cognitive decline in patients with mild cognitive impairment. Stroke. 2007 Nov;38(11):2924–2930. doi: 10.1161/STROKEAHA.107.488403. [DOI] [PubMed] [Google Scholar]
  • 52.de Groot JC, De Leeuw FE, Oudkerk M, Van Gijn J, Hofman A, Jolles J, et al. Periventricular cerebral white matter lesions predict rate of cognitive decline. Ann Neurol. 2002 Sep;52(3):335–341. doi: 10.1002/ana.10294. [DOI] [PubMed] [Google Scholar]
  • 53.Filley CM. The behavioral neurology of cerebral white matter. Neurology. 1998 Jun;50(6):1535–1540. doi: 10.1212/wnl.50.6.1535. [DOI] [PubMed] [Google Scholar]
  • 54.DeCarli C, Fletcher E, Ramey V, Harvey D, Jagust WJ. Anatomical mapping of white matter hyperintensities (WMH): exploring the relationships between periventricular WMH, deep WMH, and total WMH burden. Stroke. 2005 Jan;36(1):50–55. doi: 10.1161/01.STR.0000150668.58689.f2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Firbank MJ, Lloyd AJ, Ferrier N, O'Brien JT. A volumetric study of MRI signal hyperintensities in late-life depression. Am J Geriatr Psychiatry. 2004;12:606–612. doi: 10.1176/appi.ajgp.12.6.606. [DOI] [PubMed] [Google Scholar]
  • 56.MacFall JR, Payne ME, Provenzale JM, Krishnan KRR. Medial orbital frontal lesions in late-onset depression. Biol Psychiatry. 2001;49:803–806. doi: 10.1016/s0006-3223(00)01113-6. [DOI] [PubMed] [Google Scholar]
  • 57.MacFall JR, Taylor WD, Rex DE, Pieper S, Payne ME, McQuoid DR, et al. Lobar distribution of lesion volumes in late-life depression: the Biomedical Informatics Research Network (BIRN). Neuropsychopharmacology. 2005;31:1500–1507. doi: 10.1038/sj.npp.1300986. [DOI] [PubMed] [Google Scholar]
  • 58.Taylor WD, MacFall JR, Steffens DC, Payne ME, Provenzale JM, Krishnan KRR. Localization of age-associated white matter hyperintensities in late-life depression. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:539–544. doi: 10.1016/S0278-5846(02)00358-5. [DOI] [PubMed] [Google Scholar]
  • 59.O'Brien JT, Firbank MJ, Krishnan MS, van Straaten EC, van der Flier WM, Petrovic K, et al. White matter hyperintensities rather than lacunar infarcts are associated with depressive symptoms in older people: the LADIS study. Am J Geriatr Psychiatry. 2006 Oct;14(10):834–841. doi: 10.1097/01.JGP.0000214558.63358.94. [DOI] [PubMed] [Google Scholar]
  • 60.Taylor WD, Zhao Z, Ashley-Koch A, Payne ME, Steffens DC, Krishnan RR, et al. Fiber tract-specific white matter lesion severity Findings in late-life depression and by AGTR1 A1166C genotype. Hum Brain Mapp. 2011 Oct 22; doi: 10.1002/hbm.21445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Sheline YI, Price JL, Vaishnavi SN, Mintun MA, Barch DM, Epstein AA, et al. Regional white matter hyperintensity burden in automated segmentation distinguishes late-life depressed subjects from comparison subjects matched for vascular risk factors. Am J Psychiatry. 2008 Apr;165(4):524–532. doi: 10.1176/appi.ajp.2007.07010175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Dalby RB, Chakravarty MM, Ahdidan J, Sorensen L, Frandsen J, Jonsdottir KY, et al. Localization of white-matter lesions and effect of vascular risk factors in late-onset major depression. Psychol Med. 2010 Aug;40(8):1389–1399. doi: 10.1017/S0033291709991656. [DOI] [PubMed] [Google Scholar]
  • 63.Patel MJ, Boada FE, Price JC, Sheu LK, Reynolds CF, 3rd, Aizenstein HJ. Association of small vessel ischemic white matter changes with BOLD fMRI imaging in the elderly. Psychiatry Res. 2012 doi: 10.1016/j.pscychresns.2012.09.006. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mayda AB, Westphal A, Carter CS, DeCarli C. Late life cognitive control deficits are accentuated by white matter disease burden. Brain. 2011 Jun;134(Pt 6):1673–1683. doi: 10.1093/brain/awr065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Venkatraman VK, Aizenstein H, Guralnik J, Newman AB, Glynn NW, Taylor C, et al. Executive control function, brain activation and white matter hyperintensities in older adults. Neuroimage. 2010 Feb 15;49(4):3436–3442. doi: 10.1016/j.neuroimage.2009.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wu M, Andreescu C, Butters MA, Tamburo R, Reynolds CF, 3rd, Aizenstein H. Default-mode network connectivity and white matter burden in late-life depression. Psychiatry Res. 2011 Oct 31;194(1):39–46. doi: 10.1016/j.pscychresns.2011.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Aizenstein HJ, Andreescu C, Edelman KL, Cochran JL, Price J, Butters MA, et al. fMRI correlates of white matter hyperintensities in late-life depression. Am J Psychiatry. 2011 Oct;168(10):1075–1082. doi: 10.1176/appi.ajp.2011.10060853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Fazekas F, Kleinert R, Offenbacher H, Schmidt R, Kleinert G, Payer F, et al. Pathologic correlates of incidental MRI white matter signal hyperintensities. Neurology. 1993;43:1683–1689. doi: 10.1212/wnl.43.9.1683. [DOI] [PubMed] [Google Scholar]
  • 69.Chimowitz MI, Estes ML, Furlan AJ, Awad IA. Further observations on the pathology of subcortical lesions identified on magnetic resonance imaging. Arch Neurol. 1992;49:747–752. doi: 10.1001/archneur.1992.00530310095018. [DOI] [PubMed] [Google Scholar]
  • 70.Schmidt R, Schmidt H, Haybaeck J, Loitfelder M, Weis S, Cavalieri M, et al. Heterogeneity in age-related white matter changes. Acta neuropathologica. 2011 Aug;122(2):171–185. doi: 10.1007/s00401-011-0851-x. [DOI] [PubMed] [Google Scholar]
  • 71.Thomas AJ, Ferrier IN, Kalaria RN, Davis S, O'Brien JT. Cell adhesion molecule expression in the dorsolateral prefrontal cortex and anterior cingulate cortex in major depression in the elderly. Br J Psychiatry. 2002;181:129–134. doi: 10.1017/s0007125000161847. [DOI] [PubMed] [Google Scholar]
  • 72.Thomas AJ, Ferrier IN, Kalaria RN, Woodward SA, Ballard C, Oakley A, et al. Elevation in late-life depression of intercellular adhesion molecule-1 expression in the dorsolateral prefrontal cortex. Am J Psychiatry. 2000;157:1682–1684. doi: 10.1176/appi.ajp.157.10.1682. [DOI] [PubMed] [Google Scholar]
  • 73.Thomas AJ, Perry R, Kalaria RN, Oakley A, McMeekin W, O'Brien JT. Neuropathological evidence for ischemia in the white matter of the dorsolateral prefrontal cortex in late-life depression. Int J Geriatr Psychiatry. 2003;18:7–13. doi: 10.1002/gps.720. [DOI] [PubMed] [Google Scholar]
  • 74.Rajkowska G, Miguel-Hidalgo JJ, Dubey P, Stockmeier CA, Krishnan KR. Prominent reduction in pyramidal neurons density in the orbitofrontal cortex of elderly depressed patients. Biol Psychiatry. 2005;58:297–306. doi: 10.1016/j.biopsych.2005.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Miguel-Hidalgo JJ, Jiang W, Konick L, Overholser JC, Jurjus GJ, Stockmeier CA, et al. Morphometric analysis of vascular pathology in the orbitofrontal cortex of older subjects with major depression. Int J Geriatr Psychiatry. 2012 Dec 3; doi: 10.1002/gps.3911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Miguel-Hidalgo JJ, Overholser JC, Jurjus GJ, Meltzer HY, Dieter L, Konick L, et al. Vascular and extravascular immunoreactivity for intercellular adhesion molecule 1 in the orbitofrontal cortex of subjects with major depression: age-dependent changes. J Affect Disord. 2011 Aug;132(3):422–431. doi: 10.1016/j.jad.2011.03.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Santos M, Gold G, Kovari E, Herrmann FR, Hof PR, Bouras C, et al. Neuropathological analysis of lacunes and microvascular lesions in late-onset depression. Neuropathol Appl Neurobiol. 2010 Dec;36(7):661–672. doi: 10.1111/j.1365-2990.2010.01101.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Tsopelas C, Stewart R, Savva GM, Brayne C, Ince P, Thomas A, et al. Neuropathological correlates of late-life depression in older people. Br J Psychiatry. 2011 Feb;198(2):109–114. doi: 10.1192/bjp.bp.110.078816. [DOI] [PubMed] [Google Scholar]
  • 79.Thomas AJ, Ferrier IN, Kalaria RN, Perry RH, Brown A, O'Brien JT. A neuropathological study of vascular factors in late-life depression. J Neurol Neurosurg Psychiatry. 2001;70:83–87. doi: 10.1136/jnnp.70.1.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Murphy CF, Gunning-Dixon FM, Hoptman MJ, Lim KO, Ardekani B, Shields JK, et al. White-matter integrity predicts Stroop performance in patients with geriatric depression. Biol Psychiatry. 2007;61:1007–1010. doi: 10.1016/j.biopsych.2006.07.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lockwood KA, Alexopoulos GS, van Gorp WG. Executive dysfunction in geriatric depression. Am J Psychiatry. 2002;159:1119–1126. doi: 10.1176/appi.ajp.159.7.1119. [DOI] [PubMed] [Google Scholar]
  • 82.Elderkin-Thompson V, Kumar A, Bilker WB, Dunkin JJ, Mintz J, Moberg PJ, et al. Neuropsychological deficits among patients with late-onset minor and major depression. Archives of Clinical Neuropsychology. 2003 Jul;18:529–549. doi: 10.1016/s0887-6177(03)00022-2. [DOI] [PubMed] [Google Scholar]
