Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Aug 19.
Published in final edited form as: Psychiatr Clin North Am. 2011 Jun;34(2):423–ix. doi: 10.1016/j.psc.2011.02.001

Structural Neuroimaging of Geriatric Depression

Sophiya Benjamin 1, David C Steffens 1,2
PMCID: PMC3746760  NIHMSID: NIHMS493894  PMID: 21536166

Abstract

There is a large literature on the neuroanatomy of late-life depression which continues to grow with the discovery of novel structural imaging techniques along with innovative methods to analyze the images. Such advances have helped identify specific areas as well characteristic lesions in the brain and changes in the chemical composition in these regions that might be important in the pathophysiology of this complex disease. In this article we review the relevant findings by each structural neuroimaging technique. When validated across many studies, such findings can serve as neuroanatomic markers that can help generate rational hypotheses for future studies to further our understanding of geriatric depression.

Keywords: Geriatric depression, late life depression, neuroimaging, MRI, white matter hyperintensities, magnetic resonance spectroscopy

Introduction

Neuroimaging has been a powerful tool in our search to identify neuroanatomic markers that provide information about the diagnostic and prognostic status of elderly patients with depressive symptoms. After decades of research, we now know several regions in the brain that are implicated in depression such as the anterior cingulate, orbitofrontal cortex and the hippocampus. Successes in delineating the neural circuits in depression have closely paralleled progress in neuroimaging techniques and advances in image analysis. Such non-invasive methods are critical as access to the tissue of interest, the brain, is otherwise impossible except in post-mortem samples.

Magnetic resonance (MR) or nuclear magnetic resonance occurs when protons in the nuclei of certain atoms (usually hydrogen) are subjected to a static magnetic field and then, exposed to a second oscillating magnetic field (pulse). During the application of the pulse, the alignment of the protons within the static magnetic field is disturbed. Following the application of the pulse, the misaligned protons relax and return to their original alignment emitting energy signals in the process. The relaxation times of different tissues vary, forming the basis of magnetic resonance imaging (MRI).(1) MR based methods such as MR morphometry, diffusion tensor imaging and MR spectroscopy among others are used detect differences between the brains of depressed and non-depressed elderly. Readers interested in further details about individual methods, their strengths and weaknesses in the context of geriatric depression are referred to the excellent review by Hoptman et al.(1)

Several differences in the structure of the brain between depressed and non-depressed elderly have been found and replicated. In this review, we summarize some of the salient structural imaging findings in geriatric depression and their implications to the neurobiology of late life depression. First we discuss morphometric studies focusing on volumetric differences; then, we discuss the findings from studies that examine white matter pathology using different imaging techniques. Finally, we summarize the biochemical correlates of depression found by magnetic resonance spectroscopy.

Volumetric Differences in Specific Brain Structures

Volumetric studies examine differences in the volumes of different brain structures among patients with depression compared with those who are not depressed. This can be accomplished either by traditional morphometric studies which focus on a predetermined, specific structure, based on previous knowledge about pathophysiology or voxel based morphometry which is not biased towards any one structure and assesses anatomical differences throughout the brain.

Morphometric studies

Most morphometric studies have utilized T1- weighted images to compare the volumes of specific structures such as the hippocampus or a particular region in the frontal cortex. These areas are called regions of interest (ROI). This hypothesis driven approach has identified several volumetric differences between depressed and non-depressed elderly.

The hippocampus

The hippocampus is one of the most commonly studied structures in depression. Studies in late life depression have repeatedly demonstrated a reduction in hippocampal volume among the depressed. (2-7) Though some studies did find negative results,(8, 9) two recent meta-analyses of MRI studies have confirmed that there is indeed an association between depression and decreased hippocampal volume.(10, 11) It has been suggested that earlier negative findings may be due either to the inclusion of the amygdala along with the hippocampus (12) or lower resolution of images. (13) Further exploration of this association has revealed that age of onset of depression correlates negatively with hippocampal volume as patients with late-onset depression had smaller hippocampal volumes when compared to those with early onset depression and controls. (4) In a younger cohort of patients followed over 3 years, depressed participants exhibited greater decline in bilateral hippocampal volume. (14)

Evidence from animal studies indicates that the mechanism underlying decreased hippocampal volumes is stress induced decrease in cell proliferation in the hippocampal region. (15, 16) Further, administration of antidepressants has been shown to prevent such stress induced suppression of neurogenisis.(17) Hippocampal volume reduction has also been shown to correlate with serotonin transporter promoter region polymorphism (5-HTTLPR) where individuals who were homozygous for the L allele exhibited smaller hippocampal volumes. (18) A similar correlation with the val66met polymorphism of the brain derived neurotrophic factor (BDNF) gene has been found to be positive in younger adults (19) but was not replicated in an elderly sample. (20) Though decrease in hippocampal volume has been proposed as an endophenotypes for depression, it is by no means specific as similar decreases occur in mild cognitive impairment and Alzheimer’s dementia which are common co-morbidities in older adults with depression.(21, 22)

The amygdala

The amygdala is an important structure in emotion regulation as it identifies and integrates the emotional salience with perception. Functional studies have demonstrated that patients with depression have increased reactivity of the amygdala to negative stimuli which can be reversed by antidepressant treatment. (23) However, findings from structural studies have been conflicting as studies in younger cohorts have shown an increase in volume during the first episode of depression but not in those with recurrent episodes; further, it did not correlate with age of onset, illness duration or severity of symptoms.(24) Earlier studies included both the amygdala and hippocampus together making the results hard to interpret.(8) A meta-analysis combining amygdala volumes in depression from 14 studies found that there was a significant heterogeneity among the studies and that they was no significant association between amygdala volume and depression.(10) There is some evidence that the differences in volume might be heritable thereby making it harder to detect differences between depressed and non-depressed samples(25).

The striatum

Decreased volumes of the caudate (26) and putamen(27) which are important structures in the corticostriatal circuit were observed in depressed samples. This was later replicated in depressed elderly (28, 29) supporting the role of subcortical structures in geriatric depression. Another study found that a decrease in caudate volume predicted psychomotor slowing in older adults with depression. (30) Some studies did not find the above mentioned differences in basal ganglia volumes, however, this may have been due to differences in sample selection as the patients were younger (31) and free of medical co-morbidities, specifically, cardiovascular risk.(32)

The anterior cingulate cortex (ACC)

Though the entire cingulate cortex was initially thought to be involved with emotion and behavior, the more recent understanding is that the ACC is important in emotion while the posterior part is more important in visuospatial function and memory. (33) The ACC is further subdivided into ‘affect’ and ‘cognition’ subdivisions (33) both of which are implicated in geriatric depression.(34) Some of the functions of the ACC salient to depression include conditioned emotional learning, vocalizations associated with expressing internal states, assessment of motivational content and assigning emotional valence to stimuli.(33)

In a meta-analysis of several structures involved in depression, the ACC had the largest effect, with depressed individuals having smaller ACC volumes.(10) Though not specific to elderly samples, some studies have demonstrated decreased volumes of the ACC in depressed individuals (35, 36) while others failed to do so. (37, 38) Another study that limited it’s sample to the elderly found significant bilateral reductions in gray matter volume in the ACC.(39)

Orbitofrontal cortex (OFC)

The OFC functions as part of a network which includes the hippocampus, amygdala and basal ganglia. It is involved in integrating sensory experiences and in emotional and reward related learning and decision making.(40) In elderly samples, compared with nondepressed subjects, depressed cohorts exhibit smaller OFC volumes.(39, 41, 42) There have been similar findings in younger adults (37, 43) though negative results have also been reported. (44) The decreased in OFC volume in the depressed is consistent with postmortem findings which show a reduction in the density of pyramidal neurons in this region in the depressed.(45)

In a study that examined the functional implications of the OFC in depressed elderly, decreased left OFC volume was associated with poorer performance on the Benton Visual Retention Test (BVRT). (46) The BVRT measures perception of spatial relations and memory for newly learned material. Depressed elderly with smaller left OFC volumes made more preservative errors and scored lower on the overall test.(46) Further, smaller medial orbitofrontal gyri volumes was found to be associated with impairment in both basic as well as instrumental activities of daily living.(47) Though no causal inferences can be made from either of the above cross sectional studies, they do expand our current understanding of the mechanisms that underlie cognitive dysfunction and functional impairment in depression.

Voxel Based Morphometry

Voxel based morphometry (VBM) is a highly automated method with a hypothesis free approach, not requiring a priori assumptions about the relevance of specific brain regions. VBM consists of the following four steps: spatial normalization which transforms all the subjects’ data into the same stereotactic space; partitioning the spatially normalized images into segments such as gray matter, white matter and CSF; preprocessing the gray matter segment to make enable further voxel-by-voxel analysis to be comparable to the ROI approach; and, comparing the segment of interest such as the gray matter between the groups voxel-by-voxel.(48) VBM is a more recently described method compared to the ROI approach and the literature in geriatric depression using this technique is still in its early stages. Results from recent studies using this technique are encouraging in that the regions that have been identified are by often those that have been identified by previous structural and functional studies. As the literature from VBM is not as voluminous as that from traditional morphometric methods, results about different brain structures are presented together in this section.

In a study of 30 depressed and 47 non depressed elderly, depressed patients were found to have smaller right hippocampal volumes compared to control subjects and the volume of the hippocampal-entotrhinal cortex was inversely associated with the duration since the first episode of depression. (49) In another study, VBM revealed decreases in the volume of the right rostral hippocampus, in the right amygdala and the medial orbito-frontal cortex bilaterally.(50) Additionally, the grey matter volume of both the right and left medial orbito-frontal cortex correlated negatively with scores on the geriatric depression scale.(50)

In-patients with late-onset depression were found to have smaller volumes in several regions of grey matter including the insula and the posterior cingulate region and white matter including the subcallosal cingulate cortex, floor of lateral ventricles, parahippocampal region, insula, and the cerebellum.(51) Compared with the depressed who did not attempt suicide, those who attempted suicide had decreased grey matter and white matter volume in the frontal, parietal, and temporal regions, and the insula, lentiform nucleus, midbrain, and the cerebellum. (51)

In a study that examined first-episode remitted geriatric depression, patients with remitted depression had smaller volumes of right superior frontal cortex, left postcentral cortex, and right middle temporal gyrus and larger left cingulate gyus volume compared with healthy control subjects.(52) In patients with remitted depression, the volume of the left cingulated gyrus correlated negatively with scores on the Rey Auditory Verbal Learning Test and delayed recall(52) providing further evidence that specific brain regions involved in depression might also be involved in cognitive impairment seen so often in the depressed elderly.

A limitation of the aforementioned studies is their small sample size which range from thirty-four (50) to seventy-seven.(49) Though many of the published studies have found significant associations, negative findings have also been reported (53) and larger sample sizes that could clarify these discrepancies are needed. False positives are an inherent problem in any technique where the number of comparisons is large and the sample size small; as in the case of VBM, however, several statistical methods to control for this have been described. (48, 54) Another disadvantage that might be specific to geriatric depression is that this technique does not differentiate between vascular and degenerative causes of differences in regional brain volume. Despite the above limitations, early results are promising and VBM has the potential to identify new structural variations that could expand our current understanding of geriatric depression.

White Matter Pathology

White matter hyperintensities (WMH) are thought to be caused by small, silent cerebral infarctions. (55) Such silent cerebral infarctions were observed in 65.9% of patients with early or presenile-onset depression and 93.7% of those with late onset depression.(55) Early observations that older individuals with depression have a greater severity of clinically silent ischemic disease which were observable as hyperintense lesions on MRI scans(56) as well as clinical characteristics such as increased cognitive dysfunction(57) led to the advent of the “vascular depression hypothesis”.(58) White matter hyperintensities can be detected by various structural imaging methods including T2 weighted MRI scans, diffusion tensor imaging (DTI) and magnetization transfer imaging (MTI).

Findings from T2 MRI studies

On T2-weighted MRI studies, the hyperintense signals are present in the white matter and can be classified into three major groups- periventricular hyperintensities (PVH), deep white matter hyperintensities (DWMH) and subcortical hyperintensities (SCH). Older depressed individuals have consistently been found to have more white matter hyperintensities than older healthy controls without depression.(59-63) Further, such lesions are more common in late-onset depression compared with early onset depression. (56) In a cohort of post stroke patients, those who developed post-stroke depression, were more likely to have severe DWMH (12.8% vs. 1.3%; p=0.009). (64)

Beyond just diagnosis and classification, white matter hyperintensities have been shown to be important in the course of illness and outcome of depression as subcortical white matter lesions were associated with occurrence, persistence, worsening and severity of depressive symptoms. (65, 66) Additionally, greater progression of white matter lesions was associated with poorer treatment outcomes as depressed elderly who had greater WMH volume did not achieve or sustain remission when compared to depressed elderly who had lower WMH volume.(67) Even in the absence of overt cerebrovascular disease, time to relapse was shorter in those who had severe deep white matter lesions.(68)

White matter hyperintensities have also been shown to predict response to antidepressant treatment. WMH burden predicted MADRS scores over a 12-week course of sertraline. Further, WMH correlated with neuropsychological testing measures which also predicted depression outcome with treatment, and both these variables correlated with the Framingham vascular risk factor scores, supporting the vascular depression hypothesis.(69) In another study, patients with depression who failed to remit after a 12-week controlled trial of escitalopram had greater MRI signal hyperintensity burden compared to those who remitted as well as elderly comparisons but, there was no difference in signal hypenintensity burden between those who remitted and elderly control subjects.(70)

Based on the evidence that vascular depression can confer risk for adverse outcomes and is produced by a pathology that separates it from other forms of depression, it has been argued that “subcortical ischemic depression” be considered as a unique and valid diagnosis corresponding to the “vascular depression” hypothesis.(71) In a more recent study replicated in two independent clinical samples, deep white matter hyperintensities was found to be the most accurate marker for classifying depression into vascular vs. non-vascular depression.(72)

Diffusion tensor neuroimaging

Diffusion tensor imaging is an MRI based method that measures the self-diffusion of water which can be isotropic, when it occurs equally in all directions when no barriers are present, and anisotropic when the diffusion of water tends to follow along external barriers.(1) White matter tracts in the brain form organized barriers along which water can diffuse making the flow anisotropic. When such tracts are disrupted, the diffusion will be less anisotropic. Fractional anisotropy (FA) is a common measure used to characterize the integrity of the neural circuit. Using measures such as FA and apparent diffusion coefficient, DTI can help further our understanding of white matter pathology in geriatric depression by enabling researchers to locate and quantify the structure and orientation of cerebral white matter tracts.

In one of the earlier studies, regions with hyperintensities in the depressed elderly showed increase apparent diffusion coefficient and decreased fractional anisotropy when compared with normal regions though there was no significant difference in diffusion characteristics between the depressed and non depressed. (73) A more recent study showed significantly lower fractional anisotropy in the right superior frontal gyrus white matter of depressed patients. (74) Another study of one hundred and six depressed and eighty-four non depressed elderly participants found that depressed patients had significantly lower fractional anisotropy in the white matter lateral to the right anterior cingulate cortex, bilateral superior frontal gyri and the left middle frontal gyrus.(75) Such findings have been replicated in different populations: in a Chinese sample, late life-depression was associated with decrease fractional anisotropy in the frontal (superior and middle frontal gyrus), and temporal (right parahippocampal gyrus) regions.(76)

Diffusion anisotropy was lower in several regions of the brain including right superior frontal gyrus, left inferior frontal gyrus, left middle temporal gyrus, right inferior parietal lobule, right middle occipital gyrus, left lingual gyrus, right putamen and right caudate in patients with first-episode remitted geriatric depression.(77) Though most studies have concentrated on DTI imaging of white matter pathology, a recent study used DTI to examine the integrity of normal appearing white matter and found that depressed elderly had widespread abnormalities in DTI measures in the prefrontal region. (78) These studies strengthen the evidence for the possibility that decreased fractional anisotropy could affect the connectivity of the dorsolateral prefrontal circuit and the anterior cingulate circuit resulting in disconnection of cortical and subcortical structures, thereby resulting in depression. Further, decreased integrity of white matter tracts are also associated with decreased executive function, commonly seen in late life depression.(79)

Such microstructural abnormalities have also been shown to be associated with the severity of depressive symptoms and the likelihood of remission with treatment. Higher FA values in a region 8mm below the anterior commisure-posterior commisure line correlated with lower Hamilton Depression Rating Scale scores.(80) In another study, increased FA the region 15 mm above the anterior commisure-posterior commisure plane was associated with low remission after treatment with citalopram.(81) Similarly, failure to remit with sertraline was associated with increased FA in the superior frontal gyri and anterior cingulate cortices bilaterally. (82) In another study of depression treatment, depressed patients who had decreased FA compared to non-depressed showed an increase in frontal FA after electroconvulsive treatment.(83)

Magnetization Transfer Imaging

Magnetization Transfer MRI (MT-MRI) is another MRI based method used to examine biophysical properties of brain tissue. MT- MRI utilizes the two types of water molecules present in biological tissues, free water and water bound to molecules. Proton MRI detects signal from mobile protons or free water which have longer T2 relaxation times. The T2 relaxation times of protons associated with macromolecules are too short to be detected by conventional MRI. However, coupling between the bound and free protons allows the bound protons to influence the spin state of the mobile ones. MT imaging requires two image acquisitions where the first is similar to an MRI study. The second, which is the magnetization transfer acquisition, is acquired by first saturating the bound water by an off resonance radiofrequency pulse which negates the potential for the bound water to create a signal which then creates a signal reduction relative to the first image. The difference in signal intensity between the two images is the magnetization transfer ratio (MTR).(1, 84, 85)

One of the earliest studies using MTR in late-life depression found that older depressed patients had lower MTRs in the genu and splenium of the corpus callosum, the neostriatum, and the occipital WM.(84) Of note, all of these differences were in normal appearing white matter tracts which were free of hyperintense lesions.(84) In a more recent study with a larger sample size of fifty-five older patients with depression and twenty -four comparison subjects, depressed patients had lowed MTRs in multiple left hemisphere frontostriatal and limbic regions, including white matter lateral to the lentiform nuclei, dorsolateral and dorsomedial prefrontal, dorsal anterior cingulate, subcallosal, periamygdalar, insular, and posterior cingulate regions, complementing previous findings from volumetric and DTI studies.(86)

Magnetic Resonance Spectroscopy and biochemical Correlates of Depression

Magnetic Resonance Spectroscopy (MRS) is another non-invasive imaging method which unlike all the other techniques described above, characterizes chemical and cellular features in vivo. In the brain, concentrations and mobility of low molecular weight chemicals can be measured as spectral peaks which can then be used to identify abnormalities in brain regions that appear normal in MRI. (87) The measurements are usually not absolute and are presented as the ratio of the measure of interest to a standard metabolite such as creatine. (1)

In a preliminary study of twenty elderly patients with depression and 18 comparison subjects, myo-Inositol (mI)/creatine (Cr) and choline (Cho)/Cr ratios were significantly higher in the frontal white matter in the depressed group.(88) A subsequent study explored the relationship between these metabolites in a voxel in the dorsolateral cortex, and cognitive function in the elderly. Among nondepressed subjects, cognition positively correlated with Cho/Cr and mI/Cr and negatively correlated with phosphocholine/Cr in four domains of verbal learning, recognition, recall, and hypothesis generation, whereas, depressed patients did not have consistent relationships between the metabolites.(89) Thus imaging studies have revealed both neuroanatomic as well as biochemical changes in the frontostriatal circuitry that may be associated with cognitive changes in depression.

In a more recent study, patients with late-life major depressive disorder had a significantly lower N-acetyl aspartate (NAA)/Cr ratio in the left frontal white matter, and higher Cho/Cr and mI/Cr ratios in the left basal ganglia when compared with the control subjects.(90) Further, the myo-Inositol correlated with global cognitive function. (90) In older patients whose depression responded to treatment, concentrations of total NAA, choline, and creatine were significantly decreased in the prefrontal cortex while, concentrations of NAA and myo-Inositol were significantly elevated in the left medial temporal lobe.(91) These authors concluded that reduced neuronal phospolipid, and energy metabolism in the prefrontal cortex persists in clinically improved depression and that the elevated metabolites in the temporal lobe might be associated with gial cell changes in the amygdala.(91)

MRS of the brain before antidepressant treatment of post stroke depression, followed by a second MRS six months after, revealed changes in metabolites in various regions.(92) Before treatment, NAA/creatine ratios in the bilateral hippocampus and thalami were significantly lower in post stroke depression patients than in controls. Choline/creatine (Cho/Cr) ratios were significantly higher in the bilateral hippocampus and left thalamus in post stroke depression patients than in controls. Further, Hamilton Depression Rating Scale scores significantly correlated with the Cho/Cr ratios in the left and right hippocampus. After treatment, patients had significantly higher NAA/Cr ratios in the left hippocampus and bilateral thalami and significantly lower Cho/Cr ratios in bilateral hippocampus and left thalamus.(92) This longitudinal study sheds light on some of the biochemical changes that occur in the brain with antidepressant treatment. The authors postulate that these changes might be due to the neurotrophic effects of antidepressants.

Conclusions

This review has highlighted some of the important findings in geriatric depression from structural imaging. Depression is a complex disease with multiple etiologies as evidenced by the many structures involved and the different kinds of lesions that are associated with the illness. Evolution of imaging methods and the refinement of its techniques have enabled the identification and assessment of many potential biomarkers and endophenotypes in depression. The future of imaging research will be multimodal, in which identified structural variations will be used to inform and improve hypotheses for further testing and confirmation by other techniques. In the case of the orbitofrontal cortex, an initial neuroanatomic difference of decreased volume among the depressed (42) was then studied with cognitive neurological testing which found that this volumetric difference was associated with decreased performance in tests.(46) Further, lower OFC volumes were associated with functional disability.(47) Thus findings from imaging studies can then be explored using different research methods to interrogate the varied aspects of depression and the many negative outcomes it portends. When consistently replicated, such findings might serve as imaging phenotypes that can identify vulnerability and predict treatment efficacy. (93)

Footnotes

Financial Disclosures: The authors have nothing to disclose.

Contributor Information

Sophiya Benjamin, Email: sophiya.benjamin@duke.edu.

David C Steffens, Email: david.steffens@duke.edu.

References

  • 1.Hoptman MJ, Gunning-Dixon FM, Murphy CF, et al. Structural neuroimaging research methods in geriatric depression. Am J Geriatr Psychiatry. 2006 Oct;14(10):812–822. doi: 10.1097/01.JGP.0000238588.34205.bd. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Steffens DC, Byrum CE, McQuoid DR, et al. Hippocampal volume in geriatric depression. Biol Psychiatry. 2000 Aug 15;48(4):301–309. doi: 10.1016/s0006-3223(00)00829-5. [DOI] [PubMed] [Google Scholar]
  • 3.Sheline YI. Hippocampal atrophy in major depression: a result of depression-induced neurotoxicity? Mol Psychiatry. 1996 Sep;1(4):298–299. [PubMed] [Google Scholar]
  • 4.Lloyd AJ, Ferrier IN, Barber R, et al. Hippocampal volume change in depression: late- and early-onset illness compared. Br J Psychiatry. 2004 Jun;184:488–495. doi: 10.1192/bjp.184.6.488. [DOI] [PubMed] [Google Scholar]
  • 5.O’Brien JT, Lloyd A, McKeith I, et al. A longitudinal study of hippocampal volume, cortisol levels, and cognition in older depressed subjects. Am J Psychiatry. 2004 Nov;161(11):2081–2090. doi: 10.1176/appi.ajp.161.11.2081. [DOI] [PubMed] [Google Scholar]
  • 6.Hickie I, Naismith S, Ward PB, et al. Reduced hippocampal volumes and memory loss in patients with early- and late-onset depression. Br J Psychiatry. 2005 Mar;186:197–202. doi: 10.1192/bjp.186.3.197. [DOI] [PubMed] [Google Scholar]
  • 7.Ballmaier M, Narr KL, Toga AW, et al. Hippocampal morphology and distinguishing late-onset from early-onset elderly depression. Am J Psychiatry. 2008 Feb;165(2):229–237. doi: 10.1176/appi.ajp.2007.07030506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ashtari M, Greenwald BS, Kramer-Ginsberg E, et al. Hippocampal/amygdala volumes in geriatric depression. Psychol Med. 1999 May;29(3):629–638. doi: 10.1017/s0033291799008405. [DOI] [PubMed] [Google Scholar]
  • 9.Pantel J, Schroder J, Essig M, et al. Quantitative magnetic resonance imaging in geriatric depression and primary degenerative dementia. J Affect Disord. 1997 Jan;42(1):69–83. doi: 10.1016/s0165-0327(96)00105-x. [DOI] [PubMed] [Google Scholar]
  • 10.Koolschijn PC, van Haren NE, Lensvelt-Mulders GJ, et al. Brain volume abnormalities in major depressive disorder: a meta-analysis of magnetic resonance imaging studies. Hum Brain Mapp. 2009 Nov;30(11):3719–3735. doi: 10.1002/hbm.20801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Videbech P, Ravnkilde B. Hippocampal volume and depression: a meta-analysis of MRI studies. Am J Psychiatry. 2004 Nov;161(11):1957–1966. doi: 10.1176/appi.ajp.161.11.1957. [DOI] [PubMed] [Google Scholar]
  • 12.Campbell S, Marriott M, Nahmias C, et al. Lower hippocampal volume in patients suffering from depression: a meta-analysis. Am J Psychiatry. 2004 Apr;161(4):598–607. doi: 10.1176/appi.ajp.161.4.598. [DOI] [PubMed] [Google Scholar]
  • 13.Sheline YI, Mittler BL, Mintun MA. The hippocampus and depression. Eur Psychiatry. 2002 Jul;17(Suppl 3):300–305. doi: 10.1016/s0924-9338(02)00655-7. [DOI] [PubMed] [Google Scholar]
  • 14.Frodl TS, Koutsouleris N, Bottlender R, et al. Depression-related variation in brain morphology over 3 years: effects of stress? Arch Gen Psychiatry. 2008 Oct;65(10):1156–1165. doi: 10.1001/archpsyc.65.10.1156. [DOI] [PubMed] [Google Scholar]
  • 15.Gould E, McEwen BS, Tanapat P, et al. Neurogenesis in the dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA receptor activation. J Neurosci. 1997 Apr 1;17(7):2492–2498. doi: 10.1523/JNEUROSCI.17-07-02492.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gould E, Tanapat P, McEwen BS, et al. Proliferation of granule cell precursors in the dentate gyrus of adult monkeys is diminished by stress. Proc Natl Acad Sci U S A. 1998 Mar 17;95(6):3168–3171. doi: 10.1073/pnas.95.6.3168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Czeh B, Michaelis T, Watanabe T, et al. Stress-induced changes in cerebral metabolites, hippocampal volume, and cell proliferation are prevented by antidepressant treatment with tianeptine. Proc Natl Acad Sci U S A. 2001 Oct 23;98(22):12796–12801. doi: 10.1073/pnas.211427898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Taylor WD, Steffens DC, Payne ME, et al. Influence of serotonin transporter promoter region polymorphisms on hippocampal volumes in late-life depression. Arch Gen Psychiatry. 2005 May;62(5):537–544. doi: 10.1001/archpsyc.62.5.537. [DOI] [PubMed] [Google Scholar]
  • 19.Frodl T, Schule C, Schmitt G, et al. Association of the brain-derived neurotrophic factor Val66Met polymorphism with reduced hippocampal volumes in major depression. Arch Gen Psychiatry. 2007 Apr;64(4):410–416. doi: 10.1001/archpsyc.64.4.410. [DOI] [PubMed] [Google Scholar]
  • 20.Benjamin S, McQuoid DR, Potter GG, et al. The brain-derived neurotrophic factor Val66Met polymorphism, hippocampal volume, and cognitive function in geriatric depression. Am J Geriatr Psychiatry. Apr;18(4):323–331. doi: 10.1097/JGP.0b013e3181cabd2b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Convit A, De Leon MJ, Tarshish C, et al. Specific hippocampal volume reductions in individuals at risk for Alzheimer’s disease. Neurobiol Aging. 1997 Mar-Apr;18(2):131–138. doi: 10.1016/s0197-4580(97)00001-8. [DOI] [PubMed] [Google Scholar]
  • 22.Killiany RJ, Moss MB, Albert MS, et al. Temporal lobe regions on magnetic resonance imaging identify patients with early Alzheimer’s disease. Arch Neurol. 1993 Sep;50(9):949–954. doi: 10.1001/archneur.1993.00540090052010. [DOI] [PubMed] [Google Scholar]
  • 23.Sheline YI, Barch DM, Donnelly JM, et al. Increased amygdala response to masked emotional faces in depressed subjects resolves with antidepressant treatment: an fMRI study. Biol Psychiatry. 2001 Nov 1;50(9):651–658. doi: 10.1016/s0006-3223(01)01263-x. [DOI] [PubMed] [Google Scholar]
  • 24.Frodl T, Meisenzahl EM, Zetzsche T, et al. Larger amygdala volumes in first depressive episode as compared to recurrent major depression and healthy control subjects. Biol Psychiatry. 2003 Feb 15;53(4):338–344. doi: 10.1016/s0006-3223(02)01474-9. [DOI] [PubMed] [Google Scholar]
  • 25.Munn MA, Alexopoulos J, Nishino T, et al. Amygdala volume analysis in female twins with major depression. Biol Psychiatry. 2007 Sep 1;62(5):415–422. doi: 10.1016/j.biopsych.2006.11.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Krishnan KR, McDonald WM, Escalona PR, et al. Magnetic resonance imaging of the caudate nuclei in depression. Preliminary observations. Arch Gen Psychiatry. 1992 Jul;49(7):553–557. doi: 10.1001/archpsyc.1992.01820070047007. [DOI] [PubMed] [Google Scholar]
  • 27.Husain MM, McDonald WM, Doraiswamy PM, et al. A magnetic resonance imaging study of putamen nuclei in major depression. Psychiatry Res. 1991 Oct;40(2):95–99. doi: 10.1016/0925-4927(91)90001-7. [DOI] [PubMed] [Google Scholar]
  • 28.Krishnan KR. Neuroanatomic substrates of depression in the elderly. J Geriatr Psychiatry Neurol. 1993 Jan-Mar;6(1):39–58. doi: 10.1177/002383099300600107. [DOI] [PubMed] [Google Scholar]
  • 29.Parashos IA, Tupler LA, Blitchington T, et al. Magnetic-resonance morphometry in patients with major depression. Psychiatry Res. 1998 Nov 9;84(1):7–15. doi: 10.1016/s0925-4927(98)00042-0. [DOI] [PubMed] [Google Scholar]
  • 30.Naismith S, Hickie I, Ward PB, et al. Caudate nucleus volumes and genetic determinants of homocysteine metabolism in the prediction of psychomotor speed in older persons with depression. Am J Psychiatry. 2002 Dec;159(12):2096–2098. doi: 10.1176/appi.ajp.159.12.2096. [DOI] [PubMed] [Google Scholar]
  • 31.Lacerda AL, Nicoletti MA, Brambilla P, et al. Anatomical MRI study of basal ganglia in major depressive disorder. Psychiatry Res. 2003 Nov 30;124(3):129–140. doi: 10.1016/s0925-4927(03)00123-9. [DOI] [PubMed] [Google Scholar]
  • 32.Lenze EJ, Sheline YI. Absence of striatal volume differences between depressed subjects with no comorbid medical illness and matched comparison subjects. Am J Psychiatry. 1999 Dec;156(12):1989–1991. doi: 10.1176/ajp.156.12.1989. [DOI] [PubMed] [Google Scholar]
  • 33.Devinsky O, Morrell MJ, Vogt BA. Contributions of anterior cingulate cortex to behaviour. Brain. 1995 Feb;118(Pt 1):279–306. doi: 10.1093/brain/118.1.279. [DOI] [PubMed] [Google Scholar]
  • 34.Alexopoulos GS, Gunning-Dixon FM, Latoussakis V, et al. Anterior cingulate dysfunction in geriatric depression. Int J Geriatr Psychiatry. 2008 Apr;23(4):347–355. doi: 10.1002/gps.1939. [DOI] [PubMed] [Google Scholar]
  • 35.Botteron KN, Raichle ME, Drevets WC, et al. Volumetric reduction in left subgenual prefrontal cortex in early onset depression. Biol Psychiatry. 2002 Feb 15;51(4):342–344. doi: 10.1016/s0006-3223(01)01280-x. [DOI] [PubMed] [Google Scholar]
  • 36.Caetano SC, Kaur S, Brambilla P, et al. Smaller cingulate volumes in unipolar depressed patients. Biol Psychiatry. 2006 Apr 15;59(8):702–706. doi: 10.1016/j.biopsych.2005.10.011. [DOI] [PubMed] [Google Scholar]
  • 37.Bremner JD, Vythilingam M, Vermetten E, et al. Reduced volume of orbitofrontal cortex in major depression. Biol Psychiatry. 2002 Feb 15;51(4):273–279. doi: 10.1016/s0006-3223(01)01336-1. [DOI] [PubMed] [Google Scholar]
  • 38.Brambilla P, Nicoletti MA, Harenski K, et al. Anatomical MRI study of subgenual prefrontal cortex in bipolar and unipolar subjects. Neuropsychopharmacology. 2002 Nov;27(5):792–799. doi: 10.1016/S0893-133X(02)00352-4. [DOI] [PubMed] [Google Scholar]
  • 39.Ballmaier M, Toga AW, Blanton RE, et al. Anterior cingulate, gyrus rectus, and orbitofrontal abnormalities in elderly depressed patients: an MRI-based parcellation of the prefrontal cortex. Am J Psychiatry. 2004 Jan;161(1):99–108. doi: 10.1176/appi.ajp.161.1.99. [DOI] [PubMed] [Google Scholar]
  • 40.Kringelbach ML. The human orbitofrontal cortex: linking reward to hedonic experience. Nat Rev Neurosci. 2005 Sep;6(9):691–702. doi: 10.1038/nrn1747. [DOI] [PubMed] [Google Scholar]
  • 41.Taylor WD, Macfall JR, Payne ME, et al. Orbitofrontal cortex volume in late life depression: influence of hyperintense lesions and genetic polymorphisms. Psychol Med. 2007 Dec;37(12):1763–1773. doi: 10.1017/S0033291707000128. [DOI] [PubMed] [Google Scholar]
  • 42.Lai T, Payne ME, Byrum CE, et al. Reduction of orbital frontal cortex volume in geriatric depression. Biol Psychiatry. 2000 Nov 15;48(10):971–975. doi: 10.1016/s0006-3223(00)01042-8. [DOI] [PubMed] [Google Scholar]
  • 43.Lacerda AL, Keshavan MS, Hardan AY, et al. Anatomic evaluation of the orbitofrontal cortex in major depressive disorder. Biol Psychiatry. 2004 Feb 15;55(4):353–358. doi: 10.1016/j.biopsych.2003.08.021. [DOI] [PubMed] [Google Scholar]
  • 44.Hastings RS, Parsey RV, Oquendo MA, et al. Volumetric analysis of the prefrontal cortex, amygdala, and hippocampus in major depression. Neuropsychopharmacology. 2004 May;29(5):952–959. doi: 10.1038/sj.npp.1300371. [DOI] [PubMed] [Google Scholar]
  • 45.Rajkowska G. Postmortem studies in mood disorders indicate altered numbers of neurons and glial cells. Biol Psychiatry. 2000 Oct 15;48(8):766–777. doi: 10.1016/s0006-3223(00)00950-1. [DOI] [PubMed] [Google Scholar]
  • 46.Steffens DC, McQuoid DR, Welsh-Bohmer KA, et al. Left orbital frontal cortex volume and performance on the benton visual retention test in older depressives and controls. Neuropsychopharmacology. 2003 Dec;28(12):2179–2183. doi: 10.1038/sj.npp.1300285. [DOI] [PubMed] [Google Scholar]
  • 47.Taylor WD, Steffens DC, McQuoid DR, et al. Smaller orbital frontal cortex volumes associated with functional disability in depressed elders. Biol Psychiatry. 2003 Jan 15;53(2):144–149. doi: 10.1016/s0006-3223(02)01490-7. [DOI] [PubMed] [Google Scholar]
  • 48.Ashburner J, Friston KJ. Voxel-based morphometry--the methods. Neuroimage. 2000 Jun;11(6 Pt 1):805–821. doi: 10.1006/nimg.2000.0582. [DOI] [PubMed] [Google Scholar]
  • 49.Bell-McGinty S, Butters MA, Meltzer CC, et al. Brain morphometric abnormalities in geriatric depression: long-term neurobiological effects of illness duration. Am J Psychiatry. 2002 Aug;159(8):1424–1427. doi: 10.1176/appi.ajp.159.8.1424. [DOI] [PubMed] [Google Scholar]
  • 50.Egger K, Schocke M, Weiss E, et al. Pattern of brain atrophy in elderly patients with depression revealed by voxel-based morphometry. Psychiatry Res. 2008 Dec 30;164(3):237–244. doi: 10.1016/j.pscychresns.2007.12.018. [DOI] [PubMed] [Google Scholar]
  • 51.Hwang JP, Lee TW, Tsai SJ, et al. Cortical and Subcortical Abnormalities in Late-Onset Depression With History of Suicide Attempts Investigated With MRI and Voxel-Based Morphometry. J Geriatr Psychiatry Neurol. Apr 29; doi: 10.1177/0891988710363713. [DOI] [PubMed] [Google Scholar]
  • 52.Yuan Y, Zhu W, Zhang Z, et al. Regional gray matter changes are associated with cognitive deficits in remitted geriatric depression: an optimized voxel-based morphometry study. Biol Psychiatry. 2008 Sep 15;64(6):541–544. doi: 10.1016/j.biopsych.2008.04.032. [DOI] [PubMed] [Google Scholar]
  • 53.Koolschijn PC, van Haren NE, Schnack HG, et al. Cortical thickness and voxel-based morphometry in depressed elderly. Eur Neuropsychopharmacol. Jun;20(6):398–404. doi: 10.1016/j.euroneuro.2010.02.010. [DOI] [PubMed] [Google Scholar]
  • 54.Baudewig J, Dechent P, Merboldt KD, et al. Thresholding in correlation analyses of magnetic resonance functional neuroimaging. Magn Reson Imaging. 2003 Dec;21(10):1121–1130. doi: 10.1016/j.mri.2003.08.013. [DOI] [PubMed] [Google Scholar]
  • 55.Fujikawa T, Yamawaki S, Touhouda Y. Incidence of silent cerebral infarction in patients with major depression. Stroke. 1993 Nov;24(11):1631–1634. doi: 10.1161/01.str.24.11.1631. [DOI] [PubMed] [Google Scholar]
  • 56.Krishnan KR, Hays JC, Blazer DG. MRI-defined vascular depression. Am J Psychiatry. 1997 Apr;154(4):497–501. doi: 10.1176/ajp.154.4.497. [DOI] [PubMed] [Google Scholar]
  • 57.Alexopoulos GS, Meyers BS, Young RC, et al. Clinically defined vascular depression. Am J Psychiatry. 1997 Apr;154(4):562–565. doi: 10.1176/ajp.154.4.562. [DOI] [PubMed] [Google Scholar]
  • 58.Alexopoulos GS, Meyers BS, Young RC, et al. ‘Vascular depression’ hypothesis. Arch Gen Psychiatry. 1997 Oct;54(10):915–922. doi: 10.1001/archpsyc.1997.01830220033006. [DOI] [PubMed] [Google Scholar]
  • 59.Coffey CE, Wilkinson WE, Weiner RD, et al. Quantitative cerebral anatomy in depression. A controlled magnetic resonance imaging study. Arch Gen Psychiatry. 1993 Jan;50(1):7–16. doi: 10.1001/archpsyc.1993.01820130009002. [DOI] [PubMed] [Google Scholar]
  • 60.Greenwald BS, Kramer-Ginsberg E, Krishnan RR, et al. MRI signal hyperintensities in geriatric depression. Am J Psychiatry. 1996 Sep;153(9):1212–1215. doi: 10.1176/ajp.153.9.1212. [DOI] [PubMed] [Google Scholar]
  • 61.Krishnan KR, McDonald WM, Doraiswamy PM, et al. Neuroanatomical substrates of depression in the elderly. Eur Arch Psychiatry Clin Neurosci. 1993;243(1):41–46. doi: 10.1007/BF02191522. [DOI] [PubMed] [Google Scholar]
  • 62.Kumar A, Bilker W, Jin Z, et al. Atrophy and high intensity lesions: complementary neurobiological mechanisms in late-life major depression. Neuropsychopharmacology. 2000 Mar;22(3):264–274. doi: 10.1016/S0893-133X(99)00124-4. [DOI] [PubMed] [Google Scholar]
  • 63.Taylor WD, MacFall JR, Payne ME, et al. Greater MRI lesion volumes in elderly depressed subjects than in control subjects. Psychiatry Res. 2005 May 30;139(1):1–7. doi: 10.1016/j.pscychresns.2004.08.004. [DOI] [PubMed] [Google Scholar]
  • 64.Tang WK, Chen YK, Lu JY, et al. White matter hyperintensities in post-stroke depression: a case control study. J Neurol Neurosurg Psychiatry. Jun 20; doi: 10.1136/jnnp.2009.203141. [DOI] [PubMed] [Google Scholar]
  • 65.Steffens DC, Krishnan KR, Crump C, et al. Cerebrovascular disease and evolution of depressive symptoms in the cardiovascular health study. Stroke. 2002 Jun;33(6):1636–1644. doi: 10.1161/01.str.0000018405.59799.d5. [DOI] [PubMed] [Google Scholar]
  • 66.Heiden A, Kettenbach J, Fischer P, et al. White matter hyperintensities and chronicity of depression. J Psychiatr Res. 2005 May;39(3):285–293. doi: 10.1016/j.jpsychires.2004.07.004. [DOI] [PubMed] [Google Scholar]
  • 67.Taylor WD, Steffens DC, MacFall JR, et al. White matter hyperintensity progression and late-life depression outcomes. Arch Gen Psychiatry. 2003 Nov;60(11):1090–1096. doi: 10.1001/archpsyc.60.11.1090. [DOI] [PubMed] [Google Scholar]
  • 68.O’Brien J, Ames D, Chiu E, et al. Severe deep white matter lesions and outcome in elderly patients with major depressive disorder: follow up study. BMJ. 1998 Oct 10;317(7164):982–984. doi: 10.1136/bmj.317.7164.982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sheline YI, Pieper CF, Barch DM, et al. Support for the vascular depression hypothesis in late-life depression: results of a 2-site, prospective, antidepressant treatment trial. Arch Gen Psychiatry. Mar;67(3):277–285. doi: 10.1001/archgenpsychiatry.2009.204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gunning-Dixon FM, Walton M, Cheng J, et al. MRI signal hyperintensities and treatment remission of geriatric depression. J Affect Disord. May 6; doi: 10.1016/j.jad.2010.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Taylor WD, Steffens DC, Krishnan KR. Psychiatric disease in the twenty-first century: The case for subcortical ischemic depression. Biol Psychiatry. 2006 Dec 15;60(12):1299–1303. doi: 10.1016/j.biopsych.2006.05.028. [DOI] [PubMed] [Google Scholar]
  • 72.Sneed JR, Rindskopf D, Steffens DC, et al. 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]
  • 73.Taylor WD, Payne ME, Krishnan KR, et al. Evidence of white matter tract disruption in MRI hyperintensities. Biol Psychiatry. 2001 Aug 1;50(3):179–183. doi: 10.1016/s0006-3223(01)01160-x. [DOI] [PubMed] [Google Scholar]
  • 74.Taylor WD, MacFall JR, Payne ME, et al. Late-life depression and microstructural abnormalities in dorsolateral prefrontal cortex white matter. Am J Psychiatry. 2004 Jul;161(7):1293–1296. doi: 10.1176/appi.ajp.161.7.1293. [DOI] [PubMed] [Google Scholar]
  • 75.Bae JN, MacFall JR, Krishnan KR, et al. Dorsolateral prefrontal cortex and anterior cingulate cortex white matter alterations in late-life depression. Biol Psychiatry. 2006 Dec 15;60(12):1356–1363. doi: 10.1016/j.biopsych.2006.03.052. [DOI] [PubMed] [Google Scholar]
  • 76.Yang Q, Huang X, Hong N, et al. White matter microstructural abnormalities in late-life depression. Int Psychogeriatr. 2007 Aug;19(4):757–766. doi: 10.1017/S1041610207004875. [DOI] [PubMed] [Google Scholar]
  • 77.Yuan Y, Zhang Z, Bai F, et al. White matter integrity of the whole brain is disrupted in first-episode remitted geriatric depression. Neuroreport. 2007 Nov 19;18(17):1845–1849. doi: 10.1097/WNR.0b013e3282f1939f. [DOI] [PubMed] [Google Scholar]
  • 78.Shimony JS, Sheline YI, D’Angelo G, et al. Diffuse microstructural abnormalities of normal-appearing white matter in late life depression: a diffusion tensor imaging study. Biol Psychiatry. 2009 Aug 1;66(3):245–252. doi: 10.1016/j.biopsych.2009.02.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Murphy CF, Gunning-Dixon FM, Hoptman MJ, et al. White-matter integrity predicts stroop performance in patients with geriatric depression. Biol Psychiatry. 2007 Apr 15;61(8):1007–1010. doi: 10.1016/j.biopsych.2006.07.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Nobuhara K, Okugawa G, Sugimoto T, et al. Frontal white matter anisotropy and symptom severity of late-life depression: a magnetic resonance diffusion tensor imaging study. J Neurol Neurosurg Psychiatry. 2006 Jan;77(1):120–122. doi: 10.1136/jnnp.2004.055129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Alexopoulos GS, Kiosses DN, Choi SJ, et al. Frontal white matter microstructure and treatment response of late-life depression: a preliminary study. Am J Psychiatry. 2002 Nov;159(11):1929–1932. doi: 10.1176/appi.ajp.159.11.1929. [DOI] [PubMed] [Google Scholar]
  • 82.Taylor WD, Kuchibhatla M, Payne ME, et al. Frontal white matter anisotropy and antidepressant remission in late-life depression. PLoS One. 2008;3(9):e3267. doi: 10.1371/journal.pone.0003267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Nobuhara K, Okugawa G, Minami T, et al. Effects of electroconvulsive therapy on frontal white matter in late-life depression: a diffusion tensor imaging study. Neuropsychobiology. 2004;50(1):48–53. doi: 10.1159/000077941. [DOI] [PubMed] [Google Scholar]
  • 84.Kumar A, Gupta RC, Albert Thomas M, et al. Biophysical changes in normal-appearing white matter and subcortical nuclei in late-life major depression detected using magnetization transfer. Psychiatry Res. 2004 Feb 15;130(2):131–140. doi: 10.1016/j.pscychresns.2003.12.002. [DOI] [PubMed] [Google Scholar]
  • 85.Henkelman RM, Stanisz GJ, Graham SJ. Magnetization transfer in MRI: a review. NMR Biomed. 2001 Apr;14(2):57–64. doi: 10.1002/nbm.683. [DOI] [PubMed] [Google Scholar]
  • 86.Gunning-Dixon FM, Hoptman MJ, Lim KO, et al. Macromolecular White Matter Abnormalities in Geriatric Depression: A Magnetization Transfer Imaging Study. Am J Geriatr Psychiatry. 2008 Mar 10; doi: 10.1097/JGP.0b013e3181602a66. [DOI] [PubMed] [Google Scholar]
  • 87.Dager SR, Corrigan NM, Richards TL, et al. Research applications of magnetic resonance spectroscopy to investigate psychiatric disorders. Top Magn Reson Imaging. 2008 Apr;19(2):81–96. doi: 10.1097/RMR.0b013e318181e0be. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kumar A, Thomas A, Lavretsky H, et al. Frontal white matter biochemical abnormalities in late-life major depression detected with proton magnetic resonance spectroscopy. Am J Psychiatry. 2002 Apr;159(4):630–636. doi: 10.1176/appi.ajp.159.4.630. [DOI] [PubMed] [Google Scholar]
  • 89.Elderkin-Thompson V, Thomas MA, Binesh N, et al. Brain metabolites and cognitive function among older depressed and healthy individuals using 2D MR spectroscopy. Neuropsychopharmacology. 2004 Dec;29(12):2251–2257. doi: 10.1038/sj.npp.1300553. [DOI] [PubMed] [Google Scholar]
  • 90.Chen CS, Chiang IC, Li CW, et al. Proton magnetic resonance spectroscopy of late-life major depressive disorder. Psychiatry Res. 2009 Jun 30;172(3):210–214. doi: 10.1016/j.pscychresns.2009.01.003. [DOI] [PubMed] [Google Scholar]
  • 91.Venkatraman TN, Krishnan RR, Steffens DC, et al. Biochemical abnormalities of the medial temporal lobe and medial prefrontal cortex in late-life depression. Psychiatry Res. 2009 Apr 30;172(1):49–54. doi: 10.1016/j.pscychresns.2008.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Huang Y, Chen W, Li Y, et al. Effects of antidepressant treatment on N-acetyl aspartate and choline levels in the hippocampus and thalami of post-stroke depression patients: a study using (1)H magnetic resonance spectroscopy. Psychiatry Res. Apr 30;182(1):48–52. doi: 10.1016/j.pscychresns.2009.11.009. [DOI] [PubMed] [Google Scholar]
  • 93.Savitz JB, Drevets WC. Imaging phenotypes of major depressive disorder: genetic correlates. Neuroscience. 2009 Nov 24;164(1):300–330. doi: 10.1016/j.neuroscience.2009.03.082. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES