Bio Factor Affecting Mental Health

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Translational Psychiatry www.nature.

com/tp

EXPERT REVIEW OPEN

Biological factors influencing depression in later life: role of


aging processes and treatment implications
1,4 ✉
Sarah M. Szymkowicz1,5, Andrew R. Gerlach2,5, Damek Homiack3 and Warren D. Taylor

This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply 2023

Late-life depression occurring in older adults is common, recurrent, and malignant. It is characterized by affective symptoms, but
also cognitive decline, medical comorbidity, and physical disability. This behavioral and cognitive presentation results from altered
function of discrete functional brain networks and circuits. A wide range of factors across the lifespan contributes to fragility and
vulnerability of those networks to dysfunction. In many cases, these factors occur earlier in life and contribute to adolescent or
earlier adulthood depressive episodes, where the onset was related to adverse childhood events, maladaptive personality traits,
reproductive events, or other factors. Other individuals exhibit a later-life onset characterized by medical comorbidity, pro-
inflammatory processes, cerebrovascular disease, or developing neurodegenerative processes. These later-life processes may not
only lead to vulnerability to the affective symptoms, but also contribute to the comorbid cognitive and physical symptoms.
Importantly, repeated depressive episodes themselves may accelerate the aging process by shifting allostatic processes to
1234567890();,:

dysfunctional states and increasing allostatic load through the hypothalamic–pituitary–adrenal axis and inflammatory processes.
Over time, this may accelerate the path of biological aging, leading to greater brain atrophy, cognitive decline, and the
development of physical decline and frailty. It is unclear whether successful treatment of depression and avoidance of recurrent
episodes would shift biological aging processes back towards a more normative trajectory. However, current antidepressant
treatments exhibit good efficacy for older adults, including pharmacotherapy, neuromodulation, and psychotherapy, with recent
work in these areas providing new guidance on optimal treatment approaches. Moreover, there is a host of nonpharmacological
treatment approaches being examined that take advantage of resiliency factors and decrease vulnerability to depression. Thus,
while late-life depression is a recurrent yet highly heterogeneous disorder, better phenotypic characterization provides
opportunities to better utilize a range of nonspecific and targeted interventions that can promote recovery, resilience, and
maintenance of remission.

Translational Psychiatry (2023)13:160 ; https://doi.org/10.1038/s41398-023-02464-9

INTRODUCTION typically persist, and older depressed adults have an increased risk of
Late-life depression (LLD) is major depressive disorder (MDD) dementia [14].
occurring in adults age 60 years or older [1]. It is common, with Such adverse outcomes may be related to LLD’s recurrent nature
~5% of community-dwelling elders meeting DSM5 diagnostic [15]. LLD is often a recurrent or chronic illness [16], although
criteria [2] and 10–16% of older adults exhibiting clinically significant continuing antidepressant medication during remission reduces
depressive symptoms that may not meet full criteria [2, 3]. LLD is a recurrence risk [17, 18]. However, even with maintenance treatment,
malignant illness that increases disability [4], contributes to poorer ~35–40% of depressed elders experience recurrence in 2 years, with
medical outcomes [5], and is associated with increased suicide risk over 50% experiencing recurrence over four years [16, 18, 19].
and mortality [6, 7]. Recurrence is particularly relevant for individuals with an initial
LLD is further characterized by poor or impaired cognitive onset of depression in early- or midlife. These individuals often
performance. Reduced executive functioning is common, affecting experience multiple prior depressive episodes and are now in the
verbal fluency, response inhibition, set-shifting, working memory, geriatric age range. Individuals with early-onset LLD, typically
and problem-solving [8]. Individuals with LLD also exhibit poor defined as occurring before age 50–60 years, exhibit greater
performance in other cognitive domains, including episodic residual depression severity over time, more frequent suicidal
memory, visuospatial skills, and processing speed [9–11]. Slower thoughts, and a greater risk of recurrence following remission
processing speed is particularly important, partly mediating [16, 20]. Early-onset depression is characterized by stronger familial
impaired performance in other cognitive domains [11–13]. Although history and genetic loading, greater anxiety and reactions to
cognitive performance improves with successful treatment, deficits stressful life events, maladaptive personality traits, and hormonal

1
Center for Cognitive Medicine, Department of Psychiatry and Behavioral Science, Vanderbilt University Medical Center, Nashville, TN, USA. 2Department of Psychiatry, University
of Pittsburgh, Pittsburgh, PA, USA. 3Department of Psychiatry, University of Illinois-Chicago, Chicago, IL, USA. 4Geriatric Research, Education, and Clinical Center, Veterans Affairs
Tennessee Valley Health System, Nashville, TN, USA. 5These authors contributed equally: Sarah M. Szymkowicz, Andrew R. Gerlach. ✉email: [email protected]

Received: 24 November 2022 Revised: 23 April 2023 Accepted: 27 April 2023


S.M. Szymkowicz et al.
2

Fig. 1 Lifespan model of late-life depression. Symptoms of depression are the behavioral manifestation of increasingly disrupted brain
networks. Multiple influences contribute to network fragility and dysfunction across the lifespan, with some being linked to clear
developmental periods. These may be additive and cumulative over time, although other risk and resiliency factors may be modifiable and
targeted by specific treatments. Unchecked, repeated depressive episodes and their associated physiological responses may have deleterious
effects contributing to accelerated aging.

fluctuations associated with early-life reproductive events [21–26]. additional contributors [35, 36], although this has received less
Individuals with late-onset depression are more often widowed, attention in LLD [37]. These networks likely influence depressive
present with more apathy and somatic symptoms, poorer cognitive behavior across the lifespan, although the etiological factors
performance, greater cognitive decline and medical morbidity, and contributing to network dysfunction change with aging. Although
more severe atrophic and vascular changes on neuroimaging heterogeneity in LLD prevents sweeping generalizations [38],
[26–32]. Although useful for clinical characterization, this age of there is support for this network-centric model [34, 39].
onset dichotomization obfuscates potentially important differences The DMN is implicated in self-referential processes [40]
in causal factors that influence the onset or recurrence of including rumination [41], making it a target of investigation
depression across the lifespan. Moreover, it does not address a in depression [33, 42]. However, there is little consensus on how
parallel hypothesis that depression itself is toxic, with recurrent the DMN is altered in depression [43]. Early studies reported
episodes increasing the allostatic load or “wear and tear” on the hyperconnectivity within the DMN relative to healthy controls
body, contributing to accelerated brain aging and vulnerability to [42], though recent meta-analyses reported no difference in
poor longitudinal clinical, cognitive, and medical outcomes [15]. DMN connectivity [44] or even hypoconnectivity between DMN
Based on past work [15], we present a model (Fig. 1) where regions [45]. DMN functional connectivity appears to be altered
disruption of functional brain network homeostasis contributes in LLD [46, 47] and such differences may persist into
first to subclinical depressive symptoms and decreased stress remission [46].
tolerance. If unchecked, this progresses to discrete depressive The CCN is primarily involved in top-down executive functions
episodes and reduced cognitive performance. Various biological [48]. Substantial evidence suggests CCN integrity differentiates
and environmental factors across the lifespan increase the healthy controls from depressed individuals [42] and influences
vulnerability of key networks to disequilibrium, with potentially treatment response [49]. The CCN may be especially relevant in
modifiable behavioral and social factors contributing either to LLD characterized by executive dysfunction [50, 51], where
vulnerability or resilience. In turn, depressive episodes alter aberrant functional connectivity of the CCN (particularly the
physiological systems that accelerate aging processes and dorsolateral prefrontal cortex [DLPFC] hub), is associated with
contribute to adverse longer-term outcomes. executive deficits [52].
Building on this model, we first present a network-based model The ASN is implicated in switching and control of attentional
of depression. We then focus on etiological influences across the processes [53], and ASN dysfunction in depression biases
lifespan that increase depression risk, primarily focusing on those individuals towards negative stimuli and processing [54, 55].
salient to later life. Next, we consider depression as a contributor Unlike the DMN, the ASN has proven relatively reliable in
to accelerated aging. Finally, we review treatment for LLD and distinguishing between healthy controls and depressed indivi-
how interventions may support resilience to future episodes. duals [42, 44, 56]. Both structural and functional connectivity of
the ASN are reduced in LLD [57].
Alterations in the positive valence system are additive.
NEURAL NETWORKS IMPLICATED IN DEPRESSION Conceptually, much work focuses on the dopaminergic mesolim-
Altered neural network function is thought to result in the bic pathway, projecting from the ventral tegmental area to the
behavioral manifestations of depression [33]. The triple network ventral striatum, nucleus accumbens, and medial temporal
model (Fig. 2) [33, 34] posits that depression is related to the structures [37]. Dysfunction in this system influences a range of
aberrant function of the default mode network (DMN), cognitive reward functions including valuation, decision-making, effort, and
control network (CCN), and anterior salience network (ASN). learning [37, 58]. Behaviorally, this contributes to anhedonia,
Positive valence system circuits involved in reward function are motivational disturbances, and willingness to expend effort [37].

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
3

Fig. 2 Network model of late-life depression. The model details findings within each intrinsic functional network and between functional
networks, specifically the default mode network (DMN), the cognitive control network (CCN), and the anterior salience network (ASN).
Disruption in network connectivity influences the cognitive processes, giving risk to the behavioral manifestations of depression. Impaired
function of the positive valence system involving the mesolimbic system likely also contributes to depressive behavior [37], although how this
system interacts with interacts with intrinsic functional networks in LLD is not entirely clear. The model and figure [34] used with permission.
Reprinted from Gunning et al. [34], Copyright 2021, with permission from Elsevier.

Neural networks and aging In order to influence depression risk, such genetic factors would
The connectivity and function of these networks change with age. need to directly or indirectly alter brain network function or
Cross-sectional and longitudinal studies demonstrate that frontal stability [76]. However, concerns persist about translating these
regions comprising the associative networks described above (plus findings to the individual level, both due to the contributions of
the dorsal attention network) exhibit reduced intra-network small-effect polymorphisms that may be missed on GWAS, and
connectivity and increased inter-network connectivity with aging due to diagnostic heterogeneity within MDD [77]. What risk
[59–69]. These changes may reflect a decline in network efficiency genes contribute to depression vulnerability may change across
and/or serve as a compensatory mechanism that maintains normal the lifespan, particularly if they affect brain aging. For example,
brain function in context of gray matter loss or white matter some work supports that vascular risk genes may be germane in
degradation [70, 71]. Supporting this latter hypothesis are studies LLD [78].
showing increased activation in frontal regions in older adults Adverse childhood experiences (ACEs), such as abuse, parental
compared to younger adults that are associated with better loss, and bullying, are associated with a host of health disorders,
cognitive performance with aging [72]. Increased prefrontal including depression [79]. They are also associated with differences
activation is further associated with reduced white matter integrity, on neuroimaging and cognitive testing [80]. ACEs may contribute to
again supporting compensation [73, 74]. These age-related network depression vulnerability through hypothalamic–pituitary–adrenal
changes have been replicated using both structural and functional (HPA) axis responses leading to increased activity of corticotropic
network measures, further associating aging with lower strength releasing factor neurons [81]. ACEs can further result in epigenetic
and density of structural connections and decoupling of structural changes [24] that may increase depression vulnerability by
and functional connectivity, particularly in network hubs [63]. This influencing glucocorticoid signaling, serotonergic function, and
may represent a rerouting of information flow in the brain intended neurotrophic factors [24]. The relationship between ACEs and
to circumnavigate degraded white matter pathways or avoid depression vulnerability persists into later life [82, 83], where the
regions that have suffered neuronal loss. The ability of the brain to relationship between ACEs and depression may be mediated by
successfully “rewire” during aging may be crucial for maintaining inflammation [82]. Stressful events occurring in adulthood or later
cognitive performance and reflect cognitive reserve [70]. Moreover, life also increase the risk for new depressive episodes and
age-related changes may contribute to network fragility, increasing depression persistence [84, 85].
risk for LLD. While there is some evidence that network organization Personality traits are similarly associated with depression. Traits
properties may differ according to age of onset [57], clear that influence how individuals interact with and respond to their
differences in neural network configuration between early- and environments originate from variability in functional brain networks
late-onset LLD have not been identified. [22]. They arise from a complex interplay between brain develop-
ment, genetic predisposition, and early environmental exposures.
Neuroticism, a predisposition to experience psychological distress
FACTORS CONTRIBUTING TO DEPRESSION VULNERABILITY and negative mood states, is well-studied and shares some
Early- and midlife risk factors conceptual overlap with LLD [86]. Higher levels of neuroticism in
Older depressed adults carry the same vulnerabilities that LLD are associated with poorer antidepressant response and greater
increased risk for depression earlier in life. As the list of potential risk of cognitive decline [86–88].
contributors to MDD risk is beyond this review, we focus on Reproductive events including puberty, menstrual cycling,
mechanisms of relevance to LLD. pregnancy, and menopause are associated with both new-onset
Genetic factors that influence MDD vulnerability likely persist and recurrent depression [23]. These events contribute to a higher
with aging. Genome-wide association studies (GWAS) identified risk of depression for women than men [89], particularly during
several hundred potential genetic risk variants, including genes reproductive years [90]. These relationships are due to fluctuations
involved in synaptic structure and neurotransmission [75]. of ovarian hormones that influence neurotransmitter function,

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
4
Table 1. Support for etiological hypotheses of late-life depression.
Inflammation Vascular Neurodegeneration
Clinical • Neurovegetative symptoms (lethargy, • Dysphoria, anhedonia, apathy, • Apathy, subjective memory loss
reduced appetite) psychomotor retardation, • AD pathology or development of
• Greater medical morbidity functional disability dementia associated with poor
• Increased levels of pro-inflammatory • Higher rates of vascular risk factors antidepressant efficacy
cytokines; Decreased levels of anti- • Increased disability and mortality
inflammatory cytokines
• Associated with treatment resistance and
poor antidepressant efficacy
Cognitive • Executive dysfunction, slowed processing • Executive dysfunction, reduced • Depression co-occurring with
and motor speed, reduced memory processing speed and visuospatial dementia worsens cognitive
skills, retrieval-based memory performance
deficits • Amnestic cognitive profile (often, but
not always)
Imaging • Peripheral inflammatory markers linked • WMH volumes increase over time • LLD with greater beta-amyloid
with: • Higher WMH volumes worsen deposition have:
• altered fronto-subcortical activation cognitive outcomes • greater temporal lobe volume loss
• gray and white matter volume loss • Persistent depressive symptoms • lower functional connectivity in
• Higher markers of central inflammation exhibit greater change in WMH fronto-subcortical regions
found in anterior cingulate and temporal volume over time • functional DMN alterations
cortices
Negative • When excluding comorbid medical • Inconsistent findings between LLD, • Some report less beta-amyloid
findings conditions, studies do not show vascular burden, and deposition in LLD compared to
relationship between depression and antidepressant response. controls
inflammatory cytokines • APOE ε4 does not clearly influence
development of LLD
AD Alzheimer’s disease, APOE apolipoprotein E, CSF cerebrospinal fluid, DMN default mode network, LLD late-life depression, WMH white matter
hyperintensities.
Table inspired by and adapted from Alexopoulos [164].

neuroendocrine processes, and inflammation [91]. Menopause is associated with depression severity, suicide risk, and poor treatment
particularly relevant to aging, with decreased estrogen production response in adult and geriatric samples [105–107]. Pro-inflammatory
potentially reducing its neuroprotective effects [92]. A longer cytokines are associated with worse function in executive processes,
exposure to endogenous estrogens, operationalized as an older memory, and processing and motor speed [108].
age at menopause, is associated with a lower risk of subsequent Aging is itself associated with chronic, low-grade inflammation,
depression [93], while earlier menopause, including surgically- dubbed “inflammaging” [109]. This process has multiple contribu-
induced menopause, is associated with cognitive decline and tors, including immune system aging, mitochondrial changes, and
dementia [94]. gut microbiota [110, 111]. These changes may be secondary to
Other exposures influence MDD, including comorbid mental common pro-inflammatory medical conditions that increase depres-
health disorders, substance misuse, and traumatic brain injury sion risk, including diabetes, cardiovascular disease, autoimmune
[95]. Medical comorbidity also contributes, including a bidirec- disorders, rheumatoid arthritis, cirrhosis, and kidney disease
tional relationship between depression and obesity [96] that [112–115]. These comorbidities may explain the observed relation-
may be mediated through immune system activation and ship between depression and inflammation, as evidenced by a study
inflammation [96]. Obesity increases the risk for other morbid- that did not associate LLD with higher levels of central or peripheral
ities associated with LLD, including pain syndromes, vascular risk inflammatory cytokines. However, this study employed rigorous
factors, and disability [97, 98]. entry criteria for medical comorbidities that excluded almost 95% of
potentially eligible participants [116]. This raises issues about its
Later-life risk factors generalizability and highlights the extent of comorbidity between
Despite LLD being associated with a range of medical comorbid- LLD and medical illnesses.
ities, few may directly contribute to depression pathogenesis. Age- Although most work examines peripheral inflammatory markers,
related morbidities that are a focus of mechanistic models include it remains relevant to brain function. The CNS was long considered
inflammation, vascular disease, and neurodegeneration (Table 1). to be immunoprotected due to the blood-brain barrier. However,
immune responses via peripheral immune cell secretion of pro-
Inflammation. The inflammation hypothesis proposes that inflammatory cytokines can convey the inflammatory response to
immune dysregulation influences vulnerability to and the develop- brain microglia via humoral and neural pathways [117, 118].
ment of LLD [99]. Neurovegetative depressive symptoms are akin to Microglia can thus become activated by peripheral cytokines
immune responses to infectious diseases including lethargy, inducing a neuroinflammatory response [119]. Both aging and
cognitive slowing, and reduced appetite [100, 101]. In younger psychological stress further prime microglia toward an activated
adults, elevated pro-inflammatory cytokines levels in response to state, tilting the CNS toward a pro-inflammatory state [120, 121]. In
psychological stress are associated with depressive symptoms [101] the aged brain, activated microglia exhibit an exaggerated response
and induction of peripheral inflammation results in fatigue and to pro-inflammatory cytokines, inducing oxidative stress and
worsening of mood [102, 103]. Depressed patients across the adult delayed clearance of neurotoxic molecules, resulting in disrupted
lifespan can exhibit elevated levels of pro-inflammatory cytokines neuronal function, impaired neurogenesis, and neural degeneration
including c-reactive protein (CRP), interleukin-6 (IL-6) and tumor [119, 120]. Central inflammation further affects multiple neurotrans-
necrosis factor (TNF) alpha and lower anti-inflammatory cytokine mitter systems, contributing to reduced serotonin synthesis via
levels [104, 105]. Higher pro-inflammatory cytokine levels are induction of indoleamine 2,3-dioxygenase [100], glutamate system

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
5
dysregulation [122–124], and altered dopamine synthesis, binding, longitudinal decline in executive functions and increased risk for
and reuptake [37, 125–128]. dementia [162]. Individuals with persistent depressive symptoms
While less examined in LLD, chronic inflammation induces an similarly exhibit greater increases in WMH volume over time
altered cellular environment in the brain parenchyma capable of [163]. Greater vascular burden may be associated with poorer
modulating neural circuits and influencing depressive behavior response to pharmacological and nonpharmacological treat-
[15]. Pro-inflammatory cytokines including CRP and IL-6 are ments [158, 164–166], although this relationship for medication
associated with global gray matter and white matter loss [129]. A response is somewhat weak [49, 78]. The higher vascular burden
meta-analysis including participants across the lifespan asso- is further associated with greater disability [167], gait and other
ciated peripheral inflammatory markers with altered activation motor deficits [168], and frailty [169]. Depression can also
of the prefrontal cortex, insula, striatum, amygdala, hippocam- worsen cardiovascular and cerebrovascular disease outcomes
pus, and various subcortical regions [130]. These regions overlap [170, 171], suggesting a bidirectional relationship.
with the DMN, ASN, limbic, and corticostriatal networks. Studies
of individuals with depression and suicide attempts report Neurodegeneration. Aging is the strongest risk factor for demen-
increased microglial activation in the anterior cingulate cortex tia [172], a collective term for cognitive impairment negatively
(ACC), a key hub of both the ASN and CCN, in individuals with affecting independent functional activities. Alzheimer’s disease
depression and suicide attempts [131, 132]. Experimental (AD), the most common dementia, is characterized by abnormal
induction of acute inflammation similarly alters glutamate accumulation of beta-amyloid plaques and tau tangles in the
metabolism in the ACC and basal ganglia [124]. brain. Early AD typically affects memory centers, including the
Treatment implications of pro-inflammatory processes are entorhinal cortex and hippocampus. With disease progression,
unclear. Work in midlife suggests that low-grade inflammation neuropathology spreads to frontal and parietal regions and affects
may decrease antidepressant efficacy [133–136]. However, success- language, executive abilities, and social behaviors.
ful antidepressant treatment can decrease pro-inflammatory Depression and dementia exhibit a bidirectional relationship.
cytokine levels [137, 138] Conversely, antidepressant augmentation Depression in mid-to-late life increases risk for AD and all-cause
with anti-inflammatory agents may reduce depressive symptom dementia [14, 173, 174]. Depression can also be a precursor to or
severity and improve treatment outcomes [139, 140]. If existing symptom of dementia, with prevalence rates ranging from 17 to
trials are supported, such interventions may most benefit indivi- 56% across all stages of AD [175]. Depression co-occurring with
duals with higher inflammatory cytokine levels, such has been seen dementia worsens cognitive performance beyond what would be
with infliximab, a TNF-alpha antagonist [141, 142]. Inflammation expected based on neuropathology alone [176]. Dementia risk
could identify a distinct phenotype [37, 143] who would benefit may be highest in individuals exhibiting persistent or worsening
from anti-inflammatory treatments. depressive symptoms over time [177].
Such observations led to work searching for common genetic
Vascular disease. Cerebrovascular system changes are common factors. While the apolipoprotein E (APOE) ε4 allele significantly
with normal aging [144]. Cerebral small vessel disease (CSVD) increases risk for AD, it does not clearly influence the development
describes leakage of the blood-brain barrier and dysfunction of of LLD [178, 179]. A genome-wide association study found that
cerebral autoregulation, neurovascular coupling, and capillary depression had a causal role in AD through Mendelian randomiza-
blood flow [145]. This causes cerebral perfusion deficits and tion, but there was no evidence for a causal role of AD on
hypoxia, triggering inflammation and neuronal death. Vascular risk depression [180]. That study identified 53 brain transcripts and
factors including hypertension, atherosclerosis, and diabetes proteins regulated by the depression GWAS signals that also were
contribute to CSVD and result in the thickening of the penetrating associated with rates of cognitive decline over time [180].
small arteries, fibrosis of the vessel wall, and depletion of vascular The “amyloid hypothesis of LLD” [181] is supported by
smooth muscle cells. observations of increased beta-amyloid deposition in older adults
The “vascular depression hypothesis” [146, 147] posits that with a depression history [182] and in LLD patients exhibiting a
CSVD may predispose, precipitate, or perpetuate LLD. This cognitive profile suggestive of amnestic Mild Cognitive Impair-
process likely begins in adulthood, as midlife cerebrovascular ment [183]. Individuals with LLD exhibiting greater beta-amyloid
burden predicts increased depression severity over time [148]. deposition show greater volume loss in the temporal lobe, lower
The neuroradiological manifestation of “vascular depression” functional connectivity in fronto-subcortical regions, and greater
includes white matter hyperintensities (WMHs) on T2-weighted functional alterations in the DMN [184, 185]. These findings are
MRI, subcortical lacunes, and microbleeds [147, 149]. Mechan- not universal, and the Alzheimer’s Disease Neuroimaging Initiative
istically, WMHs may contribute to a disconnection syndrome depression group reported less beta-amyloid deposition in LLD
where damage to communicating cortical-subcortical pathways compared to a control group [186]. While beta-amyloid deposition
involved in mood regulation and cognitive processes increases was associated with worse memory performance in that study, the
LLD vulnerability [78, 150, 151]. Further supporting a mechan- association between amyloid and cognitive performance did not
istic role, meta-analyses have shown that late-onset depression differ between diagnostic groups. More recent work has focused
show significantly greater WMH burden in late-onset LLD than in on tau pathology as others report that individuals with elevated
early-onset LLD [152, 153]. The extent of ischemic injury extends tau, but not amyloid, are twice as likely to be depressed [187].
beyond visible WMHs, as vascular risk factors compromise Poorer cognitive performance, comorbid dementia, and AD
microstructural integrity in normal-appearing white matter pathology are associated with poorer prognosis for response to
[154, 155]. Location of WMHs and microstructural changes may antidepressant medications. Both poorer cognitive performance,
be critical, as LLD is associated with damage to the cingulum particularly executive dysfunction or slowed processing speed,
bundle, uncinate fasciculus, and superior longitudinal fasciculus and dementia are associated with poorer responses to antide-
[156–159]. pressant medications [12, 165, 188, 189]. Similarly, higher levels of
These processes influence longer-term negative outcomes. beta-amyloid deposition, particularly in the temporal lobe, are
Even without depression, vascular changes are associated with associated with poor response or treatment resistance to
cognitive deficits (including executive dysfunction and retrieval- antidepressant medications, even in cognitively intact elders
based memory deficits) and altered emotion processing [160]. [190, 191]. Alternative treatment approaches are not entirely clear,
Cross-sectionally, depressive symptoms with greater WMH although some individuals with cognitive impairment may benefit
volumes worsen cognitive outcomes in the early stages of CSVD from nonpharmacological interventions such as computerized
[161]. Greater WMH volume in LLD contributes to greater cognitive training [192].

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
6
Depression is not unique to a single neuropathological process difference between calculated brain age and chronological age was
[176]. Beyond AD, it occurs in context of alpha-synuclein, a independently replicated [205] and not associated with age of
constituent of Lewy bodies and the pathological hallmark of depression onset, recurrence, or remission [204].
synucleinopathies, including Parkinson’s disease (PD), dementia An accelerated aging hypothesis of LLD implies a bidirectional
with Lewy bodies, and multiple system atrophy. Depression is a process [206]. Depressed older adults exhibit an advanced
common non-motor symptom of PD [193] and depressed biological age on a multibiomarker index of metabolic and
individuals exhibit a 2.2-fold increase in risk of subsequent inflammatory measures [207, 208] and on structural MRI, where
parkinsonism [194]. There is a positive association between they appear approximately 4 years older than nondepressed
alpha-synuclein messenger RNA expression levels and depression individuals [198]. This accelerated brain aging is further associated
severity [195], while levels of CSF alpha-synuclein may mediate with disability and cognitive performance [198], with depression
associations between LLD, markers of synaptic dysfunction, and severity moderating the relationship between brain age and some
memory ability [196]. cognitive measures. As the difference between calculated brain age
and chronological age differs between midlife adult depressed
samples and LLD [198, 203, 204, 209], depression may be associated
DEPRESSION AND ACCELERATED AGING with an altered trajectory of biological aging [210].
Both vascular [170, 171] and neurodegenerative processes Accelerated aging also influences physical function and
[14, 173, 174] occur more frequently or more severely in LLD. contributes to physical frailty. Frailty is characterized by deficits
These may represent bidirectional relationships, where depressive in strength and mobility, decreased physical activity, and reduced
episodes may both contribute to and result from accelerated energy capacity that results from dysregulation in metabolic,
aging. Biological aging is an inevitable process at molecular, musculoskeletal, and stress-response systems [211, 212]. Frailty is
cellular, and organ levels reducing a system’s reparative or common, bidirectionally associated with LLD, and associated with
regenerative potential [197]. “Accelerated aging” is when biologi- increased mortality and poor antidepressant treatment responses
cal aging occurs more rapidly than expected, resulting in [213–215]. Frailty may be an outcome of depression, as depression
biological characteristics that are more severe than would be is associated with worsening trajectories in functional status,
expected based on chronological age [198]. In the brain, this may including reduced walking speed and hand strength [216].
include ventricular enlargement, cerebrovascular injury, or gray The model of a bidirectional relationship between depression
matter atrophy. For this review, we distinguish accelerated aging and accelerated and pathological aging (Fig. 3) may start with age-
from “pathological brain aging”, characterized by neurodegenera- related changes increasing vulnerability to depression. Individuals
tive processes involving amyloid, tau, or other abnormal protein experiencing accelerated biological aging, operationalized as
deposition. advanced medical morbidity, cerebrovascular pathology, pro-
Accelerated aging is observed in multiple neuropsychiatric inflammatory processes, or pathological aging, such as increasing
disorders and quantified using a range of markers including amyloid or tau burden, are at higher risk for LLD. Such etiological
telomere length, oxidative stress markers, epigenetic markers, factors can disrupt the structure, function, and homeostasis of key
physiological functioning, and neuroimaging [199–201]. In adult intrinsic functional networks implicated in depression [78, 164].
MDD, accelerated aging is observed on molecular and cellular These etiological factors may all occur to varying extents in the
markers, including reduced telomere length, epigenetic aging, and same individual, each challenging functional network home-
metabolomic aging [201, 202]. Accelerated aging in MDD is further ostasis. As the underlying systems become more dysregulated or
observed on both structural and functional neuroimaging, where as pathologies progress, and networks experience greater
depressed individuals appear on average 1–2 years older than homeostatic imbalance and impairment in function, the clinical
nondepressed cohorts [203, 204]. In the largest of these studies, the presentation of the depressive syndrome emerges (Fig. 1) [15]. It is

Fig. 3 Accelerated aging hypothesis of late-life depression. Aging processes such as inflammation, vascular disease, or pathological
neurodegeneration impair neurotrophic function and contribute to both gray matter atrophy and impairment of white matter microstructure.
These changes in turn alter function of key intrinsic networks, leading to the clinical manifestations of late-life depression. In turn, repeated
depressive episodes result in altered or sustained physiological responses increasing allostatic load. These effects may then further accelerate
biological aging processes, shifting an individual further away from normative aging.

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
7
possible that the predominant underlying pathology and what resulted in a remission rate of 12.8% for older individuals, which is
brain networks are most affected influence the clinical presenta- comparable to remission rates seen in patients progressing to
tion and phenotype [37, 217]. later stages of the STAR*D study [245, 246]. A randomized trial of
Depressive episodes may also accelerate biological aging esketamine in treatment-resistant LLD resulted in a comparable
(Fig. 2). One potential mechanism involves the altered neural remission rate of 17.3%, with a NNT of 10, although that study did
and physiological responses to stress observed in depression. not detect a statistically significant difference in their primary
Such altered responses are observed across a range of systems, endpoint [247]. Secondary analyses suggested that participants
including altered function of brain regions involved in emotion with an earlier life onset of depression or who were less than 75
processing, autonomic reactivity, HPA axis function, and dysre- years of age exhibited greater improvement [247]. Both ketamine
gulation of the immune system or circadian processes [78, and esketamine were well-tolerated, with common side effects
99, 218–224]. Normally, these processes are meant to facilitate including dizziness, dissociative symptoms, fatigue, and transiently
allostasis, the body’s ability to respond to environmental elevated blood pressure [245, 247].
challenges and maintain normal functioning. However, with
repeated stressors and depressive episodes, these responses Electroconvulsive therapy (ECT)
contribute to increased allostatic load, the wear-and-tear result- ECT continues to be used for severe and treatment-resistant LLD.
ing from stress and the dysregulation of processes meant to In LLD, ECT exhibits remission rates between 70 and 90%,
maintain homeostasis [15, 225]. Such canonical stress-response although rates in community samples may be lower [248].
systems interact and over time contribute to accelerated aging, Individuals with late-onset depression tend to respond better to
leading to regional brain atrophy, development of cerebrovas- ECT than individuals with early-life depression onset [249], which
cular pathology, and reduction of neurotrophic function may be related to illness chronicity or recurrence in the early-
[226–228]. This hypothesis is supported by longitudinal studies onset group. However, strong remission rates should be balanced
associating persistent or recurrent depression with greater by high relapse rates after the initial ECT course, with 40–50% of
increases in WMH volume and greater hippocampal volume patients relapsing within 6 months [250]. Cognitive side effects in
decline [163, 229]. Other mechanisms explaining this relationship older adults tend to be limited and transient [251].
are possible, including shared genetic vulnerabilities [180]. Recent work has refined ECT to improve tolerability while
Such bidirectional relationships have long-term implications. preserving efficacy. This includes administering ECT using right
Accelerated or pathological aging also contribute to impaired unilateral electrode placement with ultrabrief pulse width stimuli,
cognitive performance [198, 230], increased risk for dementia [14], an approach with fewer cognitive side effects [252]. When
and risk of physical disability, sensory function loss, and frailty combined with venlafaxine in LLD, this results in remission rates
[230]. Accelerated brain aging may be associated with higher risk of 61% and response rates of 70% [253]. Unilateral brief pulse ECT
for depression relapse after achieving remission [15]. Even if the combined with venlafaxine only modestly affects cognitive
initial antidepressant treatment were successful, progressive aging performance, specifically letter fluency and cognitive flexibility
may further challenge functional networks and result in a return of [254]. This study also included a 24-week continuation phase,
depressive symptoms [15]. where participants were randomized to either medication only
(venlafaxine plus lithium) or venlafaxine plus continuation ECT,
administered weekly for the first month with additional sessions
UPDATES ON ESTABLISHED SOMATIC TREATMENTS as needed. Continuation ECT resulted in lower levels of depression
Currently, treatment decisions for patients with LLD are guided severity at study endpoint than medication only [255] and better
more by clinical history and patient preference than potential quality of life [256].
biological causal factors. Robust blinded clinical trials data for LLD
are scant and clinical guidelines tend to derive from expert opinion Transcranial magnetic stimulation (TMS)
or are extrapolated from data in younger populations [231]. Repetitive TMS (rTMS) uses a pulsed magnetic field to induce a
local electrical field on the brain’s surface, stimulating cortical
Antidepressant medications pathways. rTMS treatment of depression typically targets the
Antidepressants are more effective than placebo in the treatment DLPFC, with the best-studied techniques including unilateral
of LLD, although the response rate is lower for older than younger high-frequency left-sided (HFL), unilateral low-frequency right-
adults [232–234]. However, while antidepressants alter DMN and sided (LFR), or sequential bilateral treatment of LFR followed by
CCN connectivity [235], age of initial onset does not influence HFL [257]. Parallel work supports that targeting the DLPFC
antidepressant medication response rates, although early-onset modulates functional connectivity within and between the DMN
patients may respond more slowly [236, 237]. Antidepressants and CCN, with clinical benefit deriving from modulation of
remain beneficial, with a number needed to treat (NNT) for an subgenual cingulate cortex connectivity [258]. While many
antidepressant response being 6.7 (95% CI, 4.8–10) [234, 238]. As randomized trials support rTMS efficacy, few have been
in younger adults, augmentation strategies in LLD are more conducted in LLD [259, 260]. However, rTMS is well-tolerated
efficacious than strategies involving a switch to a different in older adults [261], and LLD trials generally support the efficacy
antidepressant [239]. Methylphenidate augmentation of an SSRI of HFL rTMS, with bilateral treatment being more efficacious in
is superior to monotherapy with either agent alone [240]. treatment-resistant patients [257].
Augmentation with lithium, bupropion, or aripiprazole in patients Recent work has modified rTMS to improve outcomes and
who did not respond to monotherapy can be well-tolerated and reduce burden. A sham-controlled trial in LLD that examined
improve depressive symptoms [239, 241, 242]. Despite clear bilateral deep rTMS reported efficacy and good tolerability when
benefits of augmentation, the likelihood of achieving remission administered over four weeks [262]. This deep TMS approach
decreases with increasing number of failed antidepressant trials addressed concerns that age-associated atrophy may contribute
within the current episode [243]. to poor treatment responses by increasing the distance between
Few studies examine outcomes of the N-methyl-D-aspartate the scalp and cortex [263], however it requires longer adminis-
(NMDA) receptor channel inhibitors ketamine and esketamine in tration sessions. More recent work in LLD compared rTMS to theta-
LLD. Both a small, randomized trial of subcutaneously adminis- burst stimulation (TBS), a bilateral approach that reduces session
tered ketamine and larger open-label study of intravenous administration from 47 min for rTMS to 4 min for TBS. This
ketamine improved depression severity in older adults with randomized trial established non-inferiority of TBS, with compar-
treatment-resistant depression [244, 245]. Intravenous ketamine able reductions in depression severity between groups [264].

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
8
Table 2. Resilience factors influencing depressive episode risk in later life.
Domain Factors Resilience correlates Depression vulnerability correlates
Trait-like factors Temperament Positive emotionality Greater harm avoidance
Personality Extroversion, conscientiousness Neuroticism
Psychological factors Beliefs Self-esteem, self-efficacy, mastery, sense of Internalized self-blame or stigma
purpose
Coping Active or accommodative coping Avoidance or passive coping
Social factors Social support Social engagement Social withdrawal, loneliness
Altruism Formal volunteering Social role absences
Cognitive factors Cognitive reserve Maintained cognitive performance Poorer executive function and processing
speed
Physical factors Physical activity Exercise, regularly active Sedentary lifestyle
Sensory function Sustained or corrected vision and hearing Impaired sensory function
Healthy diet Good nutrition Poor nutrition, substance abuse
Healthy sleep Regular sleep patterns Disrupted, irregular sleep
Correlates of factors that may contribute to resilience from depression, or vulnerability to depression. Some factors are modifiable (psychological factors, social
factors, and lifestyle factors) while others are not (trait-like factors). Some depression correlates may both increase the risk of depression and also be an
outcome of depression. Table inspired by and adapted from Laird et al. [267] and Andreescu et al. [15].

RESILIENCE FACTORS: OPPORTUNITIES FOR INTERVENTION analyses in LLD support that psychotherapy is quite effective, with a
Although these treatments are effective, benefit depends on NNT of 3 [278]. More recently developed, “Engage” therapy targets
continued treatment. If pharmacotherapy or neuromodulation neurobiologically-informed processes in LLD using streamlined
stops, the risk of recurrence can be high [18, 250, 253]. This risk behavioral techniques that can be effectively applied in the
may be reduced through interventions that target vulnerability community [279, 280].
factors to depression and strengthen resiliency (Table 2).
Resilience is broadly defined as the capacity to maintain or regain Social factors: opportunities for engagement
psychological well-being despite challenges, or the adaptive Aging adults tend to maintain close social partners but have fewer
maintenance of homeostasis despite adversity [265–267]. Resilience peripheral social contacts [281]. In contrast, larger objective social
is a multidimensional, dynamic process influenced by both internal network size and greater perceived social support protects against
factors and external resources. Resilience factors may decrease the LLD and predicts a better response to depression interventions
risk of a depressive episode, reduce severity or duration of that [282–284]. Such benefits may occur through mechanisms includ-
episode, or increase likelihood of recovery [267, 268]. If depression ing emotional support, tangible assistance (instrumental support),
contributes to accelerated biological aging, then bolstering such or opportunities for pleasurable activities [285]. Recent work has
resilience factors and reducing the frequency or duration of focused on loneliness, or perceived social isolation that is distinct
depressive episodes could hypothetically shift the biological aging from having fewer objective social contacts. Loneliness is
process towards a more normal trajectory. We discuss resilience bidirectionally associated with a host of negative outcomes,
factors and their corresponding vulnerability factors (Table 2) in including depression, poor physical health, cognitive and func-
context of treatments. tional decline, and mortality [286–288]. Loneliness may be a
neuropsychiatric manifestation of preclinical AD, as it is associated
Psychological factors: role for brief psychotherapy with an elevated dementia risk and higher levels of amyloid and
While factors such as temperament and personality may be tau pathology [289–291].
challenging to target with brief therapy, progress can improve
negative beliefs and coping strategies. Individuals’ beliefs about Evidence for targeting social connectedness. Few intervention
themselves and their environment influence how they cope with studies directly target social factors in depression [292]. Group
stressors or challenges. Depression risk is associated with lower therapy benefits LLD [293] but does not typically focus on social
self-esteem, anxiety sensitivity, and an external locus of control connectedness. Recent novel work has examined remote, layperson-
(i.e., a feeling that one cannot influence outcomes in one’s life) delivered interventions intended to improve social connectedness
[269]. The converse of these beliefs contribute to resilience, as and reduce loneliness in younger and older adults. Although not
does a sense of purpose and grit, defined as perseverance in directly targeting individuals with a depression diagnosis, they
achieving goals despite setbacks [267, 270–272]. Greater self- reduced depressive and anxiety symptoms [294–297]. Modifying
efficacy enhances individuals’ ability to flexibly apply coping existing psychotherapies to target social disconnection may also
strategies, including active coping strategies that directly address reduce suicide risk [294].
the stressor, or accommodative strategies involving adaptation to
the stressor. Such a flexible approach improves mental health Cognitive factors: role for cognitive training
outcomes and reduces depression [273, 274]. As previously discussed, LLD is associated with cognitive changes in
executive functioning, processing speed, and episodic memory
Evidence for psychotherapy. Psychological factors addressing [8–11]. However, not all individuals with LLD have cognitive
vulnerability or promoting resilience may be particularly amenable difficulties. There appear to be separate cognitive phenotypes
to psychotherapy. Evidence-based treatments in LLD include within LLD: “High Normal”, “Reduced Normal” (with a relative
cognitive-behavioral, problem-solving, interpersonal, and life- weakness in episodic recall), and “Low Executive Functions” [298].
review therapies [275, 276]. Such therapies influence functional The “High Normal” phenotype maintained cognitive performance
network connectivity, such as cognitive-behavioral therapy increas- despite similar levels of depression severity as the other pheno-
ing connectivity between the amygdala and CCN [277]. Meta- types. They also had higher levels of education and less vascular risk

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
9
factors, suggesting that cognitive reserve and vascular health may are associated with greater depressive symptom severity and
contribute to cognitive resilience in LLD [299]. Identifying cognitive increased risk of developing LLD [217, 330–335], particularly for
difficulties unique to the depressed individual allows for the dual sensory loss affecting both vision and hearing [336]. Several
prescription of personalized cognitive training interventions. hypotheses could explain these relationships, including how
sensory loss limits social activities, leading to isolation and
Evidence for benefits of cognitive training. For computerized worsening psychological health [332, 337]. This may not account
cognitive training to work, it must have an adequate duration and for the entire relationship, as sensory loss is further associated
be appropriately intense or difficult [300]. Evidence for the potential with physical decline [338], cognitive decline, and risk of
benefit of neurobiologically-informed computerized cognitive dementia [335, 339].
training in LLD comes from the approaches by Morimoto and
others that are optimized to treat LLD with executive dysfunction Evidence for interventions improving sensory function. Optimizing
[301]. They demonstrate that by targeting the underlying deficient sensory function benefits depressive symptoms and reduces
neural circuitry and associated cognitive deficits in LLD, cognitive depression risk. For impaired vision, improving residual vision and
functions and depressive symptoms improve, benefits transfer to self-management programs can reduce depressive symptoms
non-trained domains such as memory, and there are positive [340, 341]. Integration of psychotherapy techniques, such as
changes to underlying neural structural-functional connections behavioral activation can prevent depression in high-risk patients
[192, 300, 302]. Targeted cognitive training appears to modulate with macular degeneration [342]. A similar benefit is seen in
network functional connectivity in the DMN and CCN [303, 304]. preliminary clinical trials demonstrating that hearing aids may
Interventions targeting memory deficits (such as the Mayo Clinic’s benefit depressive symptoms, quality of life, and cognitive
Healthy Action to Benefit Independence and Thinking (HABIT) performance in older adults [343–345].
program) [305] benefit Mild Cognitive Impairment both with in-
person and virtual platforms. While this intervention has not been
conducted in LLD, the approach could translate to other popula- CONCLUSIONS
tions with primary memory issues. Similarly, processing speed We repeatedly describe bidirectional relationships between LLD
training shows benefit for up to 10-years in nondepressed older and lifespan factors such as aging, inflammation, vascular disease,
adults [306] and may be particularly favorable for LLD characterized and more. These reciprocal relationships in essence describe
by predominant cognitive and motor slowing. Augmenting positive feedback loops. In the absence of counterbalancing
cognitive training with neuromodulation approaches or other forces, such feedback loops can spiral out of control. Reframed in
nonpharmacological treatments may provide additional benefit the allostatic framework, maintaining stability requires adaptive
but need study in LLD [307]. regulation and resiliency.
This dynamic nature of allostatic processes contributes to LLD
Physical disability: need for sustainable movement-based heterogeneity. Vulnerability to developing depressive episodes
interventions results from accumulated factors, many of which have an initial
Motor deficits are common with aging, including slowing, onset earlier in life. Other vulnerability factors are unique to later life
coordination deficits, and balance difficulties [308, 309] and they and may contribute to a new diagnosis of depression or a relapse of
contribute to falls, disability, and mortality [310–314]. Depressed symptoms in previously remitted individuals. We propose that these
older adults are at increased risk for these motor problems vulnerability factors (Fig. 1) have negative effects on functional brain
[310, 314, 315]. This may be due to common contributors or networks that predispose networks towards a state of fragility and
comorbidities, such as vascular disease or other brain pathology instability.
[316, 317]. However, by increasing sedentary behavior and Etiological heterogeneity creates challenges for understanding
isolation, depression may also hasten muscle atrophy, decondi- both LLD’s neurobiology and variability in treatment responses. It
tioning, and frailty [37, 213–215]. also creates opportunities to use this heterogeneity to probe
specific mechanisms and guide focused, personalized treatment
Evidence for movement-based interventions. Structured physical approaches. Such examples include examining dopaminergic
exercise benefits depression symptoms across the adult lifespan system influences on LLD in patients with psychomotor slowing,
[318], including moderately benefitting older adults [319, 320]. It has testing anti-inflammatory medications in patients with elevated
positive benefits on hippocampal volume [321], may modulate inflammation, or using targeted cognitive training to treat patients
connectivity between key DMN and CCN regions [322, 323], and with LLD and executive dysfunction. While no single treatment
also augments the response to antidepressant medications [324]. will improve symptoms in all patients with LLD, combining
Physical activity improves global cognitive function in unimpaired established treatments such as pharmacotherapy, psychotherapy,
elders and benefits cognitive domains sensitive to aging, including and neuromodulation alongside personalized interventions that
attention, executive function, and memory [325, 326]. Similarly, bolster resilience or address comorbid disability may improve
physical exercise, particularly aerobic activity, may reduce the risk of outcomes for otherwise treatment-resistant patients.
dementia and benefit older adults with existing cognitive impair-
ment or dementia [327, 328]. Interestingly, recent work suggests
that mind-body therapies that combine movement-based REFERENCES
approaches with mindfulness, such as yoga or tai chi, may be 1. Taylor WD. Clinical practice. Depression in the elderly. N Engl J Med.
superior to conventional exercise for mood and cognitive outcomes 2014;371:1228–36.
[267, 329]. However, questions remain about optimal practices 2. Blazer DG. Depression in late life: review and commentary. J Gerontol A Biol Sci
needed to obtain such benefit, including frequency, intensity, Med Sci. 2003;58:249–65.
duration, and type of exercises [303]. Strategies to facilitate initiation 3. Lyness JM, Caine ED, King DA, Cox C, Yoediono Z. Psychiatric disorders in older
primary care patients. J Gen Intern Med. 1999;14:249–54.
and maintenance of exercise in community-based elders are sorely
4. Steffens DC, Hays JC, Krishnan KR. Disability in geriatric depression. Am J Geriatr
needed, particularly for individuals with chronic pain or disabilities Psychiatry. 1999;7:34–40.
that limit physical function. 5. Jiang W, Alexander J, Christopher E, Kuchibhatla M, Gaulden LH, Cuffe MS, et al.
Relationship of depression to increased risk of mortality and rehospitalization in
Sensory impairment: can improving sensory function help? patients with congestive heart failure. Arch Intern Med. 2001;161:1849–56.
Sensory impairment, including vision and hearing loss, is also 6. Conwell Y, Thompson C. Suicidal behavior in elders. Psychiatr Clin North Am.
common in later life. Uncorrected hearing loss and vision loss 2008;31:333–56.

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
10
7. Penninx BW, Geerlings SW, Deeg DJ, van Eijk JT, van Tilburg W, Beekman AT. 33. Menon V. Large-scale brain networks and psychopathology: a unifying triple
Minor and major depression and the risk of death in older persons. Arch Gen network model. Trends Cogn Sci. 2011;15:483–506.
Psychiatry. 1999;56:889–95. 34. Gunning FM, Oberlin LE, Schier M, Victoria LW. Brain-based mechanisms of late-
8. Manning KJ, Steffens DC. State of the science of neural systems in late-life life depression: implications for novel interventions. Semin Cell Dev Biol.
depression: impact on clinical presentation and treatment outcome. J Am 2021;116:169–79.
Geriatr Soc. 2018;66:S17–S23. 35. Keren H, O’Callaghan G, Vidal-Ribas P, Buzzell GA, Brotman MA, Leibenluft E,
9. Sexton CE, McDermott L, Kalu UG, Herrmann LL, Bradley KM, Allan CL, et al. et al. Reward processing in depression: a conceptual and meta-analytic review
Exploring the pattern and neural correlates of neuropsychological impairment across fMRI and EEG studies. Am J Psychiatry. 2018;175:1111–20.
in late-life depression. Psychol Med. 2012;42:1195–202. 36. Zhang WN, Chang SH, Guo LY, Zhang KL, Wang J. The neural correlates of
10. Boone KB, Lesser IM, Miller BL, Wohl M, Berman N, Lee ABP, et al. Cognitive reward-related processing in major depressive disorder: a meta-analysis of
functioning in older depressed outpatients: relationship of presence and functional magnetic resonance imaging studies. J Affect Disord. 2013;151:531–9.
severity of depression to neuropsychological test scores. Neuropsychology. 37. Taylor WD, Zald DH, Felger JC, Christman S, Claassen DO, Horga G, et al. Influ-
1995;9:390–8. ences of dopaminergic system dysfunction on late-life depression. Mol Psy-
11. Sheline YI, Barch DM, Garcia K, Gersing K, Piper C, Welsh-Bohmer KA, et al. Cog- chiatry. 2022;27:180–91.
nitive function in late life depression: relationships to depression severity, cere- 38. Saberi A, Mohammadi E, Zarei M, Eickhoff SB, Tahmasian M. Structural and
brovascular risk factors and processing speed. Biol Psychiatry. 2006;60:58–65. functional neuroimaging of late-life depression: a coordinate-based meta-ana-
12. Butters MA, Whyte EM, Nebes RD, Begley AE, Dew MA, Mulsant BH, et al. The lysis. Brain Imaging Behav. 2022;16:518–31.
nature and determinants of neuropsychological functioning in late-life 39. Manning K, Wang L, Steffens D. Recent advances in the use of imaging in
depression. Arch Gen Psychiatry. 2004;61:587–95. psychiatry: functional magnetic resonance imaging of large-scale brain net-
13. Nebes RD, Butters MA, Mulsant BH, Pollock BG, Zmuda MD, Houck PR, et al. works in late-life depression. F1000Res. 2019;8:F1000.
Decreased working memory and processing speed mediate cognitive impair- 40. Buckner RL, Andrews-Hanna JR, Schacter DL. The brain’s default network:
ment in geriatric depression. Psychol Med. 2000;30:679–91. anatomy, function, and relevance to disease. Ann N Y Acad Sci. 2008;1124:1–38.
14. Diniz BS, Butters MA, Albert SM, Dew MA, Reynolds CF 3rd. Late-life depression and 41. Zhou HX, Chen X, Shen YQ, Li L, Chen NX, Zhu ZC, et al. Rumination and the
risk of vascular dementia and Alzheimer’s disease: systematic review and meta- default mode network: Meta-analysis of brain imaging studies and implications
analysis of community-based cohort studies. Br J Psychiatry. 2013;202:329–35. for depression. Neuroimage. 2020;206:116287.
15. Andreescu C, Ajilore O, Aizenstein HJ, Albert K, Butters MA, Landman BA, et al. 42. Kaiser RH, Andrews-Hanna JR, Wager TD, Pizzagalli DA. Large-scale network
Disruption of neural homeostasis as a model of relapse and recurrence in late- dysfunction in major depressive disorder: a meta-analysis of resting-state
life depression. Am J Geriatr Psychiatry. 2019;27:1316–30. functional connectivity. JAMA Psychiatry. 2015;72:603–11.
16. Deng Y, McQuoid DR, Potter GG, Steffens DC, Albert K, Riddle M, et al. Predictors 43. Scalabrini A, Vai B, Poletti S, Damiani S, Mucci C, Colombo C, et al. All roads lead
of recurrence in remitted late-life depression. Depress Anxiety. 2018;35:658–67. to the default-mode network-global source of DMN abnormalities in major
17. Reynolds CF, Frank E, Perel JM, Imber SD, Cornes C, Miller MD, et al. Nortriptyline depressive disorder. Neuropsychopharmacology. 2020;45:2058–69.
and interpersonal psychotherapy as maintenance therapies for recurrent major 44. Javaheripour N, Li M, Chand T, Krug A, Kircher T, Dannlowski U, et al. Altered
depression: a randomized controlled trial in patients older than 59 years. J Am resting-state functional connectome in major depressive disorder: a mega-
Med Assoc. 1999;281:39–45. analysis from the PsyMRI consortium. Transl Psychiatry. 2021;11:511.
18. Reynolds CF 3rd, Dew MA, Pollock BG, Mulsant BH, Frank E, Miller MD, et al. 45. Tozzi L, Zhang X, Chesnut M, Holt-Gosselin B, Ramirez CA, Williams LM. Reduced
Maintenance treatment of major depression in old age. N Engl J Med. functional connectivity of default mode network subsystems in depression:
2006;354:1130–8. meta-analytic evidence and relationship with trait rumination. Neuroimage Clin.
19. Alexopoulos GS, Meyers BS, Young RC, Kalayam B, Kakuma T, Gabrielle M, et al. 2021;30:102570.
Executive dysfunction and long-term outcomes of geriatric depression. Arch 46. Wu M, Andreescu C, Butters MA, Tamburo R, Reynolds CF 3rd, Aizenstein H.
Gen Psychiatry. 2000;57:285–90. Default-mode network connectivity and white matter burden in late-life
20. Sachs-Ericsson N, Corsentino E, Moxley J, Hames JL, Rushing NC, Sawyer K, et al. depression. Psychiatr Res. 2011;194:39–46.
A longitudinal study of differences in late- and early-onset geriatric depression: 47. Gandelman JA, Albert K, Boyd BD, Park JW, Riddle M, Woodward ND, et al.
depressive symptoms and psychosocial, cognitive, and neurological function- Intrinsic functional network connectivity is associated with clinical symptoms
ing. Aging Ment Health. 2013;17:1–11. and cognition in late-life depression. Biol Psychiatry Cogn Neurosci Neuroima-
21. Klein DN, Schatzberg AF, McCullough JP, Dowling F, Goodman D, Howland RH, et al. ging. 2019;4:160–70.
Age of onset in chronic major depression: relation to demographic and clinical 48. Niendam TA, Laird AR, Ray KL, Dean YM, Glahn DC, Carter CS. Meta-analytic
variables, family history, and treatment response. J Affect Disord. 1999;55:149–57. evidence for a superordinate cognitive control network subserving diverse
22. Markett S, Montag C, Reuter M. Network neuroscience and personality. Personal executive functions. Cogn Affect Behav Neurosci. 2012;12:241–68.
Neurosci. 2018;1:e14. 49. Gerlach AR, Karim HT, Pecina M, Ajilore O, Taylor WD, Butters MA, et al. MRI
23. Newhouse P, Albert K. Estrogen, stress, and depression: a neurocognitive model. predictors of pharmacotherapy response in major depressive disorder. Neuro-
JAMA Psychiatry 2015;72:727–9. image Clin. 2022;36:103157.
24. Park C, Rosenblat JD, Brietzke E, Pan Z, Lee Y, Cao B, et al. Stress, epigenetics and 50. Dotson VM, McClintock SM, Verhaeghen P, Kim JU, Draheim AA, Syzmkowicz
depression: a systematic review. Neurosci Biobehav Rev. 2019;102:139–52. SM, et al. Depression and cognitive control across the lifespan: a systematic
25. Janssen J, Beekman AT, Comijs HC, Deeg DJ, Heeren TJ. Late-life depression: the review and meta-analysis. Neuropsychol Rev. 2020;30:461–76.
differences between early- and late-onset illness in a community-based sample. 51. Lockwood KA, Alexopoulos GS, van Gorp WG. Executive dysfunction in geriatric
Int J Geriatr Psychiatry. 2006;21:86–93. depression. Am J Psychiatry. 2002;159:1119–26.
26. Baldwin RC, Tomenson B. Depression in later life: a comparison of symptoms 52. Kim J, Kim Y-K. Crosstalk between depression and dementia with resting-state
and risk factors in early and late onset cases. Br J Psychiatry. 1995;167:649–52. fMRI studies and its relationship with cognitive functioning. Biomedicines.
27. Lavretsky H, Lesser IM, Wohl M, Miller BL. Relationship of age, age at onset, and 2021;9:82.
sex to depression in older adults. Am J Geriatr Psychiatry. 1998;6:248–56. 53. Menon V, Uddin LQ. Saliency, switching, attention and control: a network model
28. Dillon C, Allegri RF, Serrano CM, Iturry M, Salgado P, Glaser FB, et al. Late- versus of insula function. Brain Struct Funct. 2010;214:655–67.
early-onset geriatric depression in a memory research center. Neuropsychiatr 54. Berman MG, Nee DE, Casement M, Kim HS, Deldin P, Kross E, et al. Neural and
Dis Treat. 2009;5:517–26. behavioral effects of interference resolution in depression and rumination. Cogn
29. Rapp MA, Dahlman K, Sano M, Grossman HT, Haroutunian V, Gorman JM. Affect Behav Neurosci. 2011;11:85–96.
Neuropsychological differences between late-onset and recurrent geriatric 55. Hamilton JP, Furman DJ, Chang C, Thomason ME, Dennis E, Gotlib IH. Default-
major depression. Am J Psychiatry. 2005;162:691–8. mode and task-positive network activity in major depressive disorder: implica-
30. Salloway S, Malloy P, Kohn R, Gillard E, Duffy J, Rogg J, et al. MRI and neu- tions for adaptive and maladaptive rumination. Biol Psychiatry. 2011;70:327–33.
ropsychological differences in early- and late-life-onset geriatric depression. 56. Goldstein-Piekarski AN, Ball TM, Samara Z, Staveland BR, Keller AS, Fleming SL,
Neurology. 1996;46:1567–74. et al. Mapping neural circuit biotypes to symptoms and behavioral dimensions
31. Riddle M, Potter GG, McQuoid DR, Steffens DC, Beyer JL, Taylor WD. Longitudinal of depression and anxiety. Biol Psychiatry. 2022;91:561–71.
cognitive outcomes of clinical phenotypes of late-life depression. Am J Geriatr 57. Yun JY, Kim YK. Graph theory approach for the structural-functional brain
Psychiatry. 2017;25:1123–34. connectome of depression. Prog Neuropsychopharmacol Biol Psychiatry.
32. Ly M, Karim HT, Becker JT, Lopez OL, Anderson SJ, Aizenstein HJ, et al. Late-life 2021;111:110401.
depression and increased risk of dementia: a longitudinal cohort study. Transl 58. Rizvi SJ, Pizzagalli DA, Sproule BA, Kennedy SH. Assessing anhedonia in
Psychiatry. 2021;11:147. depression: potentials and pitfalls. Neurosci Biobehav Rev. 2016;65:21–35.

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
11
59. Andrews-Hanna JR, Snyder AZ, Vincent JL, Lustig C, Head D, Raichle ME, et al. 86. Hayward RD, Taylor WD, Smoski MJ, Steffens DC, Payne ME. Association of five-
Disruption of large-scale brain systems in advanced aging. Neuron. 2007; factor model personality domains and facets with presence, onset, and treat-
56:924–35. ment outcomes of major depression in older adults. Am J Geriatr Psychiatry.
60. Jockwitz C, Caspers S. Resting-state networks in the course of aging—differ- 2013;21:88–96.
ential insights from studies across the lifespan vs. amongst the old. Pflügers 87. Manning KJ, Chan G, Steffens DC. Neuroticism traits selectively impact long term
Arch - Eur J Physiol. 2021;473:793–803. illness course and cognitive decline in late-life depression. Am J Geriatr Psy-
61. Coelho A, Fernandes HM, Magalhães R, Moreira PS, Marques P, Soares JM, et al. chiatry. 2017;25:220–9.
Reorganization of brain structural networks in aging: a longitudinal study. J 88. Steffens DC, McQuoid DR, Smoski MJ, Potter GG. Clinical outcomes of older
Neurosci Res. 2021;99:1354–76. depressed patients with and without comorbid neuroticism. Int Psychogeriatr.
62. Ng KK, Lo JC, Lim JKW, Chee MWL, Zhou J. Reduced functional segregation 2013;25:1985–90.
between the default mode network and the executive control network in 89. Hasin DS, Sarvet AL, Meyers JL, Saha TD, Ruan WJ, Stohl M, et al. Epidemiology
healthy older adults: a longitudinal study. NeuroImage 2016;133:321–30. of adult DSM-5 major depressive disorder and its specifiers in the United States.
63. Betzel RF, Byrge L, He Y, Goñi J, Zuo X-N, Sporns O. Changes in structural and JAMA Psychiatry. 2018;75:336–46.
functional connectivity among resting-state networks across the human life- 90. Soares CN, Zitek B. Reproductive hormone sensitivity and risk for depression
span. NeuroImage. 2014;102:345–57. across the female life cycle: a continuum of vulnerability? J Psychiatry Neurosci.
64. Chan MY, Park DC, Savalia NK, Petersen SE, Wig GS. Decreased segregation of 2008;33:331–43.
brain systems across the healthy adult lifespan. Proc Natl Acad Sci USA. 91. Payne JL, Maguire J. Pathophysiological mechanisms implicated in postpartum
2014;111:E4997–E5006. depression. Front Neuroendocrinol. 2019;52:165–80.
65. Geerligs L, Renken RJ, Saliasi E, Maurits NM, Lorist MM. A brain-wide study of 92. Arevalo MA, Azcoitia I, Garcia-Segura LM. The neuroprotective actions of oes-
age-related changes in functional connectivity. Cereb Cortex. 2014;25:1987–99. tradiol and oestrogen receptors. Nat Rev Neurosci. 2015;16:17–29.
66. Grady C, Sarraf S, Saverino C, Campbell K. Age differences in the functional 93. Georgakis MK, Thomopoulos TP, Diamantaras AA, Kalogirou EI, Skalkidou A,
interactions among the default, frontoparietal control, and dorsal attention Daskalopoulou SS, et al. Association of age at menopause and duration of
networks. Neurobiol Aging. 2016;41:159–72. reproductive period with depression after menopause: a systematic review and
67. Hughes C, Faskowitz J, Cassidy BS, Sporns O, Krendl AC. Aging relates to a meta-analysis. JAMA Psychiatry. 2016;73:139–49.
disproportionately weaker functional architecture of brain networks during rest 94. Rocca WA, Bower JH, Maraganore DM, Ahlskog JE, Grossardt BR, de Andrade M,
and task states. NeuroImage. 2020;209:116521. et al. Increased risk of cognitive impairment or dementia in women who
68. Setton R, Mwilambwe-Tshilobo L, Girn M, Lockrow AW, Baracchini G, Hughes C, underwent oophorectomy before menopause. Neurology. 2007;69:1074–83.
et al. Age differences in the functional architecture of the human brain. Cereb 95. Stein MB, Jain S, Giacino JT, Levin H, Dikmen S, Nelson LD, et al. Risk of post-
Cortex. 2022;33:114–34. traumatic stress disorder and major depression in civilian patients after mild
69. Zonneveld HI, Pruim RH, Bos D, Vrooman HA, Muetzel RL, Hofman A, et al. traumatic brain injury: a TRACK-TBI study. JAMA Psychiatry. 2019;76:249–58.
Patterns of functional connectivity in an aging population: the Rotterdam Study. 96. Milaneschi Y, Simmons WK, van Rossum EFC, Penninx BW. Depression and obesity:
Neuroimage. 2019;189:432–44. evidence of shared biological mechanisms. Mol Psychiatry. 2019;24:18–33.
70. Buckner RL. Memory and executive function in aging and AD: multiple factors that 97. Taylor WD, McQuoid DR, Krishnan KR. Medical comorbidity in late-life depres-
cause decline and reserve factors that compensate. Neuron. 2004;44:195–208. sion. Int J Geriatr Psychiatry. 2004;19:935–43.
71. Park DC, Reuter-Lorenz P. The adaptive brain: aging and neurocognitive scaf- 98. Griffin SC, Young JR, Naylor JC, Allen KD, Beckham JC, Calhoun PS. Reciprocal
folding. Annu Rev Psychol. 2009;60:173–96. effects between depressive symptoms and pain in veterans over 50 years of age
72. Morcom AM, Johnson W. Neural reorganization and compensation in aging. J or older. Pain Med. 2022;23:295–304.
Cogn Neurosci. 2015;27:1275–85. 99. Alexopoulos GS, Morimoto SS. The inflammation hypothesis in geriatric
73. Zhu Z, Johnson NF, Kim C, Gold BT. Reduced frontal cortex efficiency is asso- depression. Int J Geriatr Psychiatry. 2011;26:1109–18.
ciated with lower white matter integrity in aging. Cereb Cortex. 2013;25:138–46. 100. Dantzer R, O’Connor JC, Lawson MA, Kelley KW. Inflammation-associated
74. Zhu Z, Johnson NF, Kim C, Gold BT. Reduced frontal cortex efficiency is asso- depression: from serotonin to kynurenine. Psychoneuroendocrinology. 2011;
ciated with lower white matter integrity in aging. Cereb Cortex. 2015;25:138–46. 36:426–36.
75. Howard DM, Adams MJ, Clarke TK, Hafferty JD, Gibson J, Shirali M, et al. 101. Miller AH, Raison CL. The role of inflammation in depression: from evolutionary
Genome-wide meta-analysis of depression identifies 102 independent variants imperative to modern treatment target. Nat Rev Immunol. 2016;16:22–34.
and highlights the importance of the prefrontal brain regions. Nat Neurosci. 102. Reichenberg A, Yirmiya R, Schuld A, Kraus T, Haack M, Morag A, et al. Cytokine-
2019;22:343–52. associated emotional and cognitive disturbances in humans. Arch Gen Psy-
76. Buch AM, Liston C. Dissecting diagnostic heterogeneity in depression by inte- chiatry. 2001;58:445–52.
grating neuroimaging and genetics. Neuropsychopharmacology. 2021;46:156–75. 103. Grigoleit JS, Kullmann JS, Wolf OT, Hammes F, Wegner A, Jablonowski S, et al.
77. Nguyen TD, Harder A, Xiong Y, Kowalec K, Hagg S, Cai N, et al. Genetic het- Dose-dependent effects of endotoxin on neurobehavioral functions in humans.
erogeneity and subtypes of major depression. Mol Psychiatry. 2022;27:1667–75. PLoS ONE. 2011;6:e28330.
78. Taylor WD, Aizenstein HJ, Alexopoulos GS. The vascular depression hypoth- 104. Kohler CA, Freitas TH, Maes M, de Andrade NQ, Liu CS, Fernandes BS, et al.
esis: mechanisms linking vascular disease with depression. Mol Psychiatry. Peripheral cytokine and chemokine alterations in depression: a meta-analysis of
2013;18:963–74. 82 studies. Acta Psychiatr Scand. 2017;135:373–87.
79. Bellis MA, Hughes K, Ford K, Ramos Rodriguez G, Sethi D, Passmore J. Life course 105. Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, et al. A meta-
health consequences and associated annual costs of adverse childhood analysis of cytokines in major depression. Biol Psychiatry. 2010;67:446–57.
experiences across Europe and North America: a systematic review and meta- 106. Lindqvist D, Janelidze S, Hagell P, Erhardt S, Samuelsson M, Minthon L, et al.
analysis. Lancet Public Health. 2019;4:e517–e28. Interleukin-6 is elevated in the cerebrospinal fluid of suicide attempters and
80. Saleh A, Potter GG, McQuoid DR, Boyd B, Turner R, MacFall JR, et al. Effects of related to symptom severity. Biol Psychiatry. 2009;66:287–92.
early life stress on depression, cognitive performance and brain morphology. 107. Howren MB, Lamkin DM, Suls J. Associations of depression with C-reactive
Psychol Med. 2017;47:171–81. protein, IL-1, and IL-6: a meta-analysis. Psychosom Med. 2009;71:171–86.
81. Meaney MJ. Maternal care, gene expression, and the transmission of indivi- 108. Sartori AC, Vance DE, Slater LZ, Crowe M. The impact of inflammation on cog-
dual differences in stress reactivity across generations. Ann Rev Neurosci. nitive function in older adults: implications for healthcare practice and research.
2001;24:1161–92. J Neurosci Nurs. 2012;44:206–17.
82. Iob E, Lacey R, Steptoe A. Adverse childhood experiences and depressive 109. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, et al.
symptoms in later life: longitudinal mediation effects of inflammation. Brain Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y
Behav Immun. 2020;90:97–107. Acad Sci. 2000;908:244–54.
83. Cheong EV, Sinnott C, Dahly D, Kearney PM. Adverse childhood experiences 110. Franceschi C, Zaikin A, Gordleeva S, Ivanchenko M, Bonifazi F, Storci G, et al.
(ACEs) and later-life depression: perceived social support as a potential pro- Inflammaging 2018: an update and a model. Semin Immunol. 2018;40:1–5.
tective factor. BMJ Open. 2017;7:e013228. 111. Meszaros A, Molnar K, Nogradi B, Hernadi Z, Nyul-Toth A, Wilhelm I, et al.
84. Zannas AS, McQuoid DR, Steffens DC, Chrousos GP, Taylor WD. Stressful life Neurovascular inflammaging in health and disease. Cells. 2020;9:1614.
events, perceived stress, and 12-month course of geriatric depression: direct 112. Nair D, Cukor D, Taylor WD, Cavanaugh KL. Applying a biopsychosocial frame-
effects and moderation by the 5-HTTLPR and COMT Val158Met polymorphisms. work to achieve durable behavior change in kidney disease. Semin Nephrol.
Stress. 2012;15:425–34. 2021;41:487–504.
85. Kendler KS, Karkowski LM, Prescott CA. Causal relationship between stressful life 113. Rutledge T, Linke SE, Krantz DS, Johnson BD, Bittner V, Eastwood JA, et al.
events and the onset of major depression. Am J Psychiatry. 1999;156:837–41. Comorbid depression and anxiety symptoms as predictors of cardiovascular

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
12
events: results from the NHLBI-sponsored Women’s Ischemia Syndrome Eva- evidence from meta-analyses and Mendelian randomization studies. J Psychiatr
luation (WISE) study. Psychosom Med. 2009;71:958–64. Res. 2018;103:189–207.
114. Drosselmeyer J, Jacob L, Rathmann W, Rapp MA, Kostev K. Depression risk in 138. Liu JJ, Wei YB, Strawbridge R, Bao Y, Chang S, Shi L, et al. Peripheral cytokine
patients with late-onset rheumatoid arthritis in Germany. Qual Life Res. levels and response to antidepressant treatment in depression: a systematic
2017;26:437–43. review and meta-analysis. Mol Psychiatry. 2020;25:339–50.
115. Benros ME, Waltoft BL, Nordentoft M, Ostergaard SD, Eaton WW, Krogh J, et al. 139. Kohler-Forsberg O, C NL, Hjorthoj C, Nordentoft M, Mors O, Benros ME. Efficacy of
Autoimmune diseases and severe infections as risk factors for mood disorders: a anti-inflammatory treatment on major depressive disorder or depressive symp-
nationwide study. JAMA Psychiatry. 2013;70:812–20. toms: meta-analysis of clinical trials. Acta Psychiatr Scand. 2019;139:404–19.
116. Luning Prak ET, Brooks T, Makhoul W, Beer JC, Zhao L, Girelli T, et al. No increase 140. Kohler O, Benros ME, Nordentoft M, Farkouh ME, Iyengar RL, Mors O, et al. Effect
in inflammation in late-life major depression screened to exclude physical ill- of anti-inflammatory treatment on depression, depressive symptoms, and
ness. Transl Psychiatry. 2022;12:118. adverse effects: a systematic review and meta-analysis of randomized clinical
117. Quan N, Banks WA. Brain-immune communication pathways. Brain Behav trials. JAMA Psychiatry. 2014;71:1381–91.
Immun. 2007;21:727–35. 141. Bavaresco DV, Uggioni MLR, Ferraz SD, Marques RMM, Simon CS, Dagostin VS,
118. Yirmiya R, Rimmerman N, Reshef R. Depression as a microglial disease. Trends et al. Efficacy of infliximab in treatment-resistant depression: A systematic
Neurosci. 2015;38:637–58. review and meta-analysis. Pharm Biochem Behav. 2020;188:172838.
119. Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation 142. Raison CL, Rutherford RE, Woolwine BJ, Shuo C, Schettler P, Drake DF, et al. A
to sickness and depression: when the immune system subjugates the brain. Nat randomized controlled trial of the tumor necrosis factor antagonist infliximab
Rev Neurosci. 2008;9:46–56. for treatment-resistant depression: the role of baseline inflammatory bio-
120. Dilger RN, Johnson RW. Aging, microglial cell priming, and the discordant markers. JAMA Psychiatry. 2013;70:31–41.
central inflammatory response to signals from the peripheral immune system. J 143. Dunjic-Kostic B, Ivkovic M, Radonjic NV, Petronijevic ND, Pantovic M, Damja-
Leukoc Biol. 2008;84:932–9. novic A, et al. Melancholic and atypical major depression—connection between
121. Frank MG, Weber MD, Watkins LR, Maier SF. Stress-induced neuroinflammatory cytokines, psychopathology and treatment. Prog Neuropsychopharmacol Biol
priming: a liability factor in the etiology of psychiatric disorders. Neurobiol Psychiatry. 2013;43:1–6.
Stress. 2016;4:62–70. 144. Zimmerman B, Rypma B, Gratton G, Fabiani M. Age-related changes in cere-
122. Haroon E, Miller AH, Sanacora G. Inflammation, glutamate, and glia: a trio of brovascular health and their effects on neural function and cognition: a com-
trouble in mood disorders. Neuropsychopharmacology. 2017;42:193–215. prehensive review. Psychophysiology. 2021;58:e13796.
123. Haroon E, Felger JC, Woolwine BJ, Chen X, Parekh S, Spivey JR, et al. Age-related 145. Mustapha M, Nassir C, Aminuddin N, Safri AA, Ghazali MM. Cerebral small vessel
increases in basal ganglia glutamate are associated with TNF, reduced moti- disease (CSVD)—lessons from the animal models. Front Physiol. 2019;10:1317.
vation and decreased psychomotor speed during IFN-alpha treatment: pre- 146. Alexopoulos GS, Meyers BS, Young RC, Campbell S, Silbersweig D, Charlson M.
liminary findings. Brain Behav Immun. 2015;46:17–22. ‘Vascular depression’ hypothesis. Arch Gen Psychiatry. 1997;54:915–22.
124. Haroon E, Woolwine BJ, Chen X, Pace TW, Parekh S, Spivey JR, et al. IFN-alpha- 147. Krishnan KRR, Hays JC, Blazer DG. MRI-defined vascular depression. Am J Psy-
induced cortical and subcortical glutamate changes assessed by magnetic chiatry. 1997;154:497–501.
resonance spectroscopy. Neuropsychopharmacology. 2014;39:1777–85. 148. Blochl M, Schaare HL, Kunzmann U, Nestler S. The age-dependent association
125. Capuron L, Schroecksnadel S, Feart C, Aubert A, Higueret D, Barberger-Gateau P, between vascular risk factors and depressed mood. J Gerontol B Psychol Sci Soc
et al. Chronic low-grade inflammation in elderly persons is associated with Sci. 2022;77:284–94.
altered tryptophan and tyrosine metabolism: role in neuropsychiatric symp- 149. Krishnan KRR, Tayor WD, McQuoid DR, MacFall JR, Payne ME, Provenzale JM,
toms. Biol Psychiatry. 2011;70:175–82. et al. Clinical characteristics of magnetic resonance imaging-defined subcortical
126. Felger JC, Mun J, Kimmel HL, Nye JA, Drake DF, Hernandez CR, et al. Chronic ischemic depression. Biol Psychiatry. 2004;55:390–7.
interferon-alpha decreases dopamine 2 receptor binding and striatal dopamine 150. Fang Y, Qin T, Liu W, Ran L, Yang Y, Huang H, et al. Cerebral small-vessel disease
release in association with anhedonia-like behavior in nonhuman primates. and risk of incidence of depression: a meta-analysis of longitudinal cohort
Neuropsychopharmacology. 2013;38:2179–87. studies. J Am Heart Assoc. 2020;9:e016512.
127. Seaman KL, Smith CT, Juarez EJ, Dang LC, Castrellon JJ, Burgess LL, et al. Dif- 151. Smagula SF, Aizenstein HJ. Brain structural connectivity in late-life major
ferential regional decline in dopamine receptor availability across adulthood: depressive disorder. Biol Psychiatry Cogn Neurosci Neuroimaging. 2016;1:271–7.
linear and nonlinear effects of age. Hum Brain Mapp. 2019;40:3125–38. 152. Herrmann LL, Le Masurier M, Ebmeier KP. White matter hyperintensities in late
128. Karrer TM, Josef AK, Mata R, Morris ED, Samanez-Larkin GR. Reduced dopamine life depression: a systematic review. J Neurol Neurosurg Psychiatry.
receptors and transporters but not synthesis capacity in normal aging adults: a 2008;79:619–24.
meta-analysis. Neurobiol Aging. 2017;57:36–46. 153. Salo KI, Scharfen J, Wilden ID, Schubotz RI, Holling H. Confining the concept of
129. Gu Y, Vorburger R, Scarmeas N, Luchsinger JA, Manly JJ, Schupf N, et al. Cir- vascular depression to late-onset depression: a meta-analysis of MRI-defined
culating inflammatory biomarkers in relation to brain structural measurements hyperintensity burden in major depressive disorder and bipolar disorder. Front
in a non-demented elderly population. Brain Behav Immun. 2017;65:150–60. Psychol. 2019;10:1241.
130. Kraynak TE, Marsland AL, Wager TD, Gianaros PJ. Functional neuroanatomy of 154. Meinert S, Nowack N, Grotegerd D, Repple J, Winter NR, Abheiden I, et al.
peripheral inflammatory physiology: A meta-analysis of human neuroimaging Association of brain white matter microstructure with cognitive performance in
studies. Neurosci Biobehav Rev. 2018;94:76–92. major depressive disorder and healthy controls: a diffusion-tensor imaging
131. Enache D, Pariante CM, Mondelli V. Markers of central inflammation in major study. Mol Psychiatry. 2022;27:1103–10.
depressive disorder: a systematic review and meta-analysis of studies examining 155. Taylor WD, Payne ME, Krishnan KRR, Wagner HR, Provenzale JM, Steffens DC,
cerebrospinal fluid, positron emission tomography and post-mortem brain tis- et al. Evidence of white matter tract disruption in MRI hyperintensities. Biol
sue. Brain Behav Immun. 2019;81:24–40. Psychiatry. 2001;50:179–83.
132. Torres-Platas SG, Cruceanu C, Chen GG, Turecki G, Mechawar N. Evidence for 156. Taylor WD, Zhao Z, Ashley-Koch A, Payne ME, Steffens DC, Krishnan RR, et al.
increased microglial priming and macrophage recruitment in the dorsal Fiber tract-specific white matter lesion severity Findings in late-life depression
anterior cingulate white matter of depressed suicides. Brain Behav Immun. and by AGTR1 A1166C genotype. Hum Brain Mapp. 2013;34:295–303.
2014;42:50–9. 157. Sheline YI, Price JL, Vaishnavi SN, Mintun MA, Barch DM, Epstein AA, et al.
133. Vogelzangs N, Comijs HC, Oude Voshaar RC, Stek ML, Penninx BW. Late-life Regional white matter hyperintensity burden in automated segmentation dis-
depression symptom profiles are differentially associated with immunometa- tinguishes late-life depressed subjects from comparison subjects matched for
bolic functioning. Brain Behav Immun. 2014;41:109–15. vascular risk factors. Am J Psychiatry. 2008;165:524–32.
134. Uher R, Tansey KE, Dew T, Maier W, Mors O, Hauser J, et al. An inflammatory 158. Taylor WD, Kudra K, Zhao Z, Steffens DC, MacFall JR. Cingulum bundle white
biomarker as a differential predictor of outcome of depression treatment with matter lesions influence antidepressant response in late-life depression: a pilot
escitalopram and nortriptyline. Am J Psychiatry. 2014;171:1278–86. study. J Affect Disord. 2014;162:8–11.
135. Chamberlain SR, Cavanagh J, de Boer P, Mondelli V, Jones DNC, Drevets WC, 159. Wen MC, Steffens DC, Chen MK, Zainal NH. Diffusion tensor imaging studies in
et al. Treatment-resistant depression and peripheral C-reactive protein. Br J late-life depression: systematic review and meta-analysis. Int J Geriatr Psychiatry.
Psychiatry. 2019;214:11–9. 2014;29:1173–84.
136. Strawbridge R, Arnone D, Danese A, Papadopoulos A, Herane Vives A, Cleare AJ. 160. Zanon Zotin MC, Sveikata L, Viswanathan A, Yilmaz P. Cerebral small vessel
Inflammation and clinical response to treatment in depression: a meta-analysis. disease and vascular cognitive impairment: from diagnosis to management.
Eur Neuropsychopharmacol. 2015;25:1532–43. Curr Opin Neurol. 2021;34:246–57.
137. Kohler CA, Evangelou E, Stubbs B, Solmi M, Veronese N, Belbasis L, et al. 161. Arola A, Levanen T, Laakso HM, Pitkanen J, Koikkalainen J, Lotjonen J, et al. Neu-
Mapping risk factors for depression across the lifespan: An umbrella review of ropsychiatric symptoms are associated with exacerbated cognitive impairment in

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
13
covert cerebral small vessel disease. J Int Neuropsychol Soc. 2022; 1–8. https:// the Alzheimer’s disease neuroimaging initiative depression project. Biol Psy-
doi.org/10.1017/S1355617722000480. chiatry. 2021;89:757–65.
162. Butters MA, Young JB, Lopez O, Aizenstein HJ, Mulsant BH, Reynolds CF 3rd, 187. Babulal GM, Roe CM, Stout SH, Rajasekar G, Wisch JK, Benzinger TLS, et al.
et al. Pathways linking late-life depression to persistent cognitive impairment Depression is associated with tau and not amyloid positron emission tomo-
and dementia. Dialogues Clin Neurosci. 2008;10:345–57. graphy in cognitively normal adults. J Alzheimers Dis. 2020;74:1045–55.
163. Taylor WD, Steffens DC, MacFall JR, McQuoid DR, Payne ME, Provenzale JM, et al. 188. Alexopoulos GS, Kiosses DN, Heo M, Murphy CF, Shanmugham B, Gunning-
White matter hyperintensity progression and late-life depression outcomes. Dixon F. Executive dysfunction and the course of geriatric depression. Biol
Arch Gen Psychiatry. 2003;60:1090–6. Psychiatry. 2005;58:204–10.
164. Alexopoulos GS. Mechanisms and treatment of late-life depression. Transl Psy- 189. Rosenberg PB, Drye LT, Martin BK, Frangakis C, Mintzer JE, Weintraub D, et al.
chiatry. 2019;9:188. Sertraline for the treatment of depression in Alzheimer disease. Am J Geriatr
165. Sheline YI, Pieper CF, Barch DM, Welsh-Bohmer K, McKinstry RC, MacFall JR, et al. Psychiatry. 2010;18:136–45.
Support for the vascular depression hypothesis in late-life depression: results of 190. Taylor WD, Boyd BD, Elson D, Andrews P, Albert K, Vega J, et al. Preliminary
a 2-site, prospective, antidepressant treatment trial. Arch Gen Psychiatry. evidence that cortical amyloid burden predicts poor response to antidepressant
2010;67:277–85. medication treatment in cognitively intact individuals with late-life depression.
166. Boutzoukas EM, O’Shea A, Kraft JN, Hardcastle C, Evangelista ND, Hausman HK, et al. Am J Geriatr Psychiatry. 2021;29:448–57.
Higher white matter hyperintensity load adversely affects pre-post proximal cog- 191. Li P, Hsiao IT, Liu CY, Chen CH, Huang SY, Yen TC, et al. Beta-amyloid deposition
nitive training performance in healthy older adults. Geroscience. 2022;44:1441–55. in patients with major depressive disorder with differing levels of treatment
167. Cui K, Song R, Xu H, Shang Y, Qi X, Buchman AS, et al. Association of cardio- resistance: a pilot study. EJNMMI Res. 2017;7:24.
vascular risk burden with risk and progression of disability: mediating role of 192. Morimoto SS, Altizer RA, Gunning FM, Hu W, Liu J, Cote SE, et al. Targeting
cardiovascular disease and cognitive decline. J Am Heart Assoc. 2020;9:e017346. cognitive control deficits with neuroplasticity-based computerized cognitive
168. Hou Y, Li Y, Yang S, Qin W, Yang L, Hu W. Gait impairment and upper extremity remediation in patients with geriatric major depression: a randomized, double-
disturbance are associated with total magnetic resonance imaging cerebral blind, controlled trial. Am J Geriatr Psychiatry. 2020;28:971–80.
small vessel disease burden. Front Aging Neurosci. 2021;13:640844. 193. Weintraub D, Aarsland D, Chaudhuri KR, Dobkin RD, Leentjens AF, Rodriguez-
169. Stewart R. Cardiovascular disease and frailty: what are the mechanistic links? Violante M, et al. The neuropsychiatry of Parkinson’s disease: advances and
Clin Chem. 2019;65:80–6. challenges. Lancet Neurol. 2022;21:89–102.
170. Harshfield EL, Pennells L, Schwartz JE, Willeit P, Kaptoge S, Bell S, et al. Asso- 194. Wang S, Mao S, Xiang D, Fang C. Association between depression and the
ciation between depressive symptoms and incident cardiovascular diseases. J subsequent risk of Parkinson’s disease: a meta-analysis. Prog Neuropsycho-
Am Med Assoc. 2020;324:2396–405. pharmacol Biol Psychiatry. 2018;86:186–92.
171. Zahodne LB, Gilsanz P, Glymour MM, Gibbons LE, Brewster P, Hamilton J, et al. 195. Rotter A, Lenz B, Pitsch R, Richter-Schmidinger T, Kornhuber J, Rhein C. Alpha-
Comparing variability, severity, and persistence of depressive symptoms as synuclein RNA expression is increased in major depression. Int J Mol Sci.
predictors of future stroke risk. Am J Geriatr Psychiatry. 2017;25:120–8. 2019;20:2029.
172. Jorm AF, Jolley D. The incidence of dementia: a meta-analysis. Neurology. 196. Bruno D, Reichert Plaska C, Clark DPA, Zetterberg H, Blennow K, Verbeek MM,
1998;51:728–33. et al. CSF alpha-synuclein correlates with CSF neurogranin in late-life depres-
173. Dotson VM, Beydoun MA, Zonderman AB. Recurrent depressive symptoms and the sion. Int J Neurosci. 2021;131:357–61.
incidence of dementia and mild cognitive impairment. Neurology. 2010;75:27–34. 197. Khan SS, Singer BD, Vaughan DE. Molecular and physiological manifestations
174. Barnes DE, Yaffe K, Byers AL, McCormick M, Schaefer C, Whitmer RA. Midlife vs and measurement of aging in humans. Aging Cell. 2017;16:624–33.
late-life depressive symptoms and risk of dementia: differential effects for Alz- 198. Christman S, Bermudez C, Hao L, Landman BA, Boyd B, Albert K, et al. Accelerated
heimer disease and vascular dementia. Arch Gen Psychiatry. 2012;69:493–8. brain aging predicts impaired cognitive performance and greater disability in
175. Leung DKY, Chan WC, Spector A, Wong GHY. Prevalence of depression, anxiety, geriatric but not midlife adult depression. Transl Psychiatry. 2020;10:317.
and apathy symptoms across dementia stages: a systematic review and meta- 199. Cole JH, Marioni RE, Harris SE, Deary IJ. Brain age and other bodily ‘ages’:
analysis. Int J Geriatr Psychiatry. 2021;36:1330–44. implications for neuropsychiatry. Mol Psychiatry. 2019;24:266–81.
176. Wilson RS, Capuano AW, Boyle PA, Hoganson GM, Hizel LP, Shah RC, et al. 200. Miller MW, Sadeh N. Traumatic stress, oxidative stress and post-traumatic stress
Clinical-pathologic study of depressive symptoms and cognitive decline in old disorder: neurodegeneration and the accelerated-aging hypothesis. Mol Psy-
age. Neurology. 2014;83:702–9. chiatry. 2014;19:1156–62.
177. Mirza SS, Wolters FJ, Swanson SA, Koudstaal PJ, Hofman A, Tiemeier H, et al. 10- 201. Han LKM, Aghajani M, Clark SL, Chan RF, Hattab MW, Shabalin AA, et al. Epi-
year trajectories of depressive symptoms and risk of dementia: a population- genetic aging in major depressive disorder. Am J Psychiatry. 2018;175:774–82.
based study. Lancet Psychiatry. 2016;3:628–35. 202. Robinson O, Chadeau Hyam M, Karaman I, Climaco Pinto R, Ala-Korpela M,
178. Krishnan KR, Tupler LA, Ritchie JCJ, McDonald WM, Knight DL, Nemeroff CB, Handakas E, et al. Determinants of accelerated metabolomic and epigenetic
et al. Apolipoprotein E-e4 frequency in geriatric depression. Biol Psychiatry. aging in a UK cohort. Aging Cell. 2020;19:e13149.
1996;40:69–71. 203. Dunlop K, Victoria LW, Downar J, Gunning FM, Liston C. Accelerated brain aging
179. Steffens DC, Norton MC, Hart AD, Skoog I, Corcoran C, Breitner JC. Apolipo- predicts impulsivity and symptom severity in depression. Neuropsycho-
protein E genotype and major depression in a community of older adults. The pharmacology. 2021;46:911–9.
Cache County Study. Psychol Med. 2003;33:541–7. 204. Han LKM, Dinga R, Hahn T, Ching CRK, Eyler LT, Aftanas L, et al. Brain aging in
180. Harerimana NV, Liu Y, Gerasimov ES, Duong D, Beach TG, Reiman EM, et al. major depressive disorder: results from the ENIGMA major depressive disorder
Genetic evidence supporting a causal role of depression in Alzheimer’s disease. working group. Mol Psychiatry. 2021;26:5124–39.
Biol Psychiatry. 2022;92:25–33. 205. Han LKM, Dinga R, Leenings R, Hahn T, Cole JH, Aftanas LI, et al. A large-scale
181. Mahgoub N, Alexopoulos GS. Amyloid hypothesis: is there a role for antiamyloid ENIGMA multisite replication study of brain age in depression. Neuroimage Rep.
treatment in late-life depression? Am J Geriatr Psychiatry. 2016;24:239–47. 2022;2:100149.
182. Wu KY, Hsiao IT, Chen CS, Chen CH, Hsieh CJ, Wai YY, et al. Increased brain 206. Taylor WD. Coexisting depression and frailty as an accelerated aging phenotype
amyloid deposition in patients with a lifetime history of major depression: of late-life depression. Int Psychogeriatr. 2023;1–7. https://doi.org/10.1017/
evidenced on 18F-florbetapir (AV-45/Amyvid) positron emission tomography. S1041610223000170.
Eur J Nucl Med Mol Imaging. 2014;41:714–22. 207. Brown PJ, Wall MM, Chen C, Levine ME, Yaffe K, Roose SP, et al. Biological age,
183. Wu KY, Liu CY, Chen CS, Chen CH, Hsiao IT, Hsieh CJ, et al. Beta-amyloid not chronological age, is associated with late-life depression. J Gerontol A Biol
deposition and cognitive function in patients with major depressive disorder Sci Med Sci. 2018;73:1370–6.
with different subtypes of mild cognitive impairment: (18)F-florbetapir (AV-45/ 208. Diniz BS, Mulsant BH, Reynolds CF 3rd, Blumberger DM, Karp JF, Butters MA, et al.
Amyvid) PET study. Eur J Nucl Med Mol Imaging. 2016;43:1067–76. Association of molecular senescence markers in late-life depression with clinical
184. Hyung WSW, Kang J, Kim J, Lee S, Youn H, Ham BJ, et al. Cerebral amyloid characteristics and treatment outcome. JAMA Netw Open. 2022;5:e2219678.
accumulation is associated with distinct structural and functional alterations in 209. Ballester PL, Suh JS, Nogovitsyn N, Hassel S, Strother SC, Arnott SR, et al.
the brain of depressed elders with mild cognitive impairment. J Affect Disord. Accelerated brain aging in major depressive disorder and antidepressant
2021;281:459–66. treatment response: a CAN-BIND report. Neuroimage Clin. 2021;32:102864.
185. Wang SM, Kim NY, Um YH, Kang DW, Na HR, Lee CU, et al. Default mode 210. Han LKM, Verhoeven JE, Tyrka AR, Penninx B, Wolkowitz OM, Mansson KNT, et al.
network dissociation linking cerebral beta amyloid retention and depression in Accelerating research on biological aging and mental health: current challenges
cognitively normal older adults. Neuropsychopharmacology. 2021;46:2180–7. and future directions. Psychoneuroendocrinology. 2019;106:293–311.
186. Mackin RS, Insel PS, Landau S, Bickford D, Morin R, Rhodes E, et al. Late-life 211. Elliott ML, Caspi A, Houts RM, Ambler A, Broadbent JM, Hancox RJ, et al. Dis-
depression is associated with reduced cortical amyloid burden: findings from parities in the pace of biological aging among midlife adults of the same

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
14
chronological age have implications for future frailty risk and policy. Nat Aging. outcome in early-onset versus late-onset late-life depression. Am J Geriatr
2021;1:295–308. Psychiatry. 2014;22:1039–46.
212. Fried LP, Cohen AA, Xue QL, Walston J, Bandeen-Roche K, Varadhan R. The 237. Reynolds CF 3rd, Dew MA, Frank E, Begley AE, Miller MD, Cornes C, et al. Effects
physical frailty syndrome as a transition from homeostatic symphony to of age at onset of first lifetime episode of recurrent major depression on
cacophony. Nat Aging. 2021;1:36–46. treatment response and illness course in elderly patients. Am J Psychiatry.
213. Brown PJ, Roose SP, Zhang J, Wall M, Rutherford BR, Ayonayon HN, et al. 1998;155:795–9.
Inflammation, depression, and slow gait: a high mortality phenotype in later life. 238. Kok RM, Reynolds CF 3rd. Management of depression in older adults: a review. J
J Gerontol A Biol Sci Med Sci. 2016;71:221–7. Am Med Assoc. 2017;317:2114–22.
214. Brown PJ, Ciarleglio A, Roose SP, Montes Garcia C, Chung S, Fernandes S, et al. 239. Lenze EJ, Mulsant BH, Roose SP, Lavretsky H, Reynolds CF3rd, Blumberger DM,
Frailty and depression in late life: a high-risk comorbidity with distinctive clinical et al. Antidepressant augmentation versus switch in treatment-resistant geriatric
presentation and poor antidepressant response. J Gerontol A Biol Sci Med Sci. depression. N Engl J Med. 2023;388:1067–79.
2022;77:1055–62. 240. Lavretsky H, Reinlieb M, St Cyr N, Siddarth P, Ercoli LM, Senturk D. Citalopram,
215. Brown PJ, Roose SP, O’Boyle KR, Ciarleglio A, Maas B, Igwe KC, et al. Frailty and methylphenidate, or their combination in geriatric depression: a randomized,
its correlates in adults with late life depression. Am J Geriatr Psychiatry. double-blind, placebo-controlled trial. Am J Psychiatry. 2015;172:561–9.
2020;28:145–54. 241. Cooper C, Katona C, Lyketsos K, Blazer D, Brodaty H, Rabins P, et al. A systematic
216. Lever-van Milligen BA, Lamers F, Smit JH, Penninx BW. Six-year trajectory of review of treatments for refractory depression in older people. Am J Psychiatry.
objective physical function in persons with depressive and anxiety disorders. 2011;168:681–8.
Depress Anxiety. 2017;34:188–97. 242. Lenze EJ, Mulsant BH, Blumberger DM, Karp JF, Newcomer JW, Anderson SJ,
217. Rutherford BR, Taylor WD, Brown PJ, Sneed JR, Roose SP. Biological aging and et al. Efficacy, safety, and tolerability of augmentation pharmacotherapy with
the future of geriatric psychiatry. J Gerontol A Biol Sci Med Sci. 2017;72:343–52. aripiprazole for treatment-resistant depression in late life: a randomised, dou-
218. McEwen BS, Karatsoreos IN. Sleep deprivation and circadian disruption: stress, ble-blind, placebo-controlled trial. Lancet. 2015;386:2404–12.
allostasis, and allostatic load. Sleep Med Clin. 2015;10:1–10. 243. Buchalter ELF, Oughli HA, Lenze EJ, Dixon D, Miller JP, Blumberger DM, et al.
219. Belvederi Murri M, Pariante C, Mondelli V, Masotti M, Atti AR, Mellacqua Z, et al. Predicting remission in late-life major depression: a clinical algorithm based
HPA axis and aging in depression: systematic review and meta-analysis. Psy- upon past treatment history. J Clin Psychiatry. 2019;80:18m12483.
choneuroendocrinology. 2014;41:46–62. 244. George D, Galvez V, Martin D, Kumar D, Leyden J, Hadzi-Pavlovic D, et al. Pilot
220. Gianaros PJ, Sheu LK, Uyar F, Koushik J, Jennings JR, Wager TD, et al. A brain randomized controlled trial of titrated subcutaneous ketamine in older
phenotype for stressor-evoked blood pressure reactivity. J Am Heart Assoc. patients with treatment-resistant depression. Am J Geriatr Psychiatry.
2017;6:e006053. 2017;25:1199–209.
221. Bagley SL, Weaver TL, Buchanan TW. Sex differences in physiological and affective 245. Lipsitz O, Di Vincenzo JD, Rodrigues NB, Cha DS, Lee Y, Greenberg D, et al.
responses to stress in remitted depression. Physiol Behav. 2011;104:180–6. Safety, tolerability, and real-world effectiveness of intravenous ketamine in older
222. Admon R, Holsen LM, Aizley H, Remington A, Whitfield-Gabrieli S, Goldstein JM, adults with treatment-resistant depression: a case series. Am J Geriatr Psy-
et al. Striatal hypersensitivity during stress in remitted individuals with recurrent chiatry. 2021;29:899–913.
depression. Biol Psychiatry. 2015;78:67–76. 246. Subramanian S, Lenze EJ. Ketamine for depression in older adults. Am J Geriatr
223. Zhong M, Wang X, Xiao J, Yi J, Zhu X, Liao J, et al. Amygdala hyperactivation and Psychiatry. 2021;29:914–6.
prefrontal hypoactivation in subjects with cognitive vulnerability to depression. 247. Ochs-Ross R, Daly EJ, Zhang Y, Lane R, Lim P, Morrison RL, et al. Efficacy and
Biol Psychol. 2011;88:233–42. safety of esketamine nasal spray plus an oral antidepressant in elderly patients
224. Ahrens T, Deuschle M, Krumm B, van der Pompe G, den Boer JA, Lederbogen F. with treatment-resistant depression-TRANSFORM-3. Am J Geriatr Psychiatry.
Pituitary-adrenal and sympathetic nervous system responses to stress in women 2020;28:121–41.
remitted from recurrent major depression. Psychosom Med. 2008;70:461–7. 248. Prudic J, Olfson M, Marcus SC, Fuller RB, Sackeim HA. Effectiveness of electro-
225. McEwen BS, Akil H. Revisiting the stress concept: implications for affective convulsive therapy in community settings. Biol Psychiatry. 2004;55:301–12.
disorders. J Neurosci. 2020;40:12–21. 249. Dols A, Bouckaert F, Sienaert P, Rhebergen D, Vansteelandt K, Ten Kate M, et al.
226. Geerlings MI, Sigurdsson S, Eiriksdottir G, Garcia ME, Harris TB, Gudnason V, et al. Early- and late-onset depression in late life: a prospective study on clinical and
Salivary cortisol, brain volumes, and cognition in community-dwelling elderly structural brain characteristics and response to electroconvulsive therapy. Am J
without dementia. Neurology. 2015;85:976–83. Geriatr Psychiatry. 2017;25:178–89.
227. Lupien SJ, de Leon M, de Santi S, Convit A, Tarshish C, Nair NPV, et al. Cortisol 250. Prudic J, Haskett RF, McCall WV, Isenberg K, Cooper T, Rosenquist PB, et al.
levels during human aging predict hippocampal atrophy and memory deficits. Pharmacological strategies in the prevention of relapse after electroconvulsive
Nat Neurosci. 1998;1:69–73. therapy. J ECT. 2013;29:3–12.
228. Cox SR, Bastin ME, Ferguson KJ, Maniega SM, MacPherson SE, Deary IJ, et al. 251. Kumar S, Mulsant BH, Liu AY, Blumberger DM, Daskalakis ZJ, Rajji TK. Systematic
Brain white matter integrity and cortisol in older men: the Lothian Birth Cohort review of cognitive effects of electroconvulsive therapy in late-life depression.
1936. Neurobiol Aging. 2015;36:257–64. Am J Geriatr Psychiatry. 2016;24:547–65.
229. Taylor WD, McQuoid DR, Payne ME, Zannas AS, MacFall JR, Steffens DC. Hip- 252. Tor PC, Bautovich A, Wang MJ, Martin D, Harvey SB, Loo C. A systematic review
pocampus atrophy and the longitudinal course of late-life depression. Am J and meta-analysis of brief versus ultrabrief right unilateral electroconvulsive
Geriatr Psychiatry. 2014;22:1504–12. therapy for depression. J Clin Psychiatry. 2015;76:e1092–8.
230. Wertz J, Caspi A, Ambler A, Broadbent J, Hancox RJ, Harrington H, et al. Asso- 253. Kellner CH, Husain MM, Knapp RG, McCall WV, Petrides G, Rudorfer MV, et al.
ciation of history of psychopathology with accelerated aging at midlife. JAMA Right unilateral ultrabrief pulse ECT in geriatric depression: phase 1 of the PRIDE
Psychiatry. 2021;78:530–9. study. Am J Psychiatry. 2016;173:1101–9.
231. Patel K, Abdool PS, Rajji TK, Mulsant BH. Pharmacotherapy of major depres- 254. Lisanby SH, McClintock SM, Alexopoulos G, Bailine SH, Bernhardt E, Briggs MC,
sion in late life: what is the role of new agents? Expert Opin Pharmacother. et al. Neurocognitive effects of combined electroconvulsive therapy (ECT) and
2017;18:599–609. venlafaxine in geriatric depression: phase 1 of the PRIDE study. Am J Geriatr
232. Tedeschini E, Levkovitz Y, Iovieno N, Ameral VE, Nelson JC, Papakostas GI. Psychiatry. 2020;28:304–16.
Efficacy of antidepressants for late-life depression: a meta-analysis and meta- 255. Kellner CH, Husain MM, Knapp RG, McCall WV, Petrides G, Rudorfer MV, et al. A
regression of placebo-controlled randomized trials. J Clin Psychiatry. novel strategy for continuation ECT in geriatric depression: phase 2 of the PRIDE
2011;72:1660–8. study. Am J Psychiatry. 2016;173:1110–8.
233. Calati R, Salvina Signorelli M, Balestri M, Marsano A, De Ronchi D, Aguglia E, et al. 256. McCall WV, Lisanby SH, Rosenquist PB, Dooley M, Husain MM, Knapp RG, et al.
Antidepressants in elderly: metaregression of double-blind, randomized clinical Effects of continuation electroconvulsive therapy on quality of life in elderly
trials. J Affect Disord. 2013;147:1–8. depressed patients: a randomized clinical trial. J Psychiatr Res. 2018;97:65–9.
234. Kok RM, Nolen WA, Heeren TJ. Efficacy of treatment in older depressed patients: 257. Trevizol AP, Goldberger KW, Mulsant BH, Rajji TK, Downar J, Daskalakis ZJ, et al.
a systematic review and meta-analysis of double-blind randomized controlled Unilateral and bilateral repetitive transcranial magnetic stimulation for
trials with antidepressants. J Affect Disord. 2012;141:103–15. treatment-resistant late-life depression. Int J Geriatr Psychiatry. 2019;34:822–7.
235. Karim HT, Andreescu C, Tudorascu D, Smagula SF, Butters MA, Karp JF, et al. 258. Liston C, Chen AC, Zebley BD, Drysdale AT, Gordon R, Leuchter B, et al. Default
Intrinsic functional connectivity in late-life depression: trajectories over the mode network mechanisms of transcranial magnetic stimulation in depression.
course of pharmacotherapy in remitters and non-remitters. Mol Psychiatry. Biol Psychiatry. 2014;76:517–26.
2017;22:450–7. 259. van Rooij SJH, Riva-Posse P, McDonald WM. The efficacy and safety of neuro-
236. Disabato BM, Morris C, Hranilovich J, D’Angelo GM, Zhou G, Wu N, et al. Com- modulation treatments in late-life depression. Curr Treat Options Psychiatry.
parison of brain structural variables, neuropsychological factors, and treatment 2020;7:337–48.

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
15
260. Jorge RE, Moser DJ, Acion L, Robinson RG. Treatment of vascular depression using 286. Donovan NJ, Wu Q, Rentz DM, Sperling RA, Marshall GA, Glymour MM. Lone-
repetitive transcranial magnetic stimulation. Arch Gen Psychiatry. 2008;65:268–76. liness, depression and cognitive function in older U.S. adults. Int J Geriatr Psy-
261. Overvliet GM, Jansen RAC, van Balkom A, van Campen DC, Oudega ML, van der chiatry. 2017;32:564–73.
Werf YD, et al. Adverse events of repetitive transcranial magnetic stimulation in 287. Perissinotto CM, Stijacic Cenzer I, Covinsky KE. Loneliness in older persons: a
older adults with depression, a systematic review of the literature. Int J Geriatr predictor of functional decline and death. Arch Intern Med. 2012;172:1078–83.
Psychiatry. 2021;36:383–92. 288. Van Orden KA. Addressing loneliness in older adults during the COVID-19
262. Kaster TS, Daskalakis ZJ, Noda Y, Knyahnytska Y, Downar J, Rajji TK, et al. Efficacy, pandemic: a commentary on “understanding psychological distress and pro-
tolerability, and cognitive effects of deep transcranial magnetic stimulation for tective factors among older adults during the COVID-19 pandemic”. Am J
late-life depression: a prospective randomized controlled trial. Neuropsycho- Geriatr Psychiatry. 2021;29:895–8.
pharmacology. 2018;43:2231–8. 289. Wilson RS, Krueger KR, Arnold SE, Schneider JA, Kelly JF, Barnes LL, et al.
263. Mosimann UP, Marre SC, Werlen S, Schmitt W, Hess CW, Fisch HU, et al. Anti- Loneliness and risk of Alzheimer disease. Arch Gen Psychiatry. 2007;64:234–40.
depressant effects of repetitive transcranial magnetic stimulation in the elderly: 290. d’Oleire Uquillas F, Jacobs HIL, Biddle KD, Properzi M, Hanseeuw B, Schultz AP,
correlation between effect size and coil-cortex distance. Arch Gen Psychiatry. et al. Regional tau pathology and loneliness in cognitively normal older adults.
2002;59:560–1. Transl Psychiatry. 2018;8:282.
264. Blumberger DM, Mulsant BH, Thorpe KE, McClintock SM, Konstantinou GN, Lee 291. Donovan NJ, Okereke OI, Vannini P, Amariglio RE, Rentz DM, Marshall GA, et al.
HH, et al. Effectiveness of standard sequential bilateral repetitive transcranial Association of higher cortical amyloid burden with loneliness in cognitively
magnetic stimulation vs bilateral theta burst stimulation in older adults with normal older adults. JAMA Psychiatry. 2016;73:1230–7.
depression: the FOUR-D randomized noninferiority clinical trial. JAMA Psy- 292. Chen JT, Wuthrich VM, Rapee RM, Draper B, Brodaty H, Cutler H, et al. Improving
chiatry. 2022;79:1065–73. mental health and social participation outcomes in older adults with depression
265. Ryff CD, Friedman EM, Morozink JA, Tsenkova V. Psychological resilience in and anxiety: Study protocol for a randomised controlled trial. PLoS ONE.
adulthood and later life: implications for health. Annu Rev Gerontol Geriatr. 2022;17:e0269981.
2012;32:73–92. 293. Tavares LR, Barbosa MR. Efficacy of group psychotherapy for geriatric depres-
266. Pfau ML, Russo SJ. Peripheral and central mechanisms of stress resilience. sion: a systematic review. Arch Gerontol Geriatr. 2018;78:71–80.
Neurobiol Stress. 2015;1:66–79. 294. Van Orden KA, Arean PA, Conwell Y. A pilot randomized trial of engage psy-
267. Laird KT, Krause B, Funes C, Lavretsky H. Psychobiological factors of resilience chotherapy to increase social connection and reduce suicide risk in later life. Am
and depression in late life. Transl Psychiatry. 2019;9:88. J Geriatr Psychiatry. 2021;29:789–800.
268. Laird KT, Lavretsky H, St Cyr N, Siddarth P. Resilience predicts remission in 295. Choi NG, Pepin R, Marti CN, Stevens CJ, Bruce ML. Improving social con-
antidepressant treatment of geriatric depression. Int J Geriatr Psychiatry. nectedness for homebound older adults: randomized controlled trial of tele-
2018;33:1596–603. delivered behavioral activation versus tele-delivered friendly visits. Am J Geriatr
269. Struijs SY, de Jong PJ, Jeronimus BF, van der Does W, Riese H, Spinhoven P. Psychiatry. 2020;28:698–708.
Psychological risk factors and the course of depression and anxiety disorders: a 296. Bruce ML, Pepin R, Marti CN, Stevens CJ, Choi NG. One year impact on social
review of 15 years NESDA research. J Affect Disord. 2021;295:1347–59. connectedness for homebound older adults: randomized controlled trial of tele-
270. Duckworth AL, Peterson C, Matthews MD, Kelly DR. Grit: perseverance and delivered behavioral activation versus tele-delivered friendly visits. Am J Geriatr
passion for long-term goals. J Pers Soc Psychol. 2007;92:1087–101. Psychiatry. 2021;29:771–6.
271. Windsor TD, Curtis RG, Luszcz MA. Sense of purpose as a psychological resource 297. Kahlon MK, Aksan N, Aubrey R, Clark N, Cowley-Morillo M, Jacobs EA, et al. Effect
for aging well. Dev Psychol. 2015;51:975–86. of layperson-delivered, empathy-focused program of telephone calls on lone-
272. Sowislo JF, Orth U. Does low self-esteem predict depression and anxiety? A liness, depression, and anxiety among adults during the COVID-19 pandemic: a
meta-analysis of longitudinal studies. Psychol Bull. 2013;139:213–40. randomized clinical trial. JAMA Psychiatry. 2021;78:616–22.
273. Laird KT, Lavretsky H, Paholpak P, Vlasova RM, Roman M, St Cyr N, et al. Clinical 298. Szymkowicz SM, Ryan C, Elson DM, Kang H, Taylor WD. Cognitive phenotypes in
correlates of resilience factors in geriatric depression. Int Psychogeriatr. late-life depression. Int Psychogeriatr. 2023;35:193–205.
2019;31:193–202. 299. Dotson VM. Can we promote cognitive resilience in late-life depression? Int
274. Cheng C, Lau HP, Chan MP. Coping flexibility and psychological adjustment to Psychogeriatr. 2023;35:171–3.
stressful life changes: a meta-analytic review. Psychol Bull. 2014;140:1582–607. 300. Morimoto SS, Wexler BE, Alexopoulos GS. Neuroplasticity-based computerized
275. Cuijpers P, Karyotaki E, Pot AM, Park M, Reynolds CF 3rd. Managing depression cognitive remediation for geriatric depression. Int J Geriatr Psychiatry. 2012;
in older age: psychological interventions. Maturitas. 2014;79:160–9. 27:1239–47.
276. Kiosses DN, Leon AC, Arean PA. Psychosocial interventions for late-life major 301. Morimoto SS, Gunning FM, Wexler BE, Hu W, Ilieva I, Liu J, et al. Executive
depression: evidence-based treatments, predictors of treatment outcomes, and dysfunction predicts treatment response to neuroplasticity-based computerized
moderators of treatment effects. Psychiatr Clin North Am. 2011;34:377–401. cognitive remediation (nCCR-GD) in elderly patients with major depression. Am
277. Shou H, Yang Z, Satterthwaite TD, Cook PA, Bruce SE, Shinohara RT, et al. J Geriatr Psychiatry. 2016;24:816–20.
Cognitive behavioral therapy increases amygdala connectivity with the cogni- 302. Gunning FM, Anguera JA, Victoria LW, Arean PA. A digital intervention targeting
tive control network in both MDD and PTSD. Neuroimage Clin. 2017;14:464–70. cognitive control network dysfunction in middle age and older adults with
278. Pinquart M, Duberstein PR, Lyness JM. Treatments for later-life depressive major depression. Transl Psychiatry. 2021;11:269.
conditions: a meta-analytic comparison of pharmacotherapy and psychother- 303. Oberlin LE, Jaywant A, Wolff A, Gunning FM. Strategies to promote cognitive health
apy. Am J Psychiatry. 2006;163:1493–501. in aging: recent evidence and innovations. Curr Psychiatry Rep. 2022;24:441–50.
279. Alexopoulos GS, Arean P. A model for streamlining psychotherapy in the RDoC 304. Cao W, Cao X, Hou C, Li T, Cheng Y, Jiang L, et al. Effects of cognitive training on
era: the example of ‘Engage’. Mol Psychiatry. 2014;19:14–9. resting-state functional connectivity of default mode, salience, and central
280. Alexopoulos GS, Raue PJ, Banerjee S, Marino P, Renn BN, Solomonov N, et al. executive networks. Front Aging Neurosci. 2016;8:70.
Comparing the streamlined psychotherapy “Engage” with problem-solving 305. Locke DEC, Khayoun R, Shandera-Ochsner AL, Cuc A, Eilertsen J, Caselli M, et al.
therapy in late-life major depression. A randomized clinical trial. Mol Psychiatry. Innovation inspired by COVID: a virtual treatment program for patients with
2021;26:5180–9. mild cognitive impairment at Mayo Clinic. Mayo Clin Proc Innov Qual Outcomes.
281. Fung HH, Carstensen LL, Lang FR. Age-related patterns in social networks among 2021;5:820–6.
European Americans and African Americans: implications for socioemotional 306. Rebok GW, Ball K, Guey LT, Jones RN, Kim HY, King JW, et al. Ten-year effects of
selectivity across the life span. Int J Aging Hum Dev. 2001;52:185–206. the advanced cognitive training for independent and vital elderly cognitive
282. Schwarzbach M, Luppa M, Forstmeier S, Konig HH, Riedel-Heller SG. Social training trial on cognition and everyday functioning in older adults. J Am Geriatr
relations and depression in late life-a systematic review. Int J Geriatr Psychiatry. Soc. 2014;62:16–24.
2014;29:1–21. 307. Szymkowicz SM, Taylor WD, Woods AJ. Augmenting cognitive training with
283. Woods A, Solomonov N, Liles B, Guillod A, Kales HC, Sirey JA. Perceived social bifrontal tDCS decreases subclinical depressive symptoms in older adults: pre-
support and interpersonal functioning as predictors of treatment response liminary findings. Brain Stimul. 2022;15:1037–9.
among depressed older adults. Am J Geriatr Psychiatry. 2021;29:843–52. 308. Nevitt MC, Cummings SR, Kidd S, Black D. Risk factors for recurrent nonsyncopal
284. Bosworth HB, McQuoid DR, George LK, Steffens DC. Time-to-remission from falls. A prospective study. J Am Med Assoc. 1989;261:2663–8.
geriatric depression: psychosocial and clinical factors. Am J Geriatr Psychiatry. 309. Seidler RD, Alberts JL, Stelmach GE. Changes in multi-joint performance with
2002;10:551–9. age. Mot Control. 2002;6:19–31.
285. King Johnson ML, Roche AI, Markon K, Denburg NL. Emotion regulation in older 310. Penninx BW, Ferrucci L, Leveille SG, Rantanen T, Pahor M, Guralnik JM. Lower
adulthood: roles of executive functioning and social relationships. Neuropsychol extremity performance in nondisabled older persons as a predictor of sub-
Dev Cogn B Aging Neuropsychol Cogn. 2023;30:336–53. sequent hospitalization. J Gerontol A Biol Sci Med Sci. 2000;55:M691–7.

Translational Psychiatry (2023)13:160


S.M. Szymkowicz et al.
16
311. Abellan van Kan G, Rolland Y, Andrieu S, Bauer J, Beauchet O, Bonnefoy M, et al. 338. Chen DS, Betz J, Yaffe K, Ayonayon HN, Kritchevsky S, Martin KR, et al. Asso-
Gait speed at usual pace as a predictor of adverse outcomes in community- ciation of hearing impairment with declines in physical functioning and the risk
dwelling older people an International Academy on Nutrition and Aging (IANA) of disability in older adults. J Gerontol A Biol Sci Med Sci. 2015;70:654–61.
Task Force. J Nutr Health Aging. 2009;13:881–9. 339. Brewster KK, Hu MC, Zilcha-Mano S, Stein A, Brown PJ, Wall MM, et al. Age-
312. White DK, Neogi T, Nevitt MC, Peloquin CE, Zhu Y, Boudreau RM, et al. Trajectories related hearing loss, late-life depression, and risk for incident dementia in older
of gait speed predict mortality in well-functioning older adults: the Health, Aging adults. J Gerontol A Biol Sci Med Sci. 2021;76:827–34.
and Body Composition study. J Gerontol A Biol Sci Med Sci. 2013;68:456–64. 340. Horowitz A, Brennan M, Reinhardt JP, Macmillan T. The impact of assistive
313. Verghese J, Holtzer R, Lipton RB, Wang C. Quantitative gait markers and incident device use on disability and depression among older adults with age-related
fall risk in older adults. J Gerontol A Biol Sci Med Sci. 2009;64:896–901. vision impairments. J Gerontol B Psychol Sci Soc Sci. 2006;61:S274–80.
314. Guralnik JM, Ferrucci L, Pieper CF, Leveille SG, Markides KS, Ostir GV, et al. Lower 341. Brody BL, Roch-Levecq AC, Kaplan RM, Moutier CY, Brown SI. Age-related
extremity function and subsequent disability: consistency across studies, pre- macular degeneration: self-management and reduction of depressive symp-
dictive models, and value of gait speed alone compared with the short physical toms in a randomized, controlled study. J Am Geriatr Soc. 2006;54:1557–62.
performance battery. J Gerontol A Biol Sci Med Sci. 2000;55:M221–31. 342. Rovner BW, Casten RJ, Hegel MT, Massof RW, Leiby BE, Ho AC, et al. Low vision
315. Montero-Odasso M, Schapira M, Soriano ER, Varela M, Kaplan R, Camera LA, et al. depression prevention trial in age-related macular degeneration: a randomized
Gait velocity as a single predictor of adverse events in healthy seniors aged 75 clinical trial. Ophthalmology. 2014;121:2204–11.
years and older. J Gerontol A Biol Sci Med Sci. 2005;60:1304–9. 343. Brewster K, Choi CJ, He X, Kim AH, Golub JS, Brown PJ, et al. Hearing rehabili-
316. Zheng JJ, Lord SR, Close JC, Sachdev PS, Wen W, Brodaty H, et al. Brain white tative treatment for older adults with comorbid hearing loss and depression:
matter hyperintensities, executive dysfunction, instability, and falls in older peo- effects on depressive symptoms and executive function. Am J Geriatr Psychiatry.
ple: a prospective cohort study. J Gerontol A Biol Sci Med Sci. 2012;67:1085–91. 2022;30:448–58.
317. Crockett RA, Hsu CL, Dao E, Tam R, Alkeridy W, Eng JJ, et al. Mind the gaps: 344. Brewster KK, Pavlicova M, Stein A, Chen M, Chen C, Brown PJ, et al. A pilot
functional networks disrupted by white matter hyperintensities are associated randomized controlled trial of hearing aids to improve mood and cognition in
with greater falls risk. Neurobiol Aging. 2022;109:166–75. older adults. Int J Geriatr Psychiatry. 2020;35:842–50.
318. Wegner M, Helmich I, Machado S, Nardi AE, Arias-Carrion O, Budde H. Effects of 345. Mulrow CD, Aguilar C, Endicott JE, Tuley MR, Velez R, Charlip WS, et al. Quality-
exercise on anxiety and depression disorders: review of meta-analyses and neu- of-life changes and hearing impairment. A randomized trial. Ann Intern Med.
robiological mechanisms. CNS Neurol Disord Drug Targets. 2014;13:1002–14. 1990;113:188–94.
319. Klil-Drori S, Klil-Drori AJ, Pira S, Rej S. Exercise intervention for late-life depres-
sion: a meta-analysis. J Clin Psychiatry. 2020;81:19r12877.
320. Rhyner KT, Watts A. Exercise and depressive symptoms in older adults: a sys- ACKNOWLEDGEMENTS
tematic meta-analytic review. J Aging Phys Act. 2016;24:234–46. This work was supported by National of Institute of Health grants R01 MH121619, R01
321. Firth J, Stubbs B, Vancampfort D, Schuch F, Lagopoulos J, Rosenbaum S, et al. MH121620, R01 MH121384, and R01 MH123662. SMS is additionally supported by
Effect of aerobic exercise on hippocampal volume in humans: a systematic NIH L30MH127619 and the McKnight Clinical Translational Research Scholarship in
review and meta-analysis. Neuroimage 2018;166:230–8. Cognitive Aging and Age-Related Memory Loss, funded by the McKnight Brain
322. Kilpatrick LA, Siddarth P, Milillo MM, Krause-Sorio B, Ercoli L, Narr KL, et al. Research Foundation through the American Brain Foundation and the American
Impact of Tai Chi as an adjunct treatment on brain connectivity in geriatric Academy of Neurology. ARG is supported by NIH T32MH019986.
depression. J Affect Disord. 2022;315:1–6.
323. Prehn K, Lesemann A, Krey G, Witte AV, Kobe T, Grittner U, et al. Using resting-
state fMRI to assess the effect of aerobic exercise on functional connectivity of
the DLPFC in older overweight adults. Brain Cogn. 2019;131:34–44. AUTHOR CONTRIBUTIONS
324. Belvederi Murri M, Amore M, Menchetti M, Toni G, Neviani F, Cerri M, et al. All authors (SMS, ARG, DH, and WDT) contributed to manuscript conceptualization
Physical exercise for late-life major depression. Br J Psychiatry. 2015;207:235–42. and writing, as well as critical review of the manuscript for important intellectual
325. Erickson KI, Hillman C, Stillman CM, Ballard RM, Bloodgood B, Conroy DE, et al. content. WDT additionally contributed to early versions of the scientific model and
Physical activity, cognition, and brain outcomes: a review of the 2018 physical supervision.
activity guidelines. Med Sci Sports Exerc. 2019;51:1242–51.
326. Northey JM, Cherbuin N, Pumpa KL, Smee DJ, Rattray B. Exercise interventions
for cognitive function in adults older than 50: a systematic review with meta- COMPETING INTERESTS
analysis. Br J Sports Med. 2018;52:154–60. The authors declare no competing interests.
327. Sanders LMJ, Hortobagyi T, la Bastide-van Gemert S, van der Zee EA, van
Heuvelen MJG. Dose-response relationship between exercise and cognitive
function in older adults with and without cognitive impairment: a systematic ADDITIONAL INFORMATION
review and meta-analysis. PLoS ONE. 2019;14:e0210036. Correspondence and requests for materials should be addressed to Warren D. Taylor.
328. Hall AJ, Febrey S, Goodwin VA. Physical interventions for people with more
advanced dementia—a scoping review. BMC Geriatr. 2021;21:675. Reprints and permission information is available at http://www.nature.com/reprints
329. Laird KT, Paholpak P, Roman M, Rahi B, Lavretsky H. Mind-body therapies for
late-life mental and cognitive health. Curr Psychiatry Rep. 2018;20:2. Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
330. Chou KL. Combined effect of vision and hearing impairment on depression in published maps and institutional affiliations.
older adults: evidence from the English Longitudinal Study of Ageing. J Affect
Disord. 2008;106:191–6.
331. Brewster KK, Ciarleglio A, Brown PJ, Chen C, Kim HO, Roose SP, et al. Age-related
hearing loss and its association with depression in later life. Am J Geriatr Psy- Open Access This article is licensed under a Creative Commons
chiatry. 2018;26:788–96. Attribution 4.0 International License, which permits use, sharing,
332. Simning A, Fox ML, Barnett SL, Sorensen S, Conwell Y. Depressive and anxiety adaptation, distribution and reproduction in any medium or format, as long as you give
symptoms in older adults with auditory, vision, and dual sensory impairment. J appropriate credit to the original author(s) and the source, provide a link to the Creative
Aging Health. 2019;31:1353–75. Commons license, and indicate if changes were made. The images or other third party
333. Casten RJ, Rovner BW. Update on depression and age-related macular degen- material in this article are included in the article’s Creative Commons license, unless
eration. Curr Opin Ophthalmol. 2013;24:239–43. indicated otherwise in a credit line to the material. If material is not included in the
334. Horowitz A, Reinhardt JP, Kennedy GJ. Major and subthreshold depression among article’s Creative Commons license and your intended use is not permitted by statutory
older adults seeking vision rehabilitation services. Am J Geriatr Psychiatry. 2005; regulation or exceeds the permitted use, you will need to obtain permission directly
13:180–7. from the copyright holder. To view a copy of this license, visit http://
335. Rutherford BR, Brewster K, Golub JS, Kim AH, Roose SP. Sensation and psy- creativecommons.org/licenses/by/4.0/.
chiatry: linking age-related hearing loss to late-life depression and cognitive
decline. Am J Psychiatry. 2018;175:215–24.
336. Heine C, Browning CJ. Mental health and dual sensory loss in older adults: a This is a U.S. Government work and not under copyright protection in the US; foreign
systematic review. Front Aging Neurosci. 2014;6:83. copyright protection may apply 2023
337. Vink D, Aartsen MJ, Schoevers RA. Risk factors for anxiety and depression in the
elderly: a review. J Affect Disord. 2008;106:29–44.

Translational Psychiatry (2023)13:160

You might also like