  • 83.Nebes RD, Butters MA, Houck PR, Zmuda MD, Aizenstein H, Pollock BG, et al. Dual-task performance in depressed geriatric patients. Psychiatry Res. 2001 Jun 1;102:139–151. doi: 10.1016/s0165-1781(01)00244-x. [DOI] [PubMed] [Google Scholar]
  • 84.Boone KB, Lesser IM, Miller BL, Wohl M, Berman N, Lee ABP, et al. Cognitive functioning in older depressed outpatients: relationship of presence and severity of depression to neuropsychological test scores. Neuropsychology. 1995;9:390–398. [Google Scholar]
  • 85.Beats BC, Sahakian BJ, Levy R. Cognitive performance in tests sensitive to frontal lobe dysfunction in the elderly depressed. Psychol Med. 1996;26(3):591–603. doi: 10.1017/s0033291700035662. [DOI] [PubMed] [Google Scholar]
  • 86.Lesser IM, Boone KB, Mehringer CM, Wohl MA, Miller BL, Berman NG. Cognition and white matter hyperintensities in older depressed patients. Am J Psychiatry. 1996;153:1280–1287. doi: 10.1176/ajp.153.10.1280. [DOI] [PubMed] [Google Scholar]
  • 87.Sexton CE, McDermott L, Kalu UG, Herrmann LL, Bradley KM, Allan CL, et al. Exploring the pattern and neural correlates of neuropsychological impairment in late-life depression. Psychol Med. 2011 Oct;26:1–8. doi: 10.1017/S0033291711002352. [DOI] [PubMed] [Google Scholar]
  • 88.Herrmann LL, Goodwin GM, Ebmeier KP. The cognitive neuropsychology of depression in the elderly. Psychol Med. 2007 Dec;37(12):1693–1702. doi: 10.1017/S0033291707001134. [DOI] [PubMed] [Google Scholar]
  • 89.Alexopoulos GS, Kiosses DN, Klimstra S, Kalayam B, Bruce ML. Clinical presentation of the “depression-executive dysfunction syndrome” of late life. Am J Geriatr Psychiatry. 2002;10:98–106. [PubMed] [Google Scholar]
  • 90.Kim BS, Lee DH, Lee DW, Bae JN, Chang SM, Kim S, et al. The role of vascular risk factors in the development of DED syndrome among an elderly community sample. Am J Geriatr Psychiatry. 2011 Feb;19(2):104–114. doi: 10.1097/JGP.0b013e31820119b6. [DOI] [PubMed] [Google Scholar]
  • 91.Hajjar I, Yang F, Sorond F, Jones RN, Milberg W, Cupples LA, et al. A novel aging phenotype of slow gait, impaired executive function, and depressive symptoms: relationship to blood pressure and other cardiovascular risks. J Gerontol A Biol Sci Med Sci. 2009 Sep;64(9):994–1001. doi: 10.1093/gerona/glp075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Alexopoulos GS, Kiosses DN, Murphy C, Heo M. Executive dysfunction, heart disease burden, and remission of geriatric depression. Neuropsychopharmacology. 2004;29:2278–2284. doi: 10.1038/sj.npp.1300557. [DOI] [PubMed] [Google Scholar]
  • 93.Bogner HR, Bruce ML, Reynolds CF, 3rd, Mulsant BH, Cary MS, Morales K, et al. The effects of memory, attention, and executive dysfunction on outcomes of depression in a primary care intervention trial: the PROSPECT study. Int J Geriatr Psychiatry. 2007 Sep;22(9):922–929. doi: 10.1002/gps.1767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kalayam B, Alexopoulos GS. Prefrontal dysfunction and treatment response in geriatric depression. Arch Gen Psychiatry. 1999;56:713–718. doi: 10.1001/archpsyc.56.8.713. [DOI] [PubMed] [Google Scholar]
  • 95.Potter GG, Kittinger JD, Wagner HR, Steffens DC, Krishnan KR. Prefrontal neuropsychological predictors of treatment remission in late-life depression. Neuropsychopharmacology. 2004;29:2266–2271. doi: 10.1038/sj.npp.1300551. [DOI] [PubMed] [Google Scholar]
  • 96.Baldwin R, Jeffries S, Jackson A, Sutcliffe C, Thacker N, Scott M, et al. Treatment response in late-onset depression: relationship to neuropsychological, neuroradiological and vascular risk factors. Psychol Med. 2004;34:125–136. doi: 10.1017/s0033291703008870. [DOI] [PubMed] [Google Scholar]
  • 97.Sneed JR, Roose SP, Keilp JG, Krishnan KR, Alexopoulos GS, Sackeim HA. Response inhibition predicts poor antidepressant treatment response in very old depressed patients. Am J Geriatr Psychiatry. 2007 Jul;15:553–563. doi: 10.1097/JGP.0b013e3180302513. [DOI] [PubMed] [Google Scholar]
  • 98.Sheline YI, Pieper CF, Barch DM, Welsh-Boehmer K, McKinstry RC, MacFall JR, et al. Support for the vascular depression hypothesis in late-life depression: results of a 2-site, prospective, antidepressant treatment trial. Arch Gen Psychiatry. 2010 Mar;67(3):277–285. doi: 10.1001/archgenpsychiatry.2009.204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Bella R, Pennisi G, Cantone M, Palermo F, Pennisi M, Lanza G, et al. Clinical presentation and outcome of geriatric depression in subcortical ischemic vascular disease. Gerontology. 2010;56(3):298–302. doi: 10.1159/000272003. [DOI] [PubMed] [Google Scholar]
  • 100.Nebes RN, Pollock BG, Houck PR, Butters MA, Mulsant BH, Zmuda MD, et al. Persistence of cognitive impairment in geriatric patients following antidepressant treament: a randomized, double-blind clinical trial with nortriptyline and paroxetine. J Psychiatr Res. 2003;37:99–108. doi: 10.1016/s0022-3956(02)00085-7. [DOI] [PubMed] [Google Scholar]
  • 101.Kramer-Ginsberg E, Greenwald BS, Krishnan KRR, Christiansen B, Hu J, Ashtari M, et al. Neuropsychological functioning and MRI signal hyperintensities in geraitric depression. Am J Psychiatry. 1999;156:438–444. doi: 10.1176/ajp.156.3.438. [DOI] [PubMed] [Google Scholar]
  • 102.Kohler S, Thomas AJ, Lloyd A, Barber R, Almeida OP, O'Brien JT. White matter hyperintensities, cortisol levels, brain atrophy and continuing cognitive deficits in late-life depression. Br J Psychiatry. 2010 Feb;196(2):143–149. doi: 10.1192/bjp.bp.109.071399. [DOI] [PubMed] [Google Scholar]
  • 103.Butters MA, Whyte EM, Nebes RD, Begley AE, Dew MA, Mulsant BH, et al. The nature and determinants of neuropsychological functioning in late-life depression. Arch Gen Psychiatry. 2004;61:587–595. doi: 10.1001/archpsyc.61.6.587. [DOI] [PubMed] [Google Scholar]
  • 104.Sheline YI, Barch DM, Garcia K, Gersing K, Piper C, Welsh-Bohmer KA, et al. Cognitive function in late life depression: relationships to depression severity, cerebrovascular risk factors and processing speed. Biol Psychiatry. 2006;60:58–65. doi: 10.1016/j.biopsych.2005.09.019. [DOI] [PubMed] [Google Scholar]
  • 105.Nebes RD, Butters MA, Mulsant BH, Pollock BG, Zmuda MD, Houck PR, et al. Decreased working memory and processing speed mediate cognitive impairment in geriatric depression. Psychol Med. 2000;30:679–691. doi: 10.1017/s0033291799001968. [DOI] [PubMed] [Google Scholar]
  • 106.Delaloye C, Baudois S, de Bilbao F, Dubois Remund C, Hofer F, Lamon M, et al. Cognitive impairment in late-onset depression. Limited to a decrement in information processing resources? Eur Neurol. 2008;60(3):149–154. doi: 10.1159/000144086. [DOI] [PubMed] [Google Scholar]
  • 107.Bhalla RK, Butters MA, Mulsant BH, Begley AE, Zmuda MD, Schoderbek B, et al. Persistence of neuropsychologic deficits in the remitted state of late-life depression. Am J Geriatr Psychiatry. 2006 May;14(5):419–427. doi: 10.1097/01.JGP.0000203130.45421.69. [DOI] [PubMed] [Google Scholar]
  • 108.Butters MA, Becker JT, Nebes RD, Zmuda MD, Mulsant BH, Pollock BG, et al. Changes in cognitive functioning following treatment of late-life depression. Am J Psychiatry. 2000;157:1949–1954. doi: 10.1176/appi.ajp.157.12.1949. [DOI] [PubMed] [Google Scholar]
  • 109.Murphy CF, Alexopoulos GS. Longitudinal association of initiation/perseveration and severity of geriatric depression. Am J Geriatr Psychiatry. 2004 Jan-Feb;12(1):50–56. [PubMed] [Google Scholar]
  • 110.Vasudev A, Saxby BK, O'Brien J T, Colloby SJ, Firbank MJ, Brooker H, et al. Relationship between cognition, magnetic resonance white matter hyperintensities, and cardiovascular autonomic changes in late-life depression. Am J Geriatr Psychiatry. 2012 Aug;20(8):691–699. doi: 10.1097/JGP.0b013e31824c0435. [DOI] [PubMed] [Google Scholar]
  • 111.Perlmuter LC, Sarda G, Casavant V, O'Hara K, Hindes M, Knott PT, et al. A review of orthostatic blood pressure regulation and its association with mood and cognition. Clinical autonomic research : official journal of the Clinical Autonomic Research Society. 2012 Apr;22(2):99–107. doi: 10.1007/s10286-011-0145-3. [DOI] [PubMed] [Google Scholar]
  • 112.Novak V, Hajjar I. The relationship between blood pressure and cognitive function. Nature reviews Cardiology. 2010 Dec;7(12):686–698. doi: 10.1038/nrcardio.2010.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Wolf PA, D'Agostino RB, Belanger AJ, Kannel WB. Probability of stroke: A risk profile from the Framingham Study. Stroke. 1991;22:312–318. doi: 10.1161/01.str.22.3.312. [DOI] [PubMed] [Google Scholar]
  • 114.Gunning-Dixon FM, Raz N. Neuroanatomical correlates of selected executive functions in middle-aged and older adults: a prospective MRI study. Neuropsychologia. 2003;41(14):1929–1941. doi: 10.1016/s0028-3932(03)00129-5. [DOI] [PubMed] [Google Scholar]
  • 115.Kramer JH, Mungas D, Reed BR, Wetzel ME, Burnett MM, Miller BL, et al. Longitudinal MRI and cognitive change in healthy elderly. Neuropsychology. 2007 Jul;21(4):412–418. doi: 10.1037/0894-4105.21.4.412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Vannorsdall TD, Waldstein SR, Kraut M, Pearlson GD, Schretlen DJ. White matter abnormalities and cognition in a community sample. Archives of Clinical Neuropsychology. 2009 May;24(3):209–217. doi: 10.1093/arclin/acp037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Wright CB, Festa JR, Paik MC, Schmiedigen A, Brown TR, Yoshita M, et al. White matter hyperintensities and subclinical infarction: associations with psychomotor speed and cognitive flexibility. Stroke. 2008 Mar;39(3):800–805. doi: 10.1161/STROKEAHA.107.484147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.de Groot JC, de Leeuw FE, Oudkerk M, van Gijn J, Hofman A, Jolles J, et al. Cerebral white matter lesions and cognitive function: the Rotterdam Scan Study. Ann Neurol. 2000 Feb;47(2):145–151. doi: 10.1002/1531-8249(200002)47:2<145::aid-ana3>3.3.co;2-g. [DOI] [PubMed] [Google Scholar]
  • 119.Au R, Massaro JM, Wolf PA, Young ME, Beiser A, Seshadri S, et al. Association of white matter hyperintensity volume with decreased cognitive functioning: the Framingham Heart Study. Arch Neurol. 2006 Feb;63(2):246–250. doi: 10.1001/archneur.63.2.246. [DOI] [PubMed] [Google Scholar]
  • 120.Marquine MJ, Attix DK, Goldstein LB, Samsa GP, Payne ME, Chelune GJ, et al. Differential patterns of cognitive decline in anterior and posterior white matter hyperintensity progression. Stroke. 2010 Sep;41(9):1946–1950. doi: 10.1161/STROKEAHA.110.587717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Longstreth WT, Jr., Arnold AM, Beauchamp NJ, Jr., Manolio TA, Lefkowitz D, Jungreis C, et al. Incidence, manifestations, and predictors of worsening white matter on serial cranial magnetic resonance imaging in the elderly: the Cardiovascular Health Study. Stroke. 2005 Jan;36(1):56–61. doi: 10.1161/01.STR.0000149625.99732.69. [DOI] [PubMed] [Google Scholar]
  • 122.Silbert LC, Nelson C, Howieson DB, Moore MM, Kaye JA. Impact of white matter hyperintensity volume progression on rate of cognitive and motor decline. Neurology. 2008 Jul 8;71(2):108–113. doi: 10.1212/01.wnl.0000316799.86917.37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Smith EE, Salat DH, Jeng J, McCreary CR, Fischl B, Schmahmann JD, et al. Correlations between MRI white matter lesion location and executive function and episodic memory. Neurology. 2011 Apr 26;76(17):1492–1499. doi: 10.1212/WNL.0b013e318217e7c8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Bocti C, Swartz RH, Gao FQ, Sahlas DJ, Behl P, Black SE. A new visual rating scale to assess strategic white matter hyperintensities within cholinergic pathways in dementia. Stroke. 2005 Oct;36(10):2126–2131. doi: 10.1161/01.STR.0000183615.07936.b6. [DOI] [PubMed] [Google Scholar]
  • 125.Alexopoulos GS, Kiosses DN, Heo M, Murphy CF, Shanmugham B, Gunning-Dixon F. Executive dysfunction and the course of geriatric depression. Biol Psychiatry. 2005;58:204–210. doi: 10.1016/j.biopsych.2005.04.024. [DOI] [PubMed] [Google Scholar]
  • 126.Alexopoulos GS, Meyers BS, Young RC, Kalayam B, Kakuma T, Gabrielle M, et al. Executive dysfunction and long-term outcomes of geriatric depression. Arch Gen Psychiatry. 2000;57:285–290. doi: 10.1001/archpsyc.57.3.285. [DOI] [PubMed] [Google Scholar]
  • 127.Butters MA, Bhalla RK, Mulsant BH, Mazumdar S, Houck PR, Begley AE, et al. Executive functioning, illness course, and relapse/recurrence in continuation and maintenance treatment in late-life depression: Is there a relationship? Am J Geriatr Psychiatry. 2004;12:387–394. doi: 10.1176/appi.ajgp.12.4.387. [DOI] [PubMed] [Google Scholar]
  • 128.Morimoto SS, Wexler BE, Alexopoulos GS. Neuroplasticity-based computerized cognitive remediation for geriatric depression. Int J Geriatr Psychiatry. 2012 Mar 27; doi: 10.1002/gps.3776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.McLennan SN, Mathias JL. The depression-executive dysfunction (DED) syndrome and response to antidepressants: a meta-analytic review. Int J Geriatr Psychiatry. 2010 Oct;25(10):933–944. doi: 10.1002/gps.2431. [DOI] [PubMed] [Google Scholar]
  • 130.Morimoto SS, Gunning FM, Murphy CF, Kanellopoulos D, Kelly RE, Alexopoulos GS. Executive function and short-term remission of geriatric depression: the role of semantic strategy. Am J Geriatr Psychiatry. 2011 Feb;19(2):115–122. doi: 10.1097/JGP.0b013e3181e751c4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Morimoto SS, Gunning FM, Kanellopoulos D, Murphy CF, Klimstra SA, Kelly RE, Jr, et al. Semantic organizational strategy predicts verbal memory and remission rate of geriatric depression. Int J Geriatr Psychiatry. 2012 May;27(5):506–512. doi: 10.1002/gps.2743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Pimontel MA, Culang-Reinlieb ME, Morimoto SS, Sneed JR. Executive dysfunction and treatment response in late-life depression. Int J Geriatr Psychiatry. 2011 Oct 18; doi: 10.1002/gps.2808. [DOI] [PubMed] [Google Scholar]
  • 133.Alexopoulos GS, Hoptman MJ, Kanellopoulos D, Murphy CF, Lim KO, Gunning FM. Functional connectivity in the cognitive control network and the default mode network in late-life depression. J Affect Disord. 2012 Jun;139(1):56–65. doi: 10.1016/j.jad.2011.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Story TJ, Potter GG, Attix DK, Welsh-Bohmer KA, Steffens DC. Neurocognitive correlates of response to treatment in late-life depression. Am J Geriatr Psychiatry. 2008 Sep;16(9):752–759. doi: 10.1097/JGP.0b013e31817e739a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Simpson S, Baldwin RC, Jackson A, Burns AS. Is subcortical disease associated with a poor response to antidepressants? Neurological, neuropsychological and neuroradiological findings in late-life depression. Psychol Med. 1998;28:1015–1026. doi: 10.1017/s003329179800693x. [DOI] [PubMed] [Google Scholar]
  • 136.Patankar TF, Baldwin R, Mitra D, Jeffries S, Sutcliffe C, Burns A, et al. Virchow-Robin space dilatation may predict resistance to antidepressant monotherapy in elderly patients with depression. J Affect Disord. 2007;97:265–270. doi: 10.1016/j.jad.2006.06.024. [DOI] [PubMed] [Google Scholar]
  • 137.Gunning-Dixon FM, Walton M, Cheng J, Acuna J, Klimstra S, Zimmerman ME, et al. MRI signal hyperintensities and treatment remission of geriatric depression. J Affect Disord. 2010 Nov;126(3):395–401. doi: 10.1016/j.jad.2010.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Sneed JR, Culang-Reinlieb ME, Brickman AM, Gunning-Dixon FM, Johnert L, Garcon E, et al. MRI signal hyperintensities and failure to remit following antidepressant treatment. J Affect Disord. 2011 Dec;135(1-3):315–320. doi: 10.1016/j.jad.2011.06.052. [DOI] [PubMed] [Google Scholar]
  • 139.Janssen J, Pol HEH, Schnack HG, Kok RM, Lampe IK, de Leeuw FE, et al. Cerebral volume measurements and subcortical white matter lesions and short-term treatment response in late life depression. Int J Geriatr Psychiatry. 2007;22:468–474. doi: 10.1002/gps.1790. [DOI] [PubMed] [Google Scholar]
  • 140.Krishnan KR, Hays JC, George LK, Blazer DG. Six-month outcomes for MRI-related vascular depression. Depress Anxiety. 1998;8:142–146. doi: 10.1002/(sici)1520-6394(1998)8:4<142::aid-da2>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
  • 141.Salloway S, Boyle PA, Correia S, Malloy PF, Cahn-Weiner DA, Schneider L, et al. The relationship of MRI subcortical hyperintensities to treatment response in a trial of sertraline in geriatric depressed outpatients. Am J Geriatr Psychiatry. 2002;10:107–111. [PubMed] [Google Scholar]
  • 142.Salloway S, Correia S, Boyle P, Malloy P, Schneider L, Lavretsky H, et al. MRI subcortical hyperintensities in old and very old depressed outpatients: the important role of age in late-life depression. J Neurol Sci. 2002;203-204:227–233. doi: 10.1016/s0022-510x(02)00296-4. [DOI] [PubMed] [Google Scholar]
  • 143.Sneed JR, Roose SP, Sackeim HA. Vascular depression: A distinct diagnostic subtype? Biol Psychiatry. 2006 Dec 15;60(12):1295–1298. doi: 10.1016/j.biopsych.2006.06.018. [DOI] [PubMed] [Google Scholar]
  • 144.Sneed JR, Culang-Reinlieb ME. The vascular depression hypothesis: an update. Am J Geriatr Psychiatry. 2011 Feb;19(2):99–103. doi: 10.1097/jgp.0b013e318202fc8a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Raz N, Rodrigue KM, Kennedy KM, Acker JD. Vascular health and longitudinal changes in brain and cognition in middle-aged and older adults. Neuropsychology. 2007;21:149–157. doi: 10.1037/0894-4105.21.2.149. [DOI] [PubMed] [Google Scholar]
  • 146.Taylor WD, Steffens DC, MacFall JR, McQuoid DR, Payne ME, Provenzale JM, et al. White matter hyperintensity progression and late-life depression outcomes. Arch Gen Psychiatry. 2003;60:1090–1096. doi: 10.1001/archpsyc.60.11.1090. [DOI] [PubMed] [Google Scholar]
  • 147.Chen PS, McQuoid DR, Payne ME, Steffens DC. White matter and subcortical gray matter lesion volume changes and late-life depression outcome: a 4-year magnetic resonance imaging study. Int Psychogeriatr. 2006;18:445–456. doi: 10.1017/S1041610205002796. [DOI] [PubMed] [Google Scholar]
  • 148.Alexopoulos GS, Murphy CF, Gunning-Dixon FM, Latoussakis V, Kanellopoulos D, Klimstra S, et al. Microstructural white matter abnormalities and remission of geriatric depression. Am J Psychiatry. 2008;165:238–244. doi: 10.1176/appi.ajp.2007.07050744. [DOI] [PubMed] [Google Scholar]
  • 149.Alexopoulos GS, Glatt CE, Hoptman MJ, Kanellopoulos D, Murphy CF, Kelly RE, Jr, et al. BDNF Val66met polymorphism, white matter abnormalities and remission of geriatric depression. J Affect Disord. 2010 Mar 24; doi: 10.1016/j.jad.2010.02.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Taylor WD, Kuchibhatla M, Payne ME, MacFall JR, Sheline YI, Krishnan KR, et al. Frontal white matter anisotropy and antidepressant response in late-life depression. PLoS ONE. 2008;3:e3267. doi: 10.1371/journal.pone.0003267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Sheline YI, Price JL, Yan Z, Mintun MA. Resting-state functional MRI in depression unmasks increased connectivity between networks via the dorsal nexus. Proc Natl Acad Sci USA. 2010 Jun 15;107(24):11020–11025. doi: 10.1073/pnas.1000446107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Alexopoulos GS, Murphy CF, Gunning-Dixon FM, Glatt CE, Latoussakis V, Kelly RE, Jr, et al. Serotonin transporter polymorphisms, microstructural white matter abnormalities and remission of geriatric depression. J Affect Disord. 2009 Apr 15; doi: 10.1016/j.jad.2009.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Lotrich FE. Gene-environment interactions in geriatric depression. Psychiatr Clin North Am. 2011 Jun;34(2):357–376. viii. doi: 10.1016/j.psc.2011.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Benjamin S, Taylor W. Nature and nurture: genetic influences and gene-environment interactions in depression. Curr Psychiatry Rev. 2010;6:82–90. [Google Scholar]
  • 155.Annerbrink K, Jonsson EG, Olsson M, Nilsson S, Sedvall GC, Anckarsater H, et al. Associations between the angiotensin-converting enzyme insertion/deletion polymorphism and monoamine metabolite concentrations in cerebrospinal fluid. Psychiatry Res. 2010 Sep 30;179(2):231–234. doi: 10.1016/j.psychres.2009.04.018. [DOI] [PubMed] [Google Scholar]
  • 156.Baghai TC, Schule C, Zwanzger P, Minov C, Zill P, Ella R, et al. Hypothalamic-pituitary-adrenocortical axis dysregulation in patients with major depression is influenced by the insertion/deletion polymorphism in the angiotensin I-converting enzyme gene. Neurosci Lett. 2002 Aug 16;328(3):299–303. doi: 10.1016/s0304-3940(02)00527-x. [DOI] [PubMed] [Google Scholar]
  • 157.Wu Y, Wang X, Shen X, Tan Z, Yuan Y. The I/D polymorphism of angiotensin-converting enzyme gene in major depressive disorder and therapeutic outcome: a case-control study and meta-analysis. J Affect Disord. 2012 Feb;136(3):971–978. doi: 10.1016/j.jad.2011.08.019. [DOI] [PubMed] [Google Scholar]
  • 158.Arinami T, Li L, Mitsushio H, Itokawa M, Hamaguchi H, Toru M. An insertion/deletion polymorphism in the angiotensin converting enzyme gene is associated with both brain substance P contents and affective disorders. Biol Psychiatry. 1996 Dec 1;40(11):1122–1127. doi: 10.1016/s0006-3223(95)00597-8. [DOI] [PubMed] [Google Scholar]
  • 159.Saab YB, Gard PR, Yeoman MS, Mfarrej B, El-Moalem H, Ingram MJ. Reninangiotensin-system gene polymorphisms and depression. Prog Neuropsychopharmacol Biol Psychiatry. 2007 Jun 30;31(5):1113–1118. doi: 10.1016/j.pnpbp.2007.04.002. [DOI] [PubMed] [Google Scholar]
  • 160.Lopez-Leon S, Janssens AC, Hofman A, Claes S, Breteler MM, Tiemeier H, et al. No association between the angiotensin-converting enzyme gene and major depression: a case-control study and meta-analysis. Psychiatr Genet. 2006 Dec;16(6):225–226. doi: 10.1097/01.ypg.0000242191.51397.3d. [DOI] [PubMed] [Google Scholar]
  • 161.Baghai TC, Binder EB, Schule C, Salyakina D, Eser D, Lucae S, et al. Polymorphisms in the angiotensin-converting enzyme gene are associated with unipolar depression, ACE activity and hypercortisolism. Mol Psychiatry. 2006;11:1003–1015. doi: 10.1038/sj.mp.4001884. [DOI] [PubMed] [Google Scholar]
  • 162.Sparks DL, Hunsaker JC, 3rd, Amouyel P, Malafosse A, Bellivier F, Leboyer M, et al. Angiotensin I-converting enzyme I/D polymorphism and suicidal behaviors. Am J Med Genet B Neuropsychiatr Genet. 2009 Mar 5;150B(2):290–294. doi: 10.1002/ajmg.b.30793. [DOI] [PubMed] [Google Scholar]
  • 163.Hishimoto A, Shirakawa O, Nishiguchi N, Hashimoto T, Yanagi M, Nushida H, et al. Association between a functional polymorphism in the renin-angiotensin system and completed suicide. J Neural Transm. 2006 Dec;113(12):1915–1920. doi: 10.1007/s00702-006-0483-9. [DOI] [PubMed] [Google Scholar]
  • 164.Fudalej S, Fudalej M, Kostrzewa G, Kuzniar P, Franaszczyk M, Wojnar M, et al. Angiotensin-converting enzyme polymorphism and completed suicide: an association in Caucasians and evidence for a link with a method of self-injury. Neuropsychobiology. 2009;59(3):151–158. doi: 10.1159/000218077. [DOI] [PubMed] [Google Scholar]
  • 165.Angunsri R, Sritharathikhun T, Suttirat S, Tencomnao T. Association of angiotensin-converting enzyme gene promoter single nucleotide polymorphisms and haplotype with major depression in a northeastern Thai population. J Renin Angiotensin Aldosterone Syst. 2009 Sep;10(3):179–184. doi: 10.1177/1470320309344151. [DOI] [PubMed] [Google Scholar]
  • 166.Firouzabadi N, Shafiei M, Bahramali E, Ebrahimi SA, Bakhshandeh H, Tajik N. Association of angiotensin-converting enzyme (ACE) gene polymorphism with elevated serum ACE activity and major depression in an Iranian population. Psychiatry Res. 2012 Dec 30;200(2-3):336–342. doi: 10.1016/j.psychres.2012.05.002. [DOI] [PubMed] [Google Scholar]
  • 167.Spiering W, Kroon AA, Fuss-Lejeune MM, Daemen MJ, de Leeuw PW. Angiotensin II sensitivity is associated with the angiotensin II type 1 receptor A(1166)C polymorphism in essential hypertensives on a high sodium diet. Hypertension. 2000 Sep;36(3):411–416. doi: 10.1161/01.hyp.36.3.411. [DOI] [PubMed] [Google Scholar]
  • 168.Taylor WD, Steffens DC, Ashley-Koch A, Payne ME, MacFall JR, Potocky C, et al. Angiotensin receptor gene polymorphisms and 2-year change in cerebral hyperintense lesion volume in men. Mol Psychiatry. 2010;15:816–822. doi: 10.1038/mp.2009.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Sleegers K, den Heijer T, van Dijk EJ, Hofman A, Bertoli-Avella AM, Koudstaal PJ, et al. ACE gene is associated with Alzheimer's disease and atrophy of hippocampus and amygdala. Neurobiol Aging. 2005 Aug-Sep;26(8):1153–1159. doi: 10.1016/j.neurobiolaging.2004.09.011. [DOI] [PubMed] [Google Scholar]
  • 170.Zhang Z, Deng L, Bai F, Shi Y, Yu H, Yuan Y, et al. ACE I/D polymorphism affects cognitive function and gray-matter volume in amnestic mild cognitive impairment. Behav Brain Res. 2011 Mar 17;218(1):114–120. doi: 10.1016/j.bbr.2010.11.032. [DOI] [PubMed] [Google Scholar]
  • 171.Taylor WD, Benjamin S, McQuoid DR, Payne ME, Krishnan RR, MacFall JR, et al. AGTR1 gene variation: association with depression and frontotemporal morphology. Psychiatry Res. 2012 May 31;202(2):104–109. doi: 10.1016/j.pscychresns.2012.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Hou Z, Yuan Y, Zhang Z, Hou G, You J, Bai F. The D-allele of ACE insertion/deletion polymorphism is associated with regional white matter volume changes and cognitive impairment in remitted geriatric depression. Neurosci Lett. 2010 Aug 2;479(3):262–266. doi: 10.1016/j.neulet.2010.05.076. [DOI] [PubMed] [Google Scholar]
  • 173.Wang Z, Yuan Y, Bai F, You J, Li L, Zhang Z. Abnormal default-mode network in angiotensin converting enzyme D allele carriers with remitted geriatric depression. Behav Brain Res. 2012 May 1;230(2):325–332. doi: 10.1016/j.bbr.2012.02.011. [DOI] [PubMed] [Google Scholar]
  • 174.Baghai TC, Schule C, Zill P, Deiml T, Eser D, Zwanzger P, et al. The angiotensin I converting enzyme insertion/deletion polymorphism influences therapeutic outcome in major depressed women, but not in men. Neurosci Lett. 2004 Jun 3;363(1):38–42. doi: 10.1016/j.neulet.2004.03.052. [DOI] [PubMed] [Google Scholar]
  • 175.Bondy B, Baghai TC, Zill P, Schule C, Eser D, Deiml T, et al. Genetic variants in the angiotensin I-converting-enzyme (ACE) and angiotensin II receptor (AT1) gene and clinical outcome in depression. Prog Neuropsychopharmacol Biol Psychiatry. 2005 Jul;29(6):1094–1099. doi: 10.1016/j.pnpbp.2005.03.015. [DOI] [PubMed] [Google Scholar]
  • 176.Kondo DG, Speer MC, Krishnan KR, McQuoid DR, Slifer SH, Pieper CF, et al. Association of AGTR1 with 18-month treatment outcome in late-life depression. Am J Geriatr Psychiatry. 2007;15:564–572. doi: 10.1097/JGP.0b013e31805470a4. [DOI] [PubMed] [Google Scholar]
  • 177.Taylor WD, Bae JN, MacFall JR, Payne ME, Provenzale JM, Steffens DC, et al. Widespread effects of hyperintense lesions on cerebral white matter structure. Am J Roentgenol. 2007;188:1695–1704. doi: 10.2214/AJR.06.1163. [DOI] [PubMed] [Google Scholar]
  • 178.Alexopoulos GS. Frontostriatal and limbic dysfunction in late-life depression. Am J Geriatr Psychiatry. 2002 Nov-Dec;10(6):687–695. [PubMed] [Google Scholar]
  • 179.Dalby RB, Frandsen J, Chakravarty MM, Ahdidan J, Sorensen L, Rosenberg R, et al. Depression severity is correlated to the integrity of white matter fiber tracts in late-onset major depression. Psychiatry Res. 2010 Oct 30;184(1):38–48. doi: 10.1016/j.pscychresns.2010.06.008. [DOI] [PubMed] [Google Scholar]
  • 180.Taylor WD, Payne ME, Krishnan KRR, Wagner HR, Provenzale JM, Steffens DC, et al. Evidence of white matter tract disruption in MRI hyperintensities. Biol Psychiatry. 2001;50:179–183. doi: 10.1016/s0006-3223(01)01160-x. [DOI] [PubMed] [Google Scholar]
  • 181.Sexton CE, Le Masurier M, Allan CL, Jenkinson M, McDermott L, Kalu UG, et al. Magnetic resonance imaging in late-life depression: vascular and glucocorticoid cascade hypotheses. Br J Psychiatry. 2012 Jul;201:46–51. doi: 10.1192/bjp.bp.111.105361. [DOI] [PubMed] [Google Scholar]
  • 182.Taylor WD, MacFall JR, Gerig G, Krishnan KR. Structural integrity of the uncinate fasciculus in geriatric depression: Relationship with age of onset. Neuropsychiatr Dis Treat. 2007;3:669–674. [PMC free article] [PubMed] [Google Scholar]
  • 183.Ebeling U, Cramon DV. Topography of the uncinate fascicle and adjacent temporal fiber tracts. Acta Neurochir (Wien) 1992;115:143–148. doi: 10.1007/BF01406373. [DOI] [PubMed] [Google Scholar]
  • 184.Kier EL, Staib LH, Davis LM, Bronen RA. MR imaging of the temporal stem: anatomic dissection tractography of the uncinate fasciculus, inferior occipitofrontal fasciculus, and Meyer's loop of the optic radiation. AJNR Am J Neuroradiol. 2004 May;25(5):677–691. [PMC free article] [PubMed] [Google Scholar]
  • 185.Petrides M, Pandya DN. Efferent association pathways from the rostral prefrontal cortex in the macaque monkey. J Neurosci. 2007 Oct 24;27(43):11573–11586. doi: 10.1523/JNEUROSCI.2419-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Mufson EJ, Pandya DN. Some observations on the course and composition of the cingulum bundle in the rhesus monkey. J Comp Neurol. 1984 May 1;225(1):31–43. doi: 10.1002/cne.902250105. [DOI] [PubMed] [Google Scholar]
  • 187.Morris R, Pandya DN, Petrides M. Fiber system linking the mid-dorsolateral frontal cortex with the retrosplenial/presubicular region in the rhesus monkey. J Comp Neurol. 1999 May 3;407(2):183–192. doi: 10.1002/(sici)1096-9861(19990503)407:2<183::aid-cne3>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
  • 188.Rudrauf D, Mehta S, Grabowski TJ. Disconnection's renaissance takes shape: Formal incorporation in group-level lesion studies. Cortex. 2008 Sep;44(8):1084–1096. doi: 10.1016/j.cortex.2008.05.005. [DOI] [PubMed] [Google Scholar]
  • 189.Gaffan D, Wilson CR. Medial temporal and prefrontal function: recent behavioural disconnection studies in the macaque monkey. Cortex. 2008 Sep;44(8):928–935. doi: 10.1016/j.cortex.2008.03.005. [DOI] [PubMed] [Google Scholar]
  • 190.Zhang A, Leow A, Ajilore O, Lamar M, Yang S, Joseph J, et al. Quantitative tract-specific measures of uncinate and cingulum in major depression using diffusion tensor imaging. Neuropsychopharmacology. 2012 Mar;37(4):959–967. doi: 10.1038/npp.2011.279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Keedwell PA, Chapman R, Christiansen K, Richardson H, Evans J, Jones DK. Cingulum White Matter in Young Women at Risk of Depression: The Effect of Family History and Anhedonia. Biol Psychiatry. 2012 Feb 29; doi: 10.1016/j.biopsych.2012.01.022. [DOI] [PubMed] [Google Scholar]
  • 192.Damoiseaux JS, Greicius MD. Greater than the sum of its parts: a review of studies combining structural connectivity and resting-state functional connectivity. Brain Struct Funct. 2009 Oct;213(6):525–533. doi: 10.1007/s00429-009-0208-6. [DOI] [PubMed] [Google Scholar]
  • 193.Skudlarski P, Jagannathan K, Calhoun VD, Hampson M, Skudlarska BA, Pearlson G. Measuring brain connectivity: diffusion tensor imaging validates resting state temporal correlations. Neuroimage. 2008 Nov 15;43(3):554–561. doi: 10.1016/j.neuroimage.2008.07.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Teipel SJ, Bokde AL, Meindl T, Amaro E, Jr., Soldner J, Reiser MF, et al. White matter microstructure underlying default mode network connectivity in the human brain. Neuroimage. 2010 Feb 1;49(3):2021–2032. doi: 10.1016/j.neuroimage.2009.10.067. [DOI] [PubMed] [Google Scholar]
  • 195.van den Heuvel M, Mandl R, Luigjes J, Hulshoff Pol H. Microstructural organization of the cingulum tract and the level of default mode functional connectivity. J Neurosci. 2008 Oct 22;28(43):10844–10851. doi: 10.1523/JNEUROSCI.2964-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Steffens DC, Taylor WD, Denny KL, Bergman SR, Wang L. Structural integrity of the uncinate fasciculus and resting state functional connectivity of the ventral prefrontal cortex in late life depression. PLoS ONE. 2011;6(7):e22697. doi: 10.1371/journal.pone.0022697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Godbout JP, Johnson RW. Age and neuroinflammation: a lifetime of psychoneuroimmune consequences. Neurologic clinics. 2006 Aug;24(3):521–538. doi: 10.1016/j.ncl.2006.03.010. [DOI] [PubMed] [Google Scholar]
  • 198.Dilger RN, Johnson RW. Aging, microglial cell priming, and the discordant central inflammatory response to signals from the peripheral immune system. J Leukoc Biol. 2008 Oct;84(4):932–939. doi: 10.1189/jlb.0208108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Maes M. Depression is an inflammatory disease, but cell-mediated immune activation is the key component of depression. Prog Neuropsychopharmacol Biol Psychiatry. 2011 Apr 29;35(3):664–675. doi: 10.1016/j.pnpbp.2010.06.014. [DOI] [PubMed] [Google Scholar]
  • 200.Raison CL, Demetrashvili M, Capuron L, Miller AH. Neuropsychiatric adverse effects of interferon-alpha: recognition and management. CNS Drugs. 2005;19(2):105–123. doi: 10.2165/00023210-200519020-00002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Alexopoulos GS, Morimoto SS. The inflammation hypothesis in geriatric depression. Int J Geriatr Psychiatry. 2011 Mar 2; doi: 10.1002/gps.2672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Brydon L, Harrison NA, Walker C, Steptoe A, Critchley HD. Peripheral inflammation is associated with altered substantia nigra activity and psychomotor slowing in humans. Biol Psychiatry. 2008 Jun 1;63(11):1022–1029. doi: 10.1016/j.biopsych.2007.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Harrison NA, Brydon L, Walker C, Gray MA, Steptoe A, Critchley HD. Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biol Psychiatry. 2009 Sep 1;66(5):407–414. doi: 10.1016/j.biopsych.2009.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Raison CL, Capuron L, Miller AH. Cytokines sing the blues: inflammation and the pathogenesis of depression. Trends Immunol. 2006 Jan;27(1):24–31. doi: 10.1016/j.it.2005.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Zorrilla EP, Luborsky L, McKay JR, Rosenthal R, Houldin A, Tax A, et al. The relationship of depression and stressors to immunological assays: a meta-analytic review. Brain Behav Immun. 2001 Sep;15(3):199–226. doi: 10.1006/brbi.2000.0597. [DOI] [PubMed] [Google Scholar]
  • 206.Sutcigil L, Oktenli C, Musabak U, Bozkurt A, Cansever A, Uzun O, et al. Pro- and anti-inflammatory cytokine balance in major depression: effect of sertraline therapy. Clin Dev Immunol. 2007;2007:76396. doi: 10.1155/2007/76396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Kim YK, Na KS, Shin KH, Jung HY, Choi SH, Kim JB. Cytokine imbalance in the pathophysiology of major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2007 Jun 30;31(5):1044–1053. doi: 10.1016/j.pnpbp.2007.03.004. [DOI] [PubMed] [Google Scholar]
  • 208.Miller AH. Norman Cousins Lecture. Mechanisms of cytokine-induced behavioral changes: psychoneuroimmunology at the translational interface. Brain Behav Immun. 2009 Feb;23(2):149–158. doi: 10.1016/j.bbi.2008.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.O'Connor JC, Andre C, Wang Y, Lawson MA, Szegedi SS, Lestage J, et al. Interferon-gamma and tumor necrosis factor-alpha mediate the upregulation of indoleamine 2,3-dioxygenase and the induction of depressive-like behavior in mice in response to bacillus Calmette-Guerin. J Neurosci. 2009 Apr 1;29(13):4200–4209. doi: 10.1523/JNEUROSCI.5032-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.O'Connor JC, Lawson MA, Andre C, Moreau M, Lestage J, Castanon N, et al. Lipopolysaccharide-induced depressive-like behavior is mediated by indoleamine 2,3-dioxygenase activation in mice. Mol Psychiatry. 2009 May;14(5):511–522. doi: 10.1038/sj.mp.4002148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R. The new ‘5-HT’ hypothesis of depression: Cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry. 2010 Dec 23; doi: 10.1016/j.pnpbp.2010.12.017. [DOI] [PubMed] [Google Scholar]
  • 212.Stone TW, Behan WM. Interleukin-1beta but not tumor necrosis factor-alpha potentiates neuronal damage by quinolinic acid: protection by an adenosine A2A receptor antagonist. J Neurosci Res. 2007 Apr;85(5):1077–1085. doi: 10.1002/jnr.21212. [DOI] [PubMed] [Google Scholar]
  • 213.Tsao CW, Lin YS, Chen CC, Bai CH, Wu SR. Cytokines and serotonin transporter in patients with major depression. Prog Neuropsychopharmacol Biol Psychiatry. 2006 Jul;30(5):899–905. doi: 10.1016/j.pnpbp.2006.01.029. [DOI] [PubMed] [Google Scholar]
  • 214.Pace TW, Hu F, Miller AH. Cytokine-effects on glucocorticoid receptor function: relevance to glucocorticoid resistance and the pathophysiology and treatment of major depression. Brain Behav Immun. 2007 Jan;21(1):9–19. doi: 10.1016/j.bbi.2006.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Koo JW, Duman RS. IL-1beta is an essential mediator of the antineurogenic and anhedonic effects of stress. Proc Natl Acad Sci USA. 2008 Jan 15;105(2):751–756. doi: 10.1073/pnas.0708092105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Slavich GM, Way BM, Eisenberger NI, Taylor SE. Neural sensitivity to social rejection is associated with inflammatory responses to social stress. Proc Natl Acad Sci U S A. 2010 Aug 17;107(33):14817–14822. doi: 10.1073/pnas.1009164107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.O'Connor MF, Irwin MR, Wellisch DK. When grief heats up: pro-inflammatory cytokines predict regional brain activation. Neuroimage. 2009 Sep;47(3):891–896. doi: 10.1016/j.neuroimage.2009.05.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Wong ML, Dong C, Maestre-Mesa J, Licinio J. Polymorphisms in inflammation-related genes are associated with susceptibility to major depression and antidepressant response. Mol Psychiatry. 2008 Aug;13(8):800–812. doi: 10.1038/mp.2008.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Cerri AP, Arosio B, Viazzoli C, Confalonieri R, Vergani C, Annoni G. The -308 (G/A) single nucleotide polymorphism in the TNF-alpha gene and the risk of major depression in the elderly. Int J Geriatr Psychiatry. 2010 Mar;25(3):219–223. doi: 10.1002/gps.2323. [DOI] [PubMed] [Google Scholar]
  • 220.Hwang JP, Tsai SJ, Hong CJ, Yang CH, Hsu CD, Liou YJ. Interleukin-1 beta -511C/T genetic polymorphism is associated with age of onset of geriatric depression. Neuromolecular Med. 2009;11(4):322–327. doi: 10.1007/s12017-009-8078-x. [DOI] [PubMed] [Google Scholar]
  • 221.Halder I, Marsland AL, Cheong J, Muldoon MF, Ferrell RE, Manuck SB. Polymorphisms in the CRP gene moderate an association between depressive symptoms and circulating levels of C-reactive protein. Brain Behav Immun. 2010 Jan;24(1):160–167. doi: 10.1016/j.bbi.2009.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Baune BT, Dannlowski U, Domschke K, Janssen DG, Jordan MA, Ohrmann P, et al. The interleukin 1 beta (IL1B) gene is associated with failure to achieve remission and impaired emotion processing in major depression. Biol Psychiatry. 2010 Mar 15;67(6):543–549. doi: 10.1016/j.biopsych.2009.11.004. [DOI] [PubMed] [Google Scholar]
  • 223.Gruver AL, Hudson LL, Sempowski GD. Immunosenescence of ageing. J Pathol. 2007 Jan;211(2):144–156. doi: 10.1002/path.2104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Penninx BW, Kritchevsky SB, Yaffe K, Newman AB, Simonsick EM, Rubin S, et al. Inflammatory markers and depressed mood in older persons: results from the Health, Aging and Body Composition study. Biol Psychiatry. 2003 Sep 1;54(5):566–572. doi: 10.1016/s0006-3223(02)01811-5. [DOI] [PubMed] [Google Scholar]
  • 225.Dentino AN, Pieper CF, Rao MK, Currie MS, Harris T, Blazer DG, et al. Association of interleukin-6 and other biologic variables with depression in older people living in the community. J Am Geriatr Soc. 1999 Jan;47(1):6–11. doi: 10.1111/j.1532-5415.1999.tb01894.x. [DOI] [PubMed] [Google Scholar]
  • 226.Tiemeier H, Hofman A, van Tuijl HR, Kiliaan AJ, Meijer J, Breteler MM. Inflammatory proteins and depression in the elderly. Epidemiology. 2003 Jan;14(1):103–107. doi: 10.1097/00001648-200301000-00025. [DOI] [PubMed] [Google Scholar]
  • 227.Bremmer MA, Beekman AT, Deeg DJ, Penninx BW, Dik MG, Hack CE, et al. Inflammatory markers in late-life depression: results from a population-based study. J Affect Disord. 2008 Mar;106(3):249–255. doi: 10.1016/j.jad.2007.07.002. [DOI] [PubMed] [Google Scholar]
  • 228.Baune BT, Smith E, Reppermund S, Air T, Samaras K, Lux O, et al. Inflammatory biomarkers predict depressive, but not anxiety symptoms during aging: The prospective Sydney Memory and Aging Study. Psychoneuroendocrinology. 2012 Sep;37(9):1521–1530. doi: 10.1016/j.psyneuen.2012.02.006. [DOI] [PubMed] [Google Scholar]
  • 229.Elderkin-Thompson V, Irwin MR, Hellemann G, Kumar A. Interleukin-6 and Memory Functions of Encoding and Recall in Healthy and Depressed Elderly Adults. Am J Geriatr Psychiatry. 2012 Aug 13; doi: 10.1097/JGP.0b013e31825d08d6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Schram MT, Euser SM, de Craen AJ, Witteman JC, Frolich M, Hofman A, et al. Systemic markers of inflammation and cognitive decline in old age. J Am Geriatr Soc. 2007 May;55(5):708–716. doi: 10.1111/j.1532-5415.2007.01159.x. [DOI] [PubMed] [Google Scholar]
  • 231.Marioni RE, Strachan MW, Reynolds RM, Lowe GD, Mitchell RJ, Fowkes FG, et al. Association between raised inflammatory markers and cognitive decline in elderly people with type 2 diabetes: the Edinburgh Type 2 Diabetes Study. Diabetes. 2010 Mar;59(3):710–713. doi: 10.2337/db09-1163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Fornage M, Chiang YA, O'Meara ES, Psaty BM, Reiner AP, Siscovick DS, et al. Biomarkers of Inflammation and MRI-Defined Small Vessel Disease of the Brain: The Cardiovascular Health Study. Stroke. 2008 Jul;39(7):1952–1959. doi: 10.1161/STROKEAHA.107.508135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.van Dijk EJ, Prins ND, Vermeer SE, Vrooman HA, Hofman A, Koudstaal PJ, et al. C-reactive protein and cerebral small-vessel disease: the Rotterdam Scan Study. Circulation. 2005 Aug 9;112(6):900–905. doi: 10.1161/CIRCULATIONAHA.104.506337. [DOI] [PubMed] [Google Scholar]
  • 234.Raz N, Yang Y, Dahle CL, Land S. Volume of white matter hyperintensities in healthy adults: contribution of age, vascular risk factors, and inflammation-related genetic variants. Biochim Biophys Acta. 2012 Mar;1822(3):361–369. doi: 10.1016/j.bbadis.2011.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Satizabal CL, Zhu YC, Mazoyer B, Dufouil C, Tzourio C. Circulating IL-6 and CRP are associated with MRI findings in the elderly: the 3C-Dijon Study. Neurology. 2012 Mar 6;78(10):720–727. doi: 10.1212/WNL.0b013e318248e50f. [DOI] [PubMed] [Google Scholar]
  • 236.Janssen DG, Caniato RN, Verster JC, Baune BT. A psychoneuroimmunological review on cytokines involved in antidepressant treatment response. Hum Psychopharmacol. 2010 Apr;25(3):201–215. doi: 10.1002/hup.1103. [DOI] [PubMed] [Google Scholar]
  • 237.Xia Z, DePierre JW, Nassberger L. Tricyclic antidepressants inhibit IL-6, IL-1 beta and TNF-alpha release in human blood monocytes and IL-2 and interferon-gamma in T cells. Immunopharmacology. 1996 Aug;34(1):27–37. doi: 10.1016/0162-3109(96)00111-7. [DOI] [PubMed] [Google Scholar]
  • 238.Maes M, Song C, Lin AH, Bonaccorso S, Kenis G, De Jongh R, et al. Negative immunoregulatory effects of antidepressants: inhibition of interferon-gamma and stimulation of interleukin-10 secretion. Neuropsychopharmacology. 1999 Apr;20(4):370–379. doi: 10.1016/S0893-133X(98)00088-8. [DOI] [PubMed] [Google Scholar]
  • 239.Szuster-Ciesielska A, Tustanowska-Stachura A, Slotwinska M, Marmurowska-Michalowska H, Kandefer-Szerszen M. In vitro immunoregulatory effects of antidepressants in healthy volunteers. Pol J Pharmacol. 2003 May-Jun;55(3):353–362. [PubMed] [Google Scholar]
  • 240.Kenis G, Maes M. Effects of antidepressants on the production of cytokines. Int J Neuropsychopharmacol. 2002 Dec;5(4):401–412. doi: 10.1017/S1461145702003164. [DOI] [PubMed] [Google Scholar]
  • 241.Hannestad J, DellaGioia N, Bloch M. The effect of antidepressant medication treatment on serum levels of inflammatory cytokines: a meta-analysis. Neuropsychopharmacology. 2011 Nov;36(12):2452–2459. doi: 10.1038/npp.2011.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Tyring S, Gottlieb A, Papp K, Gordon K, Leonardi C, Wang A, et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: double-blind placebo-controlled randomised phase III trial. Lancet. 2006 Jan 7;367(9504):29–35. doi: 10.1016/S0140-6736(05)67763-X. [DOI] [PubMed] [Google Scholar]
  • 243.Kekow J, Moots RJ, Emery P, Durez P, Koenig A, Singh A, et al. Patient-reported outcomes improve with etanercept plus methotrexate in active early rheumatoid arthritis and the improvement is strongly associated with remission: the COMET trial. Ann Rheum Dis. 2010 Jan;69(1):222–225. doi: 10.1136/ard.2008.102509. [DOI] [PubMed] [Google Scholar]
  • 244.Muller N, Schwarz MJ, Dehning S, Douhe A, Cerovecki A, Goldstein-Muller B, et al. The cyclooxygenase-2 inhibitor celecoxib has therapeutic effects in major depression: results of a double-blind, randomized, placebo controlled, add-on pilot study to reboxetine. Mol Psychiatry. 2006 Jul;11(7):680–684. doi: 10.1038/sj.mp.4001805. [DOI] [PubMed] [Google Scholar]
  • 245.Raison CL, Rutherford RE, Woolwine BJ, Shuo C, Schettler P, Drake DF, et al. A Randomized Controlled Trial of the Tumor Necrosis Factor Antagonist Infliximab for Treatment-Resistant Depression: The Role of Baseline Inflammatory Biomarkers. Arch Gen Psychiatry. 2012 Sep;3:1–11. doi: 10.1001/2013.jamapsychiatry.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Koyama A, O'Brien J, Weuve J, Blacker D, Metti AL, Yaffe K. The Role of Peripheral Inflammatory Markers in Dementia and Alzheimer's Disease: A Meta-Analysis. J Gerontol A Biol Sci Med Sci. 2012 Sep 14; doi: 10.1093/gerona/gls187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Rubio-Perez JM, Morillas-Ruiz JM. A review: inflammatory process in Alzheimer's disease, role of cytokines. TheScientificWorldJournal. 2012;2012:756357. doi: 10.1100/2012/756357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Corbett A, Pickett J, Burns A, Corcoran J, Dunnett SB, Edison P, et al. Drug repositioning for Alzheimer's disease. Nat Rev Drug Discov. 2012 Nov;11(11):833–846. doi: 10.1038/nrd3869. [DOI] [PubMed] [Google Scholar]
  • 249.Paranthaman R, Greenstein AS, Burns AS, Cruickshank JK, Heagerty AM, Jackson A, et al. Vascular function in older adults with depressive disorder. Biol Psychiatry. 2010 Jul 15;68(2):133–139. doi: 10.1016/j.biopsych.2010.04.017. [DOI] [PubMed] [Google Scholar]
  • 250.Greenstein AS, Paranthaman R, Burns A, Jackson A, Malik RA, Baldwin RC, et al. Cerebrovascular damage in late-life depression is associated with structural and functional abnormalities of subcutaneous small arteries. Hypertension. 2010 Oct;56(4):734–740. doi: 10.1161/HYPERTENSIONAHA.110.152801. [DOI] [PubMed] [Google Scholar]
  • 251.Broadley AJ, Korszun A, Jones CJ, Frenneaux MP. Arterial endothelial function is impaired in treated depression. Heart. 2002 Nov;88(5):521–523. doi: 10.1136/heart.88.5.521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Rajagopalan S, Brook R, Rubenfire M, Pitt E, Young E, Pitt B. Abnormal brachial artery flow-mediated vasodilation in young adults with major depression. Am J Cardiol. 2001 Jul 15;88(2):196–198. A197. doi: 10.1016/s0002-9149(01)01623-x. [DOI] [PubMed] [Google Scholar]
  • 253.Tiemeier H, Breteler MM, van Popele NM, Hofman A, Witteman JC. Late-life depression is associated with arterial stiffness: a population-based study. J Am Geriatr Soc. 2003 Aug;51(8):1105–1110. doi: 10.1046/j.1532-5415.2003.51359.x. [DOI] [PubMed] [Google Scholar]
  • 254.Chen CS, Chen CC, Kuo YT, Chiang IC, Ko CH, Lin HF. Carotid intima-media thickness in late-onset major depressive disorder. Int J Geriatr Psychiatry. 2006 Jan;21(1):36–42. doi: 10.1002/gps.1420. [DOI] [PubMed] [Google Scholar]
  • 255.Iadecola C. Neurovascular regulation in the normal brain and in Alzheimer's disease. Nat Rev Neurosci. 2004 May;5(5):347–360. doi: 10.1038/nrn1387. [DOI] [PubMed] [Google Scholar]
  • 256.de la Torre JC. Cerebral Hemodynamics and Vascular Risk Factors: Setting the Stage for Alzheimer's Disease. J Alzheimers Dis. 2012 Jul 27; doi: 10.3233/JAD-2012-120793. [DOI] [PubMed] [Google Scholar]
  • 257.Touyz RM. Intracellular mechanisms involved in vascular remodelling of resistance arteries in hypertension: role of angiotensin II. Exp Physiol. 2005 Jul;90(4):449–455. doi: 10.1113/expphysiol.2005.030080. [DOI] [PubMed] [Google Scholar]
  • 258.Dandona P, Chaudhuri A, Aljada A. Endothelial dysfunction and hypertension in diabetes mellitus. Med Clin North Am. 2004 Jul;88(4):911–931. x–xi. doi: 10.1016/j.mcna.2004.04.006. [DOI] [PubMed] [Google Scholar]
  • 259.Paranthaman R, Greenstein A, Burns AS, Heagerty AM, Malik RA, Baldwin RC. Relationship of endothelial function and atherosclerosis to treatment response in late-life depression. Int J Geriatr Psychiatry. 2012 Sep;27(9):967–973. doi: 10.1002/gps.2811. [DOI] [PubMed] [Google Scholar]
  • 260.Tiemeier H, Bakker SL, Hofman A, Koudstaal PJ, Breteler MM. Cerebral haemodynamics and depression in the elderly. J Neurol Neurosurg Psychiatry. 2002 Jul;73(1):34–39. doi: 10.1136/jnnp.73.1.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Direk N, Koudstaal PJ, Hofman A, Ikram MA, Hoogendijk WJ, Tiemeier H. Cerebral hemodynamics and incident depression: the rotterdam study. Biol Psychiatry. 2012 Aug 15;72(4):318–323. doi: 10.1016/j.biopsych.2012.01.019. [DOI] [PubMed] [Google Scholar]
  • 262.Mies G, Ishimaru S, Xie Y, Seo K, Hossmann KA. Ischemic thresholds of cerebral protein synthesis and energy state following middle cerebral artery occlusion in rat. J Cereb Blood Flow Metab. 1991 Sep;11(5):753–761. doi: 10.1038/jcbfm.1991.132. [DOI] [PubMed] [Google Scholar]
  • 263.Martin KC, Barad M, Kandel ER. Local protein synthesis and its role in synapse-specific plasticity. Curr Opin Neurobiol. 2000 Oct;10(5):587–592. doi: 10.1016/s0959-4388(00)00128-8. [DOI] [PubMed] [Google Scholar]
  • 264.Debiec J, LeDoux JE, Nader K. Cellular and systems reconsolidation in the hippocampus. Neuron. 2002 Oct 24;36(3):527–538. doi: 10.1016/s0896-6273(02)01001-2. [DOI] [PubMed] [Google Scholar]
  • 265.Kleim JA, Bruneau R, Calder K, Pocock D, VandenBerg PM, MacDonald E, et al. Functional organization of adult motor cortex is dependent upon continued protein synthesis. Neuron. 2003 Sep 25;40(1):167–176. doi: 10.1016/s0896-6273(03)00592-0. [DOI] [PubMed] [Google Scholar]
  • 266.Moody DM, Bell MA, Challa VR. Features of the cerebral vascular pattern that predict vulnerability to perfusion or oxygenation deficiency: an anatomic study. AJNR Am J Neuroradiol. 1990 May;11(3):431–439. [PMC free article] [PubMed] [Google Scholar]
  • 267.Matsushita K, Kuriyama Y, Nagatsuka K, Nakamura M, Sawada T, Omae T. Periventricular white matter lucency and cerebral blood flow autoregulation in hypertensive patients. Hypertension. 1994 May;23(5):565–568. doi: 10.1161/01.hyp.23.5.565. [DOI] [PubMed] [Google Scholar]
  • 268.Markus HS, Lythgoe DJ, Ostegaard L, O'Sullivan M, Williams SC. Reduced cerebral blood flow in white matter in ischaemic leukoaraiosis demonstrated using quantitative exogenous contrast based perfusion MRI. J Neurol Neurosurg Psychiatry. 2000 Jul;69(1):48–53. doi: 10.1136/jnnp.69.1.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Oda K, Okubo Y, Ishida R, Murata Y, Ohta K, Matsuda T, et al. Regional cerebral blood flow in depressed patients with white matter magnetic resonance hyperintensity. Biol Psychiatry. 2003;53:150–156. doi: 10.1016/s0006-3223(02)01548-2. [DOI] [PubMed] [Google Scholar]
  • 270.Brickman AM, Zahra A, Muraskin J, Steffener J, Holland CM, Habeck C, et al. Reduction in cerebral blood flow in areas appearing as white matter hyperintensities on magnetic resonance imaging. Psychiatry Res. 2009 May 15;172(2):117–120. doi: 10.1016/j.pscychresns.2008.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Vardi N, Freedman N, Lester H, Gomori JM, Chisin R, Lerer B, et al. Hyperintensities on T2-weighted images in the basal ganglia of patients with major depression: cerebral perfusion and clinical implications. Psychiatry Res. 2011 May 31;192(2):125–130. doi: 10.1016/j.pscychresns.2010.11.010. [DOI] [PubMed] [Google Scholar]
  • 272.Nitschke JB, Mackiewicz KL. Prefrontal and anterior cingulate contributions to volition in depression. Int Rev Neurobiol. 2005;67:73–94. doi: 10.1016/S0074-7742(05)67003-1. [DOI] [PubMed] [Google Scholar]
  • 273.Videbech P, Ravnkilde B, Pedersen TH, Hartvig H, Egander A, Clemmensen K, et al. The Danish PET/depression project: clinical symptoms and cerebral blood flow. A regions-of-interest analysis. Acta Psychiatr Scand. 2002 Jul;106(1):35–44. doi: 10.1034/j.1600-0447.2002.02245.x. [DOI] [PubMed] [Google Scholar]
  • 274.Lui S, Parkes LM, Huang X, Zou K, Chan RC, Yang H, et al. Depressive disorders: focally altered cerebral perfusion measured with arterial spin-labeling MR imaging. Radiology. 2009 May;251(2):476–484. doi: 10.1148/radiol.2512081548. [DOI] [PubMed] [Google Scholar]
  • 275.Duhameau B, Ferre JC, Jannin P, Gauvrit JY, Verin M, Millet B, et al. Chronic and treatment-resistant depression: a study using arterial spin labeling perfusion MRI at 3Tesla. Psychiatry Res. 2010 May 30;182(2):111–116. doi: 10.1016/j.pscychresns.2010.01.009. [DOI] [PubMed] [Google Scholar]
  • 276.Jarnum H, Eskildsen SF, Steffensen EG, Lundbye-Christensen S, Simonsen CW, Thomsen IS, et al. Longitudinal MRI study of cortical thickness, perfusion, and metabolite levels in major depressive disorder. Acta Psychiatr Scand. 2011 Dec;124(6):435–446. doi: 10.1111/j.1600-0447.2011.01766.x. [DOI] [PubMed] [Google Scholar]
  • 277.Asllani I, Habeck C, Borogovac A, Brown TR, Brickman AM, Stern Y. Separating function from structure in perfusion imaging of the aging brain. Hum Brain Mapp. 2009 Sep;30(9):2927–2935. doi: 10.1002/hbm.20719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Claus JJ, Breteler MM, Hasan D, Krenning EP, Bots ML, Grobbee DE, et al. Regional cerebral blood flow and cerebrovascular risk factors in the elderly population. Neurobiol Aging. 1998 Jan-Feb;19(1):57–64. doi: 10.1016/s0197-4580(98)00004-9. [DOI] [PubMed] [Google Scholar]
  • 279.Dotson VM, Beason-Held L, Kraut MA, Resnick SM. Longitudinal study of chronic depressive symptoms and regional cerebral blood flow in older men and women. Int J Geriatr Psychiatry. 2009 Aug;24(8):809–819. doi: 10.1002/gps.2298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Lesser IM, Mena I, Boone KB, Miller BL, Mehringer MC, Wohl M. Reduction of cerebral blood flow in older depressed patients. Arch Gen Psychiatry. 1994;51:677–686. doi: 10.1001/archpsyc.1994.03950090009002. [DOI] [PubMed] [Google Scholar]
  • 281.Ishizaki J, Yamamoto H, Takahashi T, Takeda M, Yano M, Mimura M. Changes in regional cerebral blood flow following antidepressant treatment in late-life depression. Int J Geriatr Psychiatry. 2008 Aug;23(8):805–811. doi: 10.1002/gps.1980. [DOI] [PubMed] [Google Scholar]
  • 282.Vasile RG, Schwartz RB, Garada B, Holman BL, Alpert M, Davidson PB, et al. Focal cerebral perfusion defects demonstrated by 99mTc-hexamethylpropyleneamine oxime SPECT in elderly depressed patients. Psychiatry Res. 1996 May 31;67(1):59–70. doi: 10.1016/0925-4927(96)02689-3. [DOI] [PubMed] [Google Scholar]
  • 283.Ebmeier KP, Glabus MF, Prentice N, Ryman A, Goodwin GM. A voxel-based analysis of cerebral perfusion in dementia and depression of old age. Neuroimage. 1998;7:199–208. doi: 10.1006/nimg.1998.0321. [DOI] [PubMed] [Google Scholar]
  • 284.Vangu MD, Esser JD, Boyd IH, Berk M. Effects of electroconvulsive therapy on regional cerebral blood flow measured by 99mtechnetium HMPAO SPECT. Prog Neuropsychopharmacol Biol Psychiatry. 2003 Feb;27(1):15–19. doi: 10.1016/s0278-5846(02)00309-3. [DOI] [PubMed] [Google Scholar]
  • 285.Kohn Y, Freedman N, Lester H, Krausz Y, Chisin R, Lerer B, et al. 99mTc-HMPAO SPECT study of cerebral perfusion after treatment with medication and electroconvulsive therapy in major depression. J Nucl Med. 2007 Aug;48(8):1273–1278. doi: 10.2967/jnumed.106.039354. [DOI] [PubMed] [Google Scholar]
  • 286.Milo TJ, Kaufman GE, Barnes WE, Konopka LM, Crayton JW, Ringelstein JG, et al. Changes in regional cerebral blood flow after electroconvulsive therapy for depression. J ECT. 2001 Mar;17(1):15–21. doi: 10.1097/00124509-200103000-00004. [DOI] [PubMed] [Google Scholar]
  • 287.Vlassenko A, Sheline YI, Fischer K, Mintun MA. Cerebral perfusion response to successful treatment of depression with different serotoninergic agents. J Neuropsychiatry Clin Neurosci. 2004;16:360–363. doi: 10.1176/jnp.16.3.360. [DOI] [PubMed] [Google Scholar]
  • 288.Bench CJ, Frackowiak RS, Dolan RJ. Changes in regional cerebral blood flow on recovery from depression. Psychol Med. 1995 Mar;25(2):247–261. doi: 10.1017/s0033291700036151. [DOI] [PubMed] [Google Scholar]
  • 289.Mazza M, Marano G, Traversi G, Bria P, Mazza S. Primary cerebral blood flow deficiency and Alzheimer's disease: shadows and lights. J Alzheimers Dis. 2011;23(3):375–389. doi: 10.3233/JAD-2010-090700. [DOI] [PubMed] [Google Scholar]
  • 290.Chao LL, Buckley ST, Kornak J, Schuff N, Madison C, Yaffe K, et al. ASL perfusion MRI predicts cognitive decline and conversion from MCI to dementia. Alzheimer Dis Assoc Disord. 2010 Jan-Mar;24(1):19–27. doi: 10.1097/WAD.0b013e3181b4f736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Chao LL, Pa J, Duarte A, Schuff N, Weiner MW, Kramer JH, et al. Patterns of cerebral hypoperfusion in amnestic and dysexecutive MCI. Alzheimer Dis Assoc Disord. 2009 Jul-Sep;23(3):245–252. doi: 10.1097/WAD.0b013e318199ff46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Heo S, Prakash RS, Voss MW, Erickson KI, Ouyang C, Sutton BP, et al. Resting hippocampal blood flow, spatial memory and aging. Brain Res. 2010 Feb 22;1315:119–127. doi: 10.1016/j.brainres.2009.12.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Terada S, Sato S, Honda H, Kishimoto Y, Takeda N, Oshima E, et al. Perseverative errors on the Wisconsin Card Sorting Test and brain perfusion imaging in mild Alzheimer's disease. Int Psychogeriatr. 2011 Dec;23(10):1552–1559. doi: 10.1017/S1041610211001463. [DOI] [PubMed] [Google Scholar]
  • 294.Takeda N, Terada S, Sato S, Honda H, Yoshida H, Kishimoto Y, et al. Wisconsin card sorting test and brain perfusion imaging in early dementia. Dement Geriatr Cogn Disord. 2010;29(1):21–27. doi: 10.1159/000261645. [DOI] [PubMed] [Google Scholar]
  • 295.Rabbitt P, Scott M, Thacker N, Lowe C, Jackson A, Horan M, et al. Losses in gross brain volume and cerebral blood flow account for age-related differences in speed but not in fluid intelligence. Neuropsychology. 2006 Sep;20(5):549–557. doi: 10.1037/0894-4105.20.5.549. [DOI] [PubMed] [Google Scholar]
  • 296.Ghiadoni L, Virdis A, Magagna A, Taddei S, Salvetti A. Effect of the angiotensin II type 1 receptor blocker candesartan on endothelial function in patients with essential hypertension. Hypertension. 2000 Jan;35(1 Pt 2):501–506. doi: 10.1161/01.hyp.35.1.501. [DOI] [PubMed] [Google Scholar]
  • 297.Nagata R, Kawabe K, Ikeda K. Olmesartan, an angiotensin II receptor blocker, restores cerebral hypoperfusion in elderly patients with hypertension. J Stroke Cerebrovasc Dis. 2010 May;19(3):236–240. doi: 10.1016/j.jstrokecerebrovasdis.2009.08.004. [DOI] [PubMed] [Google Scholar]
  • 298.Virdis A, Schiffrin EL. Vascular inflammation: a role in vascular disease in hypertension? Curr Opin Nephrol Hypertens. 2003 Mar;12(2):181–187. doi: 10.1097/00041552-200303000-00009. [DOI] [PubMed] [Google Scholar]
  • 299.Almeida OP, McCaul K, Hankey GJ, Norman P, Jamrozik K, Flicker L. Homocysteine and depression in later life. Arch Gen Psychiatry. 2008 Nov;65(11):1286–1294. doi: 10.1001/archpsyc.65.11.1286. [DOI] [PubMed] [Google Scholar]
  • 300.Weiss N. Mechanisms of increased vascular oxidant stress in hyperhomocys-teinemia and its impact on endothelial function. Curr Drug Metab. 2005 Feb;6(1):27–36. doi: 10.2174/1389200052997357. [DOI] [PubMed] [Google Scholar]
  • 301.Pariante CM. Risk factors for development of depression and psychosis. Glucocorticoid receptors and pituitary implications for treatment with antidepressant and glucocorticoids. Ann N Y Acad Sci. 2009 Oct;1179:144–152. doi: 10.1111/j.1749-6632.2009.04978.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Grinevich V, Harbuz M, Ma XM, Jessop D, Tilders FJ, Lightman SL, et al. Hypothalamic pituitary adrenal axis and immune responses to endotoxin in rats with chronic adjuvant-induced arthritis. Exp Neurol. 2002 Nov;178(1):112–123. doi: 10.1006/exnr.2002.8022. [DOI] [PubMed] [Google Scholar]
  • 303.Chalon S. Omega-3 fatty acids and monoamine neurotransmission. Prostaglandins, leukotrienes, and essential fatty acids. 2006 Oct-Nov;75(4-5):259–269. doi: 10.1016/j.plefa.2006.07.005. [DOI] [PubMed] [Google Scholar]
  • 304.Bloch MH, Hannestad J. Response to critiques on ‘Omega-3 fatty acids for the treatment of depression: systematic review and meta-analysis’. Mol Psychiatry. 2012 Dec;17(12):1163–1167. doi: 10.1038/mp.2011.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Bloch MH, Hannestad J. Omega-3 fatty acids for the treatment of depression: systematic review and meta-analysis. Mol Psychiatry. 2012 Dec;17(12):1272–1282. doi: 10.1038/mp.2011.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.Fogari R, Mugellini A, Zoppi A, Derosa G, Pasotti C, Fogari E, et al. Influence of losartan and atenolol on memory function in very elderly hypertensive patients. J Hum Hypertens. 2003 Nov;17(11):781–785. doi: 10.1038/sj.jhh.1001613. [DOI] [PubMed] [Google Scholar]
  • 307.Saxby BK, Harrington F, Wesnes KA, McKeith IG, Ford GA. Candesartan and cognitive decline in older patients with hypertension: a substudy of the SCOPE trial. Neurology. 2008 May 6;70(19 Pt 2):1858–1866. doi: 10.1212/01.wnl.0000311447.85948.78. [DOI] [PubMed] [Google Scholar]
  • 308.Hajjar I, Hart M, Chen YL, Mack W, Milberg W, Chui H, et al. Effect of antihypertensive therapy on cognitive function in early executive cognitive impairment: a double-blind randomized clinical trial. Arch Intern Med. 2012 Mar 12;172(5):442–444. doi: 10.1001/archinternmed.2011.1391. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES