You are on page 1of 23

Frontiers in Neuroendocrinology 49 (2018) 8–30

Contents lists available at ScienceDirect

Frontiers in Neuroendocrinology
journal homepage: www.elsevier.com/locate/yfrne

Review article

Redefining neuroendocrinology: Epigenetics of brain-body communication T


over the life course

Bruce S. McEwen
Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Ave, New York, NY 10065, USA

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

Keywords: The brain is the central organ of stress and adaptation to stress that perceives and determines what is threa-
Stress tening, as well as the behavioral and physiological responses to the stressor, and it does so somewhat differently
Hippocampus in males and females. The expression of steroid hormone receptors throughout the brain has broadened the
Amygdala definition of ‘neuroendocrinology’ to include the reciprocal communication between the entire brain and body
Prefrontal cortex
via hormonal and neural pathways. Mediated in part via systemic hormonal influences, the adult and developing
Immune
brain possess remarkable structural and functional plasticity in response to stress, including neuronal replace-
Metabolic
Autonomic ment, dendritic remodeling, and synapse turnover. This article is both an account of an emerging field eluci-
Neuroplasticity dating brain-body interactions at multiple levels, from molecules to social organization, as well as a personal
Sex differences account of my laboratory’s role and, most importantly, the roles of trainees and colleagues, along with my
Social environment involvement in interdisciplinary groups working on this topic.
Jel classifications:
20
30
40
60
100
120
130
150
170
180
200
220

1. Prologue brother, Dr. Craig McEwen, a Sociologist at Bowdoin College, is ful-


filling for both of us and would have made our parents, George and
This article provides an overview of a rapidly emerging field as well Esther, very proud!
as a personal account of my laboratory’s contributions to it. I highlight
the roles of trainees and colleagues in elucidating brain-body interac- 2. Introduction
tions at multiple levels, from molecules to social organization. This
ongoing process was and is immeasurably enhanced by my scientific The discovery of steroid hormone receptors in brain regions that
colleague and wife, Dr. Karen Bulloch, a neuroimmumunologist, who, mediate virtually every aspect of brain function has broadened the
in addition to her novel and pioneering research on brain dendritic definition of “neuroendocrinology” to include the reciprocal commu-
cells, microglia and RNA editing summarized in Section 13, below, nication between the brain and the body via hormonal and neural
helped me to see and enjoy life beyond science and academics. pathways. Discovery of adrenal steroid receptors in hippocampus led
Furthermore, my recent and now ongoing collaboration with my the way and became the “gateway” for other discoveries and a better


Address: Alfred E. Mirsky Professor, Head, Harold and Margaret Milliken Hatch, Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY
10065, USA.
E-mail address: mcewen@mail.rockefeller.edu.
URL: http://www.rockefeller.edu/labheads/mcewen/mcewen-lab.php.

https://doi.org/10.1016/j.yfrne.2017.11.001
Received 29 July 2017; Received in revised form 1 November 2017; Accepted 4 November 2017
Available online 10 November 2017
0091-3022/ © 2017 Elsevier Inc. All rights reserved.
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

understanding of the meaning of “stress” in terms of the concepts of child/) provided a solution.
allostasis and allostatic load and overload where life style and health For starters, “good stress” involves our taking a chance on some-
behaviors are key factors along with stressful experiences themselves. thing one wants, like interviewing for a job or school, or giving a talk
Discovery of hormone receptors in brain has led to the discovery of before strangers, and feeling rewarded when we are successful.
structural and functional plasticity in the brain, mediated in part by “Tolerable stress” means that something bad happens, like losing a job
hormones, and this has helped to facilitate the emergence of the science or death of a loved one, but we have the personal resources and support
of “epigenetics” by revealing effects of the social and physical en- systems to weather the storm. “Toxic stress” means what Kagan refers
vironment on adult as well as developing brain structure and function. to, that is, something bad happens but we don’t have the personal re-
At the same time, cellular and molecular mechanisms for this plasticity sources or support systems, and, as a result, we lack a sense that we
have emerged and have revealed other mechanisms of steroid hormone have some control. As a result, we are “knocked for a loop” and may
action than direct genomic stimulation, including actions on mi- suffer mental and physical health problems of over time as a result,
tochondria as well as regulation of cellular signaling pathways. Besides particularly if the situation is not resolved.
steroid hormones, metabolic hormones enter and affect the brain and
their relationship to brain metabolism; and mitochondrial function has 5. Allostasis and allostatic load and overload
become important for understanding disorders like diabetes, depression
and dementia. Another important hormone discovery is the widespread Now let us put these three forms of “stress” into a biological and
effects of sex hormones throughout the brain along with diverse me- behavioral context. We know that “homeostasis” means the physiolo-
chanisms for sex hormone action at both genomic and non-genomic gical state which the body maintains to keep us alive - that is, body
levels, including actions in mitochondria. This has helped highlight the temperature and pH within a narrow range and adequate oxygen
need to understand sex differences in normal brain function and dis- supply. In order to maintain homeostasis, our body activates hormone
ease. Finally, hormone actions via epigenetic mechanisms operating secretion and turns on our autonomic and central nervous system (we
over the life course are changing the way we look at the development of call these “mediators” like cortisol, adrenalin, the immune system and
disorders and the possibilities for intervention. metabolism) to help us adapt, for example, when we get out of bed in
The story begins appropriately with “stress” since it was the dis- the morning, walk up a flight of stairs, have a conversation. These
covery of glucocorticoid receptors in hippocampus in 1968 (McEwen systems are also turned on when we are surprised by something un-
et al., 1968) that started this line of research for us. expected, or get into an argument, or run to catch a train. Some of these
experiences we may refer to as “stressful” but other we do not. So using
3. “Stress” is all around us! the word stress does not really recognize all of the underlying biology.
The “mediators” help us adapt as long as they are turned on in a
“Stress” is everywhere in our daily experiences and conversations balanced way when we need them and then turned off again when the
and yet, except for describing the “fight or flight” response most people challenge is over. When that does not happen, they can cause unhealthy
do not understand what is going on in the brain and body. We become changes in brain and body. This is also the case when the “mediators”
anxious hearing about violence, chaos and discord. And, in our rela- are not produced in an orchestrated and balanced manner – for ex-
tively secure world, the pace of our daily lives and the demands upon us ample, too much or too little cortisol or an elevated or too low blood
and our children often leads us to feel that there is too much to do in so pressure. When this happens and continues over weeks and months, we
little time! This disrupts our natural biological rhythms and encourages call it “allostatic load” (see Box 1) to refer to the wear and tear on the
unhealthy behaviors, like eating too much of the wrong things, ne- body that results from the chronic overuse and imbalance of the
glecting exercise and good sleep, and it results in an unhealthy life style. “mediators”. Accumulation of belly fat is an example as is the devel-
Poverty and racial and ethnic discrimination, and lack of educa- opment of chronic hypertension.
tional opportunities and economic advancement, take their toll on a When the wear and tear is strongest, we call it allostatic overload
large segment of our population where incarceration is the rule rather and this is what is occurring in toxic stress. An example is when hy-
than the exception for some of the most vulnerable. Adverse experi- pertension leads to coronary artery blockade, and the belly fat con-
ences in infancy and childhood, including poverty, leave a life-long tributes chemicals that accelerate the coronary artery blockade. Note,
imprint on the brain and body and undermine long-term health, in- however, that we are talking, not about one mediator, like cortisol, but
creasing the incidence of cardiovascular disease, diabetes, depression, a host of mediators that are all released in allostasis in a coordinated
substance abuse, anti-social behavior and dementia. How does all of manner to help us adapt but which can also cause damage when
this stress “get under our skin”, what does it do to our brains and our overused and dysregulated as described aboe. Because cortisol is well
body’s, how does it do it, and what can we do about it? known in relation to stress, its role is often misunderstood (see Box 2).

4. What is “stress”? 6. Health promoting and health damaging behaviors contribute to


allostatic load
Jerome Kagan has noted that the word “stress” is used so much in
different ways so as to be ambiguous and almost meaningless; and he We must go one step further and recognize that bad health beha-
suggested that the word “stress” be used only in the cases where an viors, often the result of a “stressful” life style, like eating too much of
event poses a serious threat to someone’s well-being (Kagan, 2016). He the wrong things, smoking, drinking, loneliness, poor sleep and lack of
also noted the “reluctance” of researchers to give up the term “stress”. exercise all contribute to that hypertension, belly fat and blockade of
While I agree that the word “stress” is ambiguous and overused, the coronary arteries. And they do so through the same “mediators” that
word is too embedded in human language and culture to be abandoned are activated to help us adapt but which also, when overused and
- and that includes researchers! Rather, my brother, Craig and I, re- dysregulated, cause allostatic load and overload (McEwen, 2006,
cently proposed in print (McEwen and McEwen, 2016) a more biolo- 2017).
gical way of looking at stress by, first, using additional words along So the “mediators” that help us adapt and enable us to maintain our
with “stress” and, second, putting that added wording into a biological homeostasis and survive can also contribute to well-know diseases of
and behavioral context. Fortunately, my participation in an inter- modern life. These ideas are the basis of the concepts of “allostasis” and
disciplinary network, The National Scientific Council for the Devel- “allostatic load/overload”, whereas they are not so obvious from the
oping Child, of which I am now a member (http://developingchild. word “stress” which is usually explained as the “fight or flight response”
harvard.edu/science/national-scientific-council-on-the-developing- when we are, for example, threatened by a mugger and run away. What

9
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Box 1
How An Interdisciplinary Research Network Created the Allostatic Load Concept.

In 1988, Peter Sterling and Joseph Eyer published a paper on the concept of allostasis as applied to the cardiovascular system (Sterling and
Eyer, 1988). Around that time, Eliot Stellar, then Provost at Penn and a collaborator on the topic of hormonal control of salt appetite in
brain, knew of our work on stress and invited me to become a member of a MacArthur Network on Health and Behavior where I heard
Sterling speak. A MacArthur Network is an interdisciplary group of social, behavioral and medical scientists interested in a common
problem. As my role in the network developed, Stellar and I wrote a paper that broadened the allostasis concept and introduced the notion
of cumulative change as a result of excessive or dysregulated allostasis (McEwen and Stellar, 1993) that was ultimately named “allostatic
load” (McEwen, 1998). The MacArthur Network “morphed” into the MacArthur Research Network on Socioeconomic Status and Health
that continued the development and validation of allostatic load with the important role of Teresa Seeman from UCLA (McEwen and
Seeman, 1999; Seeman et al., 2010b, 2010c; Wiley et al., 2016). The basic concept behind allostatic load is an outgrowth of Robert
Sapolsky’s “glucocorticoid cascade hypothesis” of stress and aging (Sapolsky et al., 1986) that was broadened, like that of Sterling and Eyer,
to encompass not only glucocorticoids but also other interacting mediators of adaptation. One more development of the allostatic load
concept was contributed by a field biologist, John Wingfield, who pointed out that animals in the wild are continually searching for calories
to stay alive while we humans, at least in affluent societies, are oversupplied with calories (McEwen and Wingfield, 2003). Moreover,
Wingfield pointed out that forms of what we might call “allostatic load” are important adaptive features in bears putting on body fat for the
winter, during which they burn it off. Yet, bears in a zoo can develop obesity associated with cardiovascular disease out of over-feeding and
boredom. This led to Wingfield’s suggestion that “allostatic overload” might apply to those health damaging aspects of dysregulated
allostasis (McEwen and Wingfield, 2003, 2010). Yet, migrating salmon and marsupial mice experience “allostatic overload” as a natural
part of the elimination of part of the population to provide resources to the next generation (Carruth et al., 2002; Poskitt et al., 1984).

really affects our health and well-being are the more subtle, gradual and these contribute to allostatic load and overload through the same bio-
long-term influences from our social and physical environment like logical “mediators” that help us adapt and keep us alive - and they
family and neighborhood chaos and conflict, demands of a job, shift shape our brains as well!
work and jet lag, sleeping badly, living in an ugly, noisy and polluted
environment, being lonely, not getting enough physical activity, eating
too much of the wrong foods, smoking, drinking too much alcohol. All

Box 2
What cortisol does and does not tell us!.

Even though we now know that there are multiple “mediators” for allostasis and allostatic load/overload, we often hear that measuring our
cortisol levels will tell us if we are stressed. This reflects a misunderstanding at two levels. First, a single measure of cortisol will tell us
nothing since cortisol levels go up and down within minutes. Moreover, we have noted the a day-night (diurnal) rhythm of this pulsating
cortisol that goes up in the early morning and wakes us up, then declines, except for a rise at lunch time, and which is normally low in the
evening before we go to bed. This is true unless, of course, “stressful” experiences occur or unless we are depressed or sleep deprived, or
both, when the diurnal rhythm is flat. There are several ways around the measurement problem – we can collect urine overnight or over the
day or we can measure cortisol in hair from the forehead, which gives as an even longer, over multiple days, index of our cortisol
production. Or we can measure cortisol in our saliva at multiple times during the day or before, during and after a stressful challenge, such
as talking about something personal before a group of strangers. The stressful challenge gives us a picture of the efficiency of our allostasis,
namely, turning on our cortisol when we are challenged and need it and then turning off our cortisol response when the stressor is over.
Failure to turn it on when needed is bad – other systems that balance it, like inflammation, may over-react; failure to turn it off when stress
is over produces negative effects on metabolism and other processes, like obesity, diabetes, depression and eventual heart disease, that
contribute to allostatic load/overload (McEwen, 1998).
The second misunderstanding is that the cortisol response is not the “bad guy”, as it is often implied by its elevation in the context of a
bad experience; rather, cortisol has a normal physiological role not only helping us adapt to stressors but also coordinating our metabolism
with our daily activity and sleep patterns (McEwen et al., 1993). We would not live very long or well without our cortisol! So let’s not blame
it for our problems! For example, we’ll see later that diurnal fluctuations of cortisol promote the formation and elimination of synapses in
our brains and this helps us learn and adapt (Liston et al., 2013; Liston and Gan, 2011). Moreover, the diurnal early morning rise of cortisol,
as well as a stress response activates adaptive immune function so that we can fight an infection or repair a wound. This grew out of the PhD
research of a former student, Firdaus Dhabhar, now Professor at the University of Miami. So an acute stress-induced rise of cortisol and
adrenalin actually helps the immune system to respond to a pathogen or repair a wound (Dhabhar and McEwen, 1997). Likewise, the
morning awakening rise of cortisol enhances the body’s response to an immunization (Dhabhar et al., 1994). Beside cortisol and adrenalin
involvement in this important adaptive function, key biochemicals of the immune system are also necessary and, together, they help
immune cells “go to their battle stations” where they are needed! (Dhabhar et al., 2012) The body’s response is like an orchestra involving
many players that need to work in harmony! But, indeed, too much cortisol also causes problems as it does with a flat diurnal rhythm and in
Cushing’s Disease when excess cortisol is produced by a pituitary gland tumor!
A third misunderstanding is that glucocorticoids do not play the same role across the life course but rather serve different functions,
starting with their role in “wakening” of the amygdala in neonatal life, from work of Regina Sullivan (Moriceau and Sullivan, 2004) to their
role in promoting ponderal growth in adolescence, based on work of Russell Romeo (Romeo et al., 2006) and their influence on the
vulnerability of the adolescent brain to stressful experiences (Pattwell et al., 2011, 2016; Romeo, 2017) as well as the age-accelerating role
of excess cortisol in aging (Lupien et al., 1998; Sapolsky et al., 1986).

10
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Box 3
Epigenetics.

“Epigenetics” originally meant the emergence of developmentally-programmed characteristics as a fertilized egg develops into a living
organism characteristic of that species (Waddington, 1942). This is programmed into each species, but the individual characteristics are
influenced by experiences, and that is where the modern use of “epigenetics” comes from. An example of this is a pair of identical twins
with genes that predispose them to schizophrenia or bipolar illness. Even with the same DNA, the probability that one twin will develop the
disease when the other twin gets it is only in the range of 40–60%, which leave plenty of room for experiences and other environmental
factors to either prevent of precipitate the disorder. As an indicator of this, the methylation patterns of DNA diverge as identical twins grow
older (Fraga et al., 2005). Thus, “epigenetics”, now meaning “above the genome”, that is, not changing the genetic code, replaces and
makes unnecessary the old question: “which is more important, genes or environment?”. The CpG methylation of DNA is now a well-known
form of epigenetic modification (Szyf et al., 2008). But there are other mechanisms that include histone modifications that repress or
activate chromatin unfolding (Allfrey, 1970) and the actions of non-coding RNA’s (Mehler, 2008), as well as transposons and retro-
transposons (Griffiths and Hunter, 2014) and RNA editing (Mehler and Mattick, 2007).

7. Central role of the brain epigenetics, meant something quite different (see Box 3). What Allfrey
and Mirsky did was to show how proteins called histones, which wrap
Up to this point I have left out the brain. The brain, “keeps the around DNA, are modified chemically to cause them to unwrap the
score” by storing memories from bad as well as good experiences, and DNA double helix and allow genes to be expressed. Around 1960,
yet the brain, working with the body, also “knows what to do” to keep hormones like cortisol and estradiol were shown by others to use this
us alive if we give it a chance by minimizing those subtle and long-term mechanism to turn on genes in the uterus and liver, and this became a
influences that cause allostatic load and overload (McEwen and Getz, focus of my work in 1966 after I was recruited back to Rockefeller to
2013). We call that the “wisdom of the body” and it refers back to work with a famous psychologist, Neal Miller, as his biochemist, having
“allostasis”, the active process of biological adaptation, and its role in spent 2 years in Sweden and a brief period at the University of Min-
maintaining homeostasis. Indeed the brain is a plastic and vulnerable nesota.
organ of the body, and is continually sculpted by experiences. The brain Learning about how hormones regulate gene expression in the
is also a vulnerable organ, along with our heart, liver and kidneys and uterus and liver led me to my interest in how hormones of the adrenals
other organs. The brain changes its architecture and function as part of and gonads alter gene expression in brain and produce their epigenetic
“allostasis” and the development of the notion of structural plasticity of effects. This led to my discovery, in the brain in 1968, that the adrenal
the brain has come about over the last half century with an accelerating stress hormone, cortisol, acts epigenetically on a brain structure called
pace as will be described below (see Box 4) ! We also need to consider the hippocampus, which we now know mediates memory of daily
where our genes fit in and understand that they do not rigidly de- events in space and time and also regulates mood. In fact, the hippo-
termine our destiny but, rather, provide the foundation on which our campus was the subject of the Nobel Prize into UK-based researcher
experiences shape our brains and bodies over the life course via “epi- Prof John O'Keefe and Norwegian scientists, May-Britt Moser and
genetic” mechanisms (Box 3). Edvard Moser in 2014 as the “GPS” of the brain.
Getting ahead of the story for the moment, the hippocampus is
8. How we started down this path: steroid hormone actions in important for episodic and spatial memory and is now also recognized
hippocampus and beyond for its role in mood regulation. For spatial memory, the hippocampus
becomes active in London cab drivers during functional MRI imaging
Though I did not think of it that way at the time, “epigenetics” has when they remember a route from one place to another (Maguire et al.,
been the core of my laboratory’s work on the brain and brain-body 1997) and the hippocampus is also important for food caching behavior
interactions. My mentors for my dissertation, in 1964, Vincent Allfrey in squirrels and birds (Biegler et al., 2001; Burger et al., 2013; Clayton,
and Alfred Mirsky, taught me the fundamentals of “epigenetics” in the 2001). The hippocampus is also “the canary in the coal mine” as far as
1960s before there was much interest in it, and when the name, conditions such as ischemia and seizures that cause brain damage as

Box 4
The Adult as Well as Developing Brain Shows Structural Plasticity.

Long regarded as a rather static and unchanging organ, except for electrophysiological responsivity, such as long-term potentiation (Bliss
and Lomo, 1973), the brain has gradually been recognized as capable of undergoing rewiring after brain damage (Parnavelas et al., 1974)
and also able to grow and change as seen by dendritic branching, angiogenesis and glial cell proliferation during cumulated experience
(Bennett et al., 1964; Greenough and Volkmar, 1973). More specific physiological changes in synaptic connectivity were also recognized in
relation to hormone action in the spinal cord (Arnold and Breedlove, 1985), and in environmentally directed plasticity of the adult songbird
brain (DeVoogd and Nottebohm, 1981). Seasonally varying neurogenesis in restricted areas of the adult songbird brain is recognized as part
of this plasticity (Nottebohm, 2002). Indeed, adult neurogenesis in the adult mammalian brain was initially described (Altman and Das,
1965; Kaplan and Bell, 1983) and then suppressed (Kaplan, 2001), only to be rediscovered in the dentate gyrus of the hippocampus
(Cameron and Gould, 1994; Gould and McEwen, 1993) in the context of studies of neuron cell death and actions of adrenal steroids and
excitatory amino acids in relation to stress. Neurogenesis in the dentate gyrus has gone on to become a huge topic related to effects of stress
(Gould et al., 1997), exercise (van Praag et al., 1999), enriched environment (Kempermann et al., 1997), antidepressants (Duman et al.,
1997) and learning and memory (Leuner et al., 2006) More than neurogenesis, structural plasticity includes dendrite remodeling, synapse
formation and synaptic pruning. For example, a recent study shows how the brain architecture of a mother is sculpted during pregnancy as
part of the formation of attachment to the child (Hoekzema et al., 2017). Moreover, a musician’s brain develop with enhanced size and
connections of sensory and motor control regions of the cerebral cortex! (Merzenich, 1998)

11
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

well as brain aging (Sapolsky, 1992). And the hippocampus responds to receptors (Ahima et al., 1991; Ahima and Harlan, 1990). Studies in our
sex hormones with effects on spatial memory and other functions laboratory as well as by Diamond and Joëls have shown biphasic effects
(Sandstrom and Williams, 2001), as will also be discussed later. mediated by MR and GR (Diamond et al., 1992; Joels, 2006; Pavlides
But we did not begin with the hippocampus. The initial discovery of et al., 1995). Ultradian fluctuations of glucocorticoids drive GR acti-
steroid hormone receptors in the brain was based on the model of cell vation and reactivation, while MR occupancy for nuclear activation is
nuclear steroid hormone receptors developed in the 1960s (Jensen and more constant and promotes excitability (Stavreva et al., 2009).
Jacobson, 1962; Toft and Gorski, 1966). Based upon the fundamental Another important advance was made by Robert Sapolsky who
discovery of the communication between hypothalamus and pituitary found that, over the lifespan of a rat, that the cortisol equivalent in the
by Geoffrey Harris (Harris, 1970) and isolation of releasing factors in rat, corticosterone, gradually cause a “wear and tear” on the hippo-
the hypothalamus for pituitary hormones (eg., (Guillemin, 1978; campus which impairs not only memory and mood but also the ability
Schally et al., 1973; Vale et al., 1981) the hypothalamus was the initial to shut off the production of its glucocorticoids (Sapolsky, 1992). This
target. The hypothalamus was also implicated in the control of re- effect is more evident in animals and also people who have experienced
productive behavior, and my colleague Don Pfaff’s pioneering work toxic stress. The “glucocorticoid-cascade hypothesis of stress and
elucidated the brain circuit for lordosis behavior in the female rat aging”, as it is called, was one of the bases for the concept of allostatic
(Pfaff, 1980). Using steroid autoradiography with tritium labeled es- load and overload (Box 1). Sapolsky also did seminal work on dominant
tradiol, putative cell nuclear receptor sites for estradiol were demon- and subordinate baboons in Africa and laid the groundwork for how
strated in hypothalamus (Pfaff and Keiner, 1973; Stumpf, 1971) and income, education and human social hierarchies impact physical and
also by cell fractionation and sucrose density gradient centrifugation by mental health. His books “Why Zebra’s Don’t Get Ulcers”, “A Primate’s
my first graduate student, Richard Zigmond (Zigmond and McEwen, Memoir” and “Behave”, and writings in the Wall Street Journal have
1970). We continued to collaborate with Don on molecular and neu- made him an important figure for public awareness of behavioral sci-
rochemical factor for lordosis for some years before we focused on the ence, along with his popularity as a teacher at Stanford University.
hippocampus and other extra-hypothalamic brain regions. More re- While Robert Sapolsky’s findings emphasized the role of gluco-
cently, we collaborated again on epigenetics and the transposon/ret- corticoids in neural damage and systemic pathophysiology, there was
rotransposons, as is described elsewhere (Hunter et al., 2013, 2012). another side to the story, namely, the role of glucocorticoids and other
hormones in adaptive plasticity of the adult as well as developing brain.
9. Two major Steps forward
10. Hormones and structural plasticity in brain
Then came the hippocampus which I had learned about as a post-
doctoral fellow in Goteborg Sweden in 1964–66 in the Institute of Up to this point in our work and in the field of neuroscience, the
Neurobiology headed by Holger Hyden. By administering 3H corticos- brain was regarded as structurally stable in adult life and the main focus
terone into adrenalectomized rats, we discovered receptors for adrenal for understanding normal and abnormal function was upon neu-
steroids in the hippocampal formation of the rat and, later, the rhesus rochemistry and neuropharmacology. That was the era when anti-
monkey (Gerlach and McEwen, 1972; Gerlach et al., 1976; McEwen and depressants like Prozac were introduced in 1985, along with an array of
Plapinger, 1970; McEwen et al., 1968). Other studies revealed such antipsychotic medications. Now there is a totally different view of
receptors in the hippocampal equivalent in other species including plasticity (see Box 4). Here is how our lab came into investigating this
birds (Dickens et al., 2009; McEwen, 1976). While there was very little topic. In 1988, Elizabeth Gould came to my laboratory as a postdoctoral
interest in 1968 in epigenetics in the brain, the hippocampus has since fellow and started us down the path of discovering structural plasticity
become a “gateway” into learning how stress hormones, and also sex, in the adult brain which includes spine synapse turnover, dendritc re-
thyroid and metabolic hormones like insulin, enter the brain, bind to modeling and dentate gyrus neurogenesis.
receptors and act epigenetically to positively regulate structure and Neurogenesis: Neurogenesis in the adult brain is now recognized as
function throughout many part of the brain and affect behavior and our an important feature of brain plasticity (Box 4). Gould with, at that
ability to function in our daily lives (McEwen et al., 2015b). But these time, students, Catherine Woolley and Heather Cameron, also estab-
“mediators”, as we have noted, also contribute to allostatic load and lished that neurons of the dentate gyrus part of the hippocampus die
overload under conditions of toxic stress and the brain is very much a and are replaced via neurogenesis that continues over the entire life
target of this, just as are the heart and other organs. course (Cameron and Gould, 1996; Gould et al., 1992). Kara Pham and
How my laboratory participated and, in some cases, initiated these Juan Nacher later found that toxic stress suppresses that neurogenesis
discoveries involves other remarkable students, postdoctoral fellows and shrinks the hippocampus (Pham et al., 2003), while other labora-
and colleagues, some of whom I shall now mention. Ron de Kloet, a tories went on to show that physical activity increases neurogenesis not
Professor at the University of Leiden, was one of my first postdoctoral only in young but also older animals (Kempermann and Gage, 1999;
fellows. He found that synthetic glucocorticoids like dexamethasone van Praag et al., 2005). These revelations about adult brain neuro-
(DEX) (cortisol is also a glucocorticoid) are actively excluded from the genesis have huge implications through the recognition that so-called
brain via the “multi-drug resistance p glycoprotein”, while cortisol gets stem or progenitor cells might be used to treat brain damage. And they
in!) Indeed, DEX is a very potent inhibitor of inflammation and immune also have meaning for lifestyle, in that regular physical activity in-
function and stimulates of liver glucose metabolism (hence the name creases this neurogenesis, in old as well as young, and improves
“glucocorticoid”). We now know that synthetic glucocorticoids like memory and mood and even enlarges the hippocampus, which tends to
DEX, when given to quell inflammation can shut of the ability to make shrink in depression, diabetes among other conditions (McEwen, 2007).
cortisol, and when DEX treatment is terminated the body and brain Within 6 months to a year, regular aerobic activity such as walking an
become deficient in cortisol and this results in terrible mood swings and hour a day 5 out of 7 days a week not only makes the hippocampus
metabolic and immune disruption. After that, Ron de Kloet went on in larger and improves memory (Erickson et al., 2011) but it also improves
his own laboratory with his student, Hans Reul, to show that cortisol in decision making by improving blood flow and metabolic function in the
the hippocampus binds to two receptor types, called MR and GR, to prefrontal cortex (Colcombe et al., 2004b; Kramer et al., 1999), a brain
produce its myriad of important actions. Work by Reul and de Kloet region essential for self-regulation of emotions and impulses as well as
demonstrated that there are two types of adrenal steroid receptors, decision making and working memory. Regarding sex differences, there
mineralocorticoid (Type 1 or MR) and glucocorticoid (Type 2 or GR), in is evidence that women’s executive processes may benefit more from
hippocampus and other brain regions (Reul and DeKloet, 1985). This exercise than men (Barha et al., 2017). The benefits of moderate phy-
was further elaborated by immunocytochemical mapping of the sical activity on the brain as well as the body cannot be

12
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Box 5
The Brain Can Generate Steroid Hormones from Cholesterol.

Besides progesterone generation by Schwann cells and oligodendrocytes (Schumacher et al., 2012), the brain is capable of making estradiol
and also androgens. Estradiol production “on demand” from cholesterol via aromatization in an ischemic brain has been documented (Hojo
et al., 2003), and knocking out aromatase in brain exacerbates the damaging effects of ischemia (McCullough et al., 2003). Furthermore,
after ovariectomy estrogen level are detectable after 3 weeks of gonadectomy in the male or female amygdala and in the female prefrontal
cortex but not in hippocampus, suggesting a local source (Barker and Galea, 2009). For androgens, the ability of exercise to stimulate
neurogenesis in male rat hippocampus is dependent on extra-gonadal production of dihydrotestosterone (Okamoto et al., 2012). The brain
also has the capacity to generate neuroactive steroids such as A-ring reduced metabolites of progesterone that are allosteric modulators of
the GABAa receptor that mediate its anaesthetic and sedative effects (Baulieu and Robel, 1990; Gee et al., 1987).

overemphasized! Indeed, regular physical activity is the most important that are non-genomic and involve the co-opting by the hormone of
behavior that one can do to maintain brain and body health! And, as a signallng pathways used in other cell types for other purposes (Woolley,
further illustration of brain-body communication, the ability of exercise 1999). Our colleague, Teresa Milner, Weill Cornell Medical College,
to stimulate neurogenesis requires at least two hormones to be taken up together with postdoctoral fellow Steve Alves, identified im-
from the body into the brain. One of them, IGF-1, comes from the liver munochemically and later by autoradiography the non-genomic sites
(Aberg et al., 2000; Trejo et al., 2001) and the other, cathepsin B, comes for estradiol receptor alpha in post-synaptic densities and spine appa-
from muscle (Moon et al., 2016). There are likely other mediators to be ratus (Milner et al., 2008, 2001). All this was preceded by work of Neil
discovered! MacLusky as a postdoctoral fellow, to show estradiol inducible pro-
Spine synapse turnover and dendritic remodeling: While neu- gestin receptors in hippocampus (MacLusky and McEwen, 1978) that
roscience research in the mid-to-late 1980s focused on the effects of turn out to be non-nuclear (Mitterling et al., 2010) and to mediate
gonadal and adrenal steroids on neurotransmitter and neuromodulator progesterone-induced down-regulation of spine synapses in hippo-
receptors, and we were involved in this through the work in our la- campus as seen in the estrous cycle (Woolley and McEwen, 1993).
boratory of William Rostene, Scott Mendelson, Anabelle Segarra, Estradiol treatment increases NMDA receptor binding and expres-
Christine Fischette, Lori Flanagan, Anita Danielsson, Anat Biegon, Jesus sion (Fischer et al., 2002; Weiland, 1992) and increases NMDA-medi-
Angulo, Alan Johnson, Bruce Nock, Maureen Gannon, Michael ated synaptic input (Woolley et al., 1997). In addition, blocking NMDA
Schumacher, Zoltan Sarnyai and Hector Coirini, we were about to receptors during estradiol treatment of ovariectomized rats prevents
embark on yet another new direction with the arrival of Elizabeth estradiol-induced spine synapse formation in the CA1 region of the
Gould (see Box 5). hippocampus (Woolley and McEwen, 1994). An example of tipping the
Elizabeth Gould introduced us to an old method introduced in the excitatory/inhibitory balance towards excitation to facilitate plasticity
late 1800s by Camelio Golgi, an Italian neuroanatomist who won a (Bavelier et al., 2010), the up-regulation of NMDA receptors in the CA1
Nobel Prize for it. The Golgi technique, when done right, allows the region of female rat hippocampus by estradiol is indirect, in that it is
investigator to visualize and measure the dendrites (like tree branches) blocked by M2 muscarinic receptor antagonists and mimicked, by-
and even the spines (sites of synapses with other neurons) on those passing estradiol, by elevating cholinergic activity (Daniel and
dendrites. Using the Golgi technique, Gould and her student, Catherine Dohanich, 2001) and this appears to involve disinhibition of inhibitory
Woolley, along with Yoshifumi Watanabe, showed that dendrites shrink GABAergic input to CA1 pyramidal neurons (Rudick et al., 2003). In
and spines synapses are lost on hippocampal neurons after chronic this connection it is important to note, as shown by Victorial Luine, that
stress lasting several weeks due, in part to the actions of glucocorticoids estradiol up-regulate choline acetyltransferase activity (Luine et al.,
(McEwen, 2016), while, in contrast, spine synapse come and go during 1975) as well as potassium-stimulated acetylcholine release (Gibbs
the estrous cycle of the female rat because of the fluctuations of the et al., 1997) and that they may do the latter via non-genomic ER α
ovarian hormone, estradiol and progesterone (Woolley and McEwen, receptors in cholinergic terminals (Towart et al., 2003). The up-reg-
1992). These findings have lead in two different directions in terms not ulation of NMDA receptors led to initiation of a cascade of filopodial
only of application but also underlying mechanisms. Thus, circulating formation (Yuen et al., 2011) and translation of PSD-95 mRNA in
hormones not only enter the brain and bind to receptors but also par- dendrites to form scaffolding for new synapses (Akama and McEwen,
ticipate with the brain’s own neurotransmitters in what we now call 2003; Znamensky et al., 2003). These signaling pathways involving
“adaptive plasticity”, which underlies behavioral and neurological BDNF, trkB, phosphorylated Akt and LIMkinase were carefully mapped
adaptation. For example, shrinkage of dendrite in hippocampus pro- across the estrous cycle by Joanna Spencer-Segal in BDNF val/66/val
tects those neurons from damage by over-stimulation during toxic stress and met/66/met mice (McEwen et al., 2012).
(McCall et al., 2013), and cyclic fluctuations of spine synapses during There are 3 types of estradiol receptors in the brain, and, as far as
the estrous cycle (and in the human menstrual cycle) underlie differ- the respective role of ER α, ER β and the G-protein coupled GPER1, ER
ences in behavior, including mood swings (Hara et al., 2015). Re- α ko mice fail to show E induced increases in spinophilin, a marker of
markably, in both cases, the hormones did not work alone and require, spine formation, while ER β ko mice allow some E up-regulation of
among other “mediators” the main neurotransmitter in the brain, glu- spinophilin, while both ERα and ERβ agonists increase spinophilin
tamate (see Box 6). I shall first discuss the effects of estradiol. immunoreactivity in mouse hippocampus (Spencer et al., 2008).
Estradiol, progestins and the brain outside of the hypothalamus: GPER1, formerly GPR30, also appears to be involved, according to work
Elizabeth Gould together with Catherine Woolley and Maya Frankfurt spearheaded by Beth Waters and Teri Milner; the GPER1 agonist, “G1,
discovered estradiol-induced spine synapse formation in hippocampus increased PSD95-IR in strata oriens, lucidum, and radiatum of CA3 in
along with ventromedial hypothalamus (Frankfurt et al., 1990; Gould ovariectomized mice 6 h after administration. In contrast, estradiol but
et al., 1990). The seminal work of Catherine Woolley as a graduate not G1 increased Akt phosphorylation levels. Thus, GPER1 actions in
student and in her academic career to date paved the way not only to the synapse may be due to interactions with synaptic scaffolding pro-
recognize synaptic turnover, first in hippocampus and then in cortex teins, such as SAP97. These results suggest that although estradiol’s
and other brain regions, dependent on estradiol and progestins, but also actions via GPER1 may converge on the same synaptic elements, dif-
to recognize mechanisms of estradiol and other steroid hormone actions ferent pathways are used to achieve these actions” (Waters et al., 2015).

13
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Box 6
Translation to human brain.

Recognition that hormones play an important role in modulating brain structure and function has opened the way to show how circulating
hormones contribute to brain disorders when their normal functions to promote adaptation go awry. Here are some additional examples
showing the value of translation and also reverse translation. Regarding glucocorticoids, Cushing’s Disease is a prominent example. In
Cushing’s disease, there are depressive symptoms along with a smaller hippocampus and memory problems that can be relieved by surgical
correction of the hypercortisolemia (Murphy, 1991; Starkman and Schteingart, 1981). However, there are residual effects associated with
the exposure to glucocorticoids in Cushing’s (Bas-Hoogendam et al., 2015; van der Werff et al., 2015) Both major depression and Cushing’s
disease are associated with chronic elevation of cortisol that results in gradual loss of minerals from bone and also can contribute to
abdominal obesity. In major depressive illness, as well as in Cushing’s disease, the duration of the illness and not the age of the subjects
predicts a progressive reduction in volume of the hippocampus, determined by structural magnetic resonance imaging (MacQueen et al.,
2003; Sheline et al., 1999; Starkman et al., 1992).
The hippocampus figures prominently, as there are a variety of other anxiety-related disorders, such as post-traumatic stress disorder
(PTSD) (Pitman, 2001; Vythilingam et al., 2002) and borderline personality disorder (Driessen et al., 2000), in which atrophy of the
hippocampus has been reported, suggesting that this is a common process reflecting chronic imbalance in the activity of adaptive systems,
such as the HPA axis, but also including endogenous neuro-transmitters, such as glutamate. Inflammation is also a factor related to many
disorders of modern life, and elevation of IL6 has been linked to reduced hippocampal volume (Marsland et al., 2008) as well as to poor
sleep (Friedman et al., 2005). Although there is little evidence regarding the effects of ordinary life stressors on brain structure, there are
indications from functional imaging of individuals undergoing ordinary stressors, such as counting backwards that there are lasting changes
in neural activity (Wang et al., 2005). Moreover, a 20 year history of chronic perceived stress has been linked in a cross-sectional study to
smaller hippocampal volume (Gianaros et al., 2007). Moreover, jet-lag and lack of adequate recovery is reported to lead to a smaller
temporal lobe, memory impairment and dysregulated and elevated cortiisol (Cho, 2001).
Brain regions besides the hippocampus, particularly the amygdala and prefrontal cortex, show altered patterns of activity in PET and
fMRI and also demonstrate changes in volume with recurrent depression: decreased volume of hippocampus and prefrontal cortex and
amygdala (Drevets et al., 1997; Sheline, 2003; Sheline et al., 1998, 1999). Interestingly, however, amygdala volume has been reported to
increase in the first episode of depression, whereas hippocampal volume is not decreased (Frodl et al., 2003; MacQueen et al., 2008).
Moreover, amygdala volume is greater in children living with a depressed mother for a decade (Lupien et al., 2011).
It has been known for some time that stress hormones, such as cortisol, are involved in psychopathology, reflecting emotional arousal
and psychic disorganization rather than the specific disorder per se (Sachar et al., 1973). But there is more than cortisol affecting the brain
and, in particular, metabolic hormones have many effects that alter systemic physiology and mental health and there is co-morbidity of
systemic disorders with mental health disorders (Tomasdottir et al., 2015). See Box 1. Toxic stress can increase anxiety by causing neurons
in the amygdala, a brain region controlling anxiety and aggression, to become larger; and, yet, a successful mindfulness stress reduction can
ameliorate that (Holzel et al., 2010), as. And regular physical activity, like walking every day, causes genesis of new neurons in the
hippocampus, a brain region that is essential for memory of daily events and spatial orientation; and it improves memory and also mood
(Erickson et al., 2011).

In contrast to the female hippocampus, where there is the complex at UC Davis, some in collaboration with us, has been very influential
interaction between cholinergic, GABAergic and glutamatergic neu- (Dumitriu et al., 2010; Hara et al., 2015, 2016). Likewise, the con-
ronal activity for the induction of NMDA receptor expression and sub- tributions by former postdoctoral fellow, Roberta Brinton, now at the
sequent estradiol-dependent spine synapse formation, the male hippo- University of Arizona, have opened new avenues for use of the hor-
campus shows androgen-induced NMDA receptor up-regulation that mone, progesterone, as a protective agent for the aging and a damaged
does not involve any androgen effect on the cholinergic system (Romeo brain (Wang et al., 2010a).
et al., 2005) and shows an androgen-induced spine formation that is Similarly, former postdoctoral fellow Michael Schumacher, now
abolished by fimbria-fornix transection implying the importance of head of an INSERM Unit (the French NIH) in Paris has found that the
some form of neuronal activity (Kovacs et al., 2003). cells in our brains that make myelin, the insulation of the brain, can
Focusing on estradiol-induced synapse formation, Susan Lee, Russ make the hormone, progesterone, and respond to it to make myelin
Romeo and Wayne Brake showed the effect of estradiol up-regulation of (Schumacher et al., 2012). He and a senior colleague, Professor Ale-
spine synapses in female hippocampus and a lack of that up-regulation jandro DeNicola from Buenos Aires, are investigating use of proges-
in males using radioimmunocytochemistry with antibodies to spino- terone and progesterone-like drugs to repair and regenerate myelin in
phillin (Lee et al., 2004). Before that, Wayne Brake had used that diseases like multiple sclerosis where there is loss of myelin
technique to demonstrate spine formation immunocytochemically (Labombarda et al., 2015). There seems to be no end to the uses and
(Brake et al., 2001); and Chenjian Li used radioimmunocytochemistry potential uses for these sex hormones that go well beyond reproduction
in mouse brain along with Golgi to look at spine density after estradiol and this is a topic we shall return to later.
treatment along with size and shape that showed an estradiol effect to Stress-Induced Structural Plasticity: Our demonstration of stress-
promote spine maturation (Li et al., 2004) In parallel, Steve Alves with induced remodeling of dendrites in hippocampal CA3 neurons provided
Brake, Victoria Luine and Russ Romeo showed that pharmacological a neuroanatomical mechanism that helped to explain behavioral effects
depletion of serotonin reduced spine density in the CA1 region of the rat of stress on memory and related processes (Conrad et al., 1996; Luine
hippocampus but did not prevent estradiol-induced increases in spine et al., 1994; McEwen, 1999, 2007; McEwen, 2016). These discoveries
density (Alves et al., 2002). have led to a cascade of investigations in stress neurobiology that have
The actions of estradiol on cognitive function and their absence increasing relevance to human mental and physical health. What these
after the menopause have become the focus of hormone therapy to slow findings helped to demonstrate is the remarkable feature of the adult as
cognitive aging and prevent Alzheimer’s disease, and the work on this well as developing brain, namely, recognition of its capacity for re-
topic by colleague John Morrison, now Director of the Primate Center modeling of dendrites, turnover of synapses and neurogenesis that

14
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Fig. 1. Inverted U-shaped dose response curve for ef-


fects of adrenal steroids interacting with excitatory
amino acids to affect brain plasticity and vulnerability
to damage.

began with the enriched environment studies on brain cortex thickness disappeared, showing the resilience, at least of the young adult brain!
(Bennett et al., 1964; Greenough et al., 1973) based on the work of Parallel studies on an animal model of perceived stress allowed Liston
Donald Hebb (Hebb, 1949). For stress-induced remodeling, which can and Jason Radley, now at the University of Iowa, working also with
be mimicked by chronic glucocorticoid treatment, excitatory amino John Morrison, to see shrinkage of neuronal dendrites and reduction of
acids and other cellular mediators are involved (McEwen, 1999, 2010). synapses in the prefrontal cortex that explained the deficits in cognitive
The work of Robert Sapolsky emphasized damaging aspects of gluco- flexibility (Liston et al., 2006; Radley et al., 2005).
corticoid action on hippocampus mediated also by excitatory amino To complete the present story of brain plasticity we need to describe
acids (Popoli et al., 2012; Sapolsky, 1992; Sapolsky et al., 1986). Put- what happens in the amygdala under the same stressors that cause
ting this together with stress induced neural remodeling, what emerged dendrites to shrink and synapses to be lost in the prefrontal cortex and
is an inverted U shaped dose-response curve (see Fig. 1) in which hippocampus, namely, that dendrites in the basolateral amygdala grow
physiological levels of glucocorticoids and excitatory amino acids op- and become more branched and, as a result, there is increased anxiety.
erate synergistically and beneficially to facilitate LTP and memory. This This was discovered by a scientific colleague and now a collaborator,
was shown by Constantine Pavlides at Rockefeller and also by David Sumantra Chattarji, a Professor at the National Centre for Biological
Diamond (Diamond et al., 1992; Pavlides et al., 1995) over a short time Sciences in Bangalore, India (Vyas et al., 2002). Conor Liston found that
frame of minutes to hours. Over a time frame of days to weeks, the same dendrites in the orbitofrontal part of the prefrontal cortex also expand
mediators promote dendritic remodeling that shows resilience and re- with chronic stress, and there is increased vigilance (Liston et al.,
covery in a healthy brain (McEwen, 1999). In contrast, acute traumatic 2006). In the short term, these changes may be adaptive, as anxiety and
events such as stroke, seizures and head trauma cause permanent da- vigilance are adaptive in a dangerous or uncertain environment; but, if
mage and neuron loss via synergy between glutamate and glucocorti- the threat passes and the behavioral state “gets stuck” and persists
coids (Conrad et al., 1999a; Sapolsky, 1992). along with the changes in neural circuitry, such maladaptation requires
Cheryl Conrad, whose work as a postdoctoral fellow showed that the intervention to open “windows of plasticity” with a combination of
novel antidepressant, tianeptine, prevented chronic stress induced CA3 pharmacological and behavioral therapies.
dendritic atrophy and spatial memory deficits while not preventing Again, regular physical activity, as described earlier, can strengthen
chronic stress enhancement of fear conditioning (Conrad et al., 1996, both prefrontal cortex and hippocampus control of the amygdala
1999b) went on at the Arizona State University to show, among other (Colcombe et al., 2004a). Another approach to chronic anxiety is
achievements, that conditions that cause CA3 dendrites to shrink make mindfulness based stress reduction (MBSR) that has been shown to
those neurons more vulnerable to excitotoxic damage (Conrad et al., reduce the size of the amygdala in those anxiety sufferers who are able
2007) while elevated CORT levels at the time of testing are responsible to reduce their anxiety by MBSR (Holzel et al., 2010). Both MBSR and
for the spatial memory impairment and not the CA3 atrophy per se meditation are gaining in popularity as a way of reducing anxiety and
apparently because of an altered sensitivity to CORT (Wright et al., thus reducing perceived stress, with increasing evidence of changes in
2006). the brain (McEwen, 2016).
The prefrontal cortex also responds to what we can call “tolerable Some of this research on traumatic stress also has implications for
stress”. During his MD-PhD thesis research, the same Conor Liston as- PTSD. In a collaboration with Shona Chattarji’s laboratory, we found
sessed perceived stress (how much or little they felt in control of their that a single, traumatic stressor causes new synapses to form in baso-
daily lives) in a group of medical students during their studies and we lateral amygdala with a delay of a week or so that is accompanied by a
found that those with the highest perceived stress were slower in doing gradual increase in anxiety (Mitra et al., 2005). This type of delay is a
a cognitive-flexibility test and also had slower functional connectivity feature of PTSD in people. What we have further shown with the
in a brain circuit involving the prefrontal cortex when tested in a Chattarji group is that a timed elevation of cortisol at the time or, or
functional MRI machine (Liston et al., 2009). The reason we can call shortly after, a traumatic stressor actually prevents the delayed increase
this “tolerable stress” is that, after a vacation, these impairments in amygdala synapses and anxiety-like behavior (Rao et al., 2012). Now

15
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

there is evidence for human PTSD that low cortisol at the time of 12. Glutamate overflow in depression and dementia
trauma, eg during open heart surgery or after a traffic accident, is a risk
factor and that elevating cortisol during or right after trauma can re- Not only is glutamate an essential excitatory transmitter throughout
duce later PTSD symptoms (Schelling et al., 2004; Zohar et al., 2011). the brain, but also dysregulated glutamate contributes to disorders such
See Box 6 for other examples of translation to the human brain. as major depression and also aging and dementia, as documented by
ongoing work in the McEwen laboratory (see Box 8).
Depression: An unregulated overflow of glutamate appears to play a
11. Psychosocial stressors provides a clue to mechanism role in depressive-like behavior in animal models in which shrinkage of
dendrites in the hippocampus and suppression of neurogenesis occurs.
Although glucocorticoids like cortisol in humans and corticosterone Carla Nasca arrived in our laboratory in 2012 and introduced us to
in rats and mice play an important role in brain structural remodeling acetyl-L-carnitine (LAC), a natural molecule that she showed rapidly
and neurochemistry, they are not working alone and require other up-regulates the metabotrophic glutamate receptor, mGlu2, via an
mediators and modulators that participate. Realization of this in my epigenetic mechanism involving acetylation of lysine 27 on histone H3;
laboratory came from an unusual direction, namely, the study of psy- this is mimicked by histone deacetylase (HDAC) inhibitors, which act in
chosocial stress in the rat visible burrow system, developed by Caroline a few days, and by SSRI’s which act more slowly (Nasca et al., 2013,
and Robert Blanchard, in which Randall Sakai, Christine McKittrick and 2015). Thus LAC is a putative rapidly-acting antidepressant candidate
Kellie Tamashiro played key roles (McEwen et al., 2015c). Chronic as shown in several animal models that have a deficiency of LAC with
social stress in this model, producing a dominant male and 4 sub- normal levels of carnitine (Bigio et al., 2016; Nasca et al., 2013). On-
ordinates led to changes in serotonin transporter (5HTT) binding, as going studies in human major depression, which now involve a pro-
well as shrinkage of dendritic arborization in both dominants and ductive collaboration with Natalie Rasgon of Stanford University, also
subordinates, compared to rats housed in ordinary group housed cages. shows that LAC deficiency may be a factor (unpublished). Moreover,
These changes occurred despite apparent differences in stressor severity LAC deficiency is also associated with insulin resistance that can be
experienced by the animals (greater in subordinates which had larger corrected by LAC supplementation (Bigio et al., 2016), as will be dis-
adrenals), suggesting that these changes may be part of the normal cussed further below (see Box 9).
adaptive response to chronic social stress (McEwen et al., 2015c). Yet, Aging and dementia: Unregulated glutamate overflow is also im-
because of the greater dendritic shrinkage in dominants compared to plicated in aging and dementia. During aging in the rat, treatment with
subordinates, in spite of the larger adrenals in subordinates, these riluzole, which is known to retard glutamate release and promote glu-
findings took us away from simplistic idea that corticosterone (CORT) tamate reuptake by astrocytes, retards aging in the hippocampus as
caused it all, and it also turned attention to the existence of other measured by preservation of spatial memory and thin spines that are
mediators and processes synergisitic with glucocorticoid functions. In- found in young hippocampal neurons, as shown by Cognitive
deed, the mechanism for dendritic shrinkage has subsequently been Neurologist, Ana Pereira, in collaboration with the John Morrison la-
shown to involve multiple interacting mediators, with excitatory amino boratory (Pereira et al., 2014). Moreover, assessing gene expression
acid receptors playing a major role, along with other mediators such as changes associated with aging in rodents using RNA-sequencing (RNA-
brain derived neurotrophic factor (BDNF), tissue plasminogen activator seq), riluzole prevented many of the hippocampal age-related gene
(tPA), corticotrophin releasing hormone (CRH) and others (see Table 1, expression changes. Moreover, a comparison of the effects of riluzole in
Box 7 and Fig. 2). rats against human AD data sets revealed that many of the gene changes
in AD are reversed by riluzole (Pereira et al., 2016). Glutamate also
plays a role in plasticity in the prefrontal cortex and amygdala, de-
Table 1
scribed above. Dr. Pereira, a Clinical Assistant Professor associated with
Molecules that are necessary/permissive for remodeling.
the McEwen lab is conducting a clinical trial of early Alzheimer’s Dis-
A. Secreted signaling molecules ease with riluzole. Because of her interest in etiologies of dementia, she
* BDNF - brain derived neurotrophic factor (Bath et al., 2013; Chen et al., 2006; has conducted a study of 33 older adults, average age 64 years, and
Govindarajan et al., 2006) show that levels of the inhibitory neurotransmitter γ-aminobutyric acid
- Facilitator of plasticity or growth; FLOOR AND CEILING
(GABA) in the left dorsolateral prefrontal cortex (DLPFC), a brain re-
* CRF – corticotrophin releasing factor (Chen et al., 2016)
- Down-regulates thin spines via RhoA signaling gion that controls executive function, were lower in people with mod-
* tPA - tissue plasminogen activator (Bennur et al., 2007; Matys et al., 2004; Pawlak erate to severe sleep disordered breathing (SDB) than in controls; this
et al., 2003, 2005) correlated with minimum oxygen saturation and severity of disease,
- required for stress-induced spine loss in CA1 hippocampus and medial
suggesting that SDB-induced hypoxia could reduce DLPFC GABA levels,
amygdala
- CRF regulates tPA release.
leading to neuronal hyperexcitability, neuronal damage and cognitive
* Lipocalin-2 - secreted protein (Mucha et al., 2011; Skrzypiec et al., 2013) decline (Pereira et al., 2017).
- Acute stress induces Lipocalin-
- Lipocalin-2 ko increases neuronal excitability and anxiety 13. Microglia and brain dendritic cells
* Endocannabinoids (Gunduz-Cinar et al., 2013; Hill et al., 2013, 2010; Hill and
McEwen, 2010)
- Induced via glucocorticoids Astrocytes and their role in glutamate reuptake and oligoden-
- Regulate emotionality and HPA habituation and shut off drocytes and their role in myelin formation are complemented in the
- Buffer against stress induced remodeling brain by “microglia” that are related to the circulating monocytes of the
B. Cell surface and nucleo-cytoplasmic interaction immune system, a subset of which are brain dendritic cells (D'Agostino
*PSA-NCAM – cell surface “anti-stickiness” (McCall et al., 2013; Nacher et al., 2013; et al., 2012a). Karen Bulloch’s Neuroimmune and Neuroinflammation
Rutishauser, 2008; Sandi, 2004) Program at Rockefeller began with a study of the role of microglia in
- Endonuclease N removes PSA from NCAM; dendrite expansion
- Facilitate plasticity but also limits it.
the increased inflammation that occurs with aging in the brain (Sierra
* Cell adhesion molecules: neuroligin-2; nectin-3 (van der Kooij et al., 2014a, 2014b) et al., 2007). Using a mouse with a transgene expression in peripheral
- Chronic stress disrupts neuroligin-neurexin interaction dendritic cells, she and her colleagues went on to demonstrate the
- Chronic stress reduces nectin-3 via MMP9 protease. presence of dendritic-like cells in the developing and adult brain
* Nuclear pore complex NUP-62 (Kinoshita et al., 2014)
(Bulloch et al., 2008) that increase with age (Kaunzner et al., 2012) and
- Reduction leads to dendrite shrinkage
- Possibly due to nuclear-cytoplasmic communication after stroke damage (Felger et al., 2010) and show unique properties in
the pituitary gland (Glennon et al., 2015). They also found that virally-

16
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Box 7
Central Role of Glutamate and Other Mediators in Plasticity and Damage.

Excitatory amino acids, particularly glutamate, play a key role in structural as well as functional changes in the brain since glutamate is the
major excitatory transmitter, while, at the same time, excess glutamate causes damage and inflammation (McEwen, 1999). Using chronic
corticosterone treatment and, separately, repeated restraint stress, Yoshi Watanabe and Gould, caused shrinkage of apical dendrites of
hippocampal CA3 neurons that could be blocked by phenytoin, implicating glutamate (McEwen, 2016). Indeed, acute restraint stress
elevates extracellular glutamate levels via a process that is blocked in adrenalectomized animals, implicating the adrenal cortex and
possible direct effects of adrenal steroids on glutamate release (Lowy et al., 1993). Indeed, corticosterone was subsequently shown to act
directly via membrane associated MR and GR to cause glutamate release (Karst et al., 2005; Treccani et al., 2014). Blocking NMDA
receptors and interfering with excitatory stimulation of ion channels blocks stress induced dendritic remodeling, as also does blockade of
adrenal steroid synthesis (Magarinos and McEwen, 1995; Watanabe et al., 1992). A stress-induced NMDA-dependent dendritic remodeling
has also been reported in medial prefrontal cortex neurons (Martin and Wellman, 2011).
Besides glutamate, there are other mediators that participate in structural and functional remodeling of brain circuits with experience,
as summarized in Table 1. These included ongoing studies of BDNF with our collaborators Francis Lee (Weill Cornell) and Kevin Bath
(Brown), past investigations of tissue plasminogen activator and lipocalin 2 with Robert Pawlak and Sid Strickland and Shona Chattarji and
extensive investigations of endocannabinoids with Matt Hill and Cecilia Hillard, as well as the important studies by Tallie Baram and
colleagues at UC Irvine.
Moreover, Table 1 shows a number of cellular processes are involved in dendritic remodeling including cytoskeletal depolymerization
caused by a soluble phosphorylated tau (Arendt et al., 2003) and stress-induced reduction in expression of a cell nuclear pore complex
protein (Kinoshita et al., 2014), suggesting that ongoing gene expression may be involved in maintaining the dendritic tree. Excess glu-
tamatergic activity, without adequate reuptake in the aftermath of trauma from seizures, ischemia and head trauma, leads to permanent
neuronal loss by a process that is exacerbated by glucorticoids (Sapolsky et al., 1986).
These relationships can be summarized in an inverted U-shaped dose and time response curve in Fig. 1.

varies over the circadian cycle and it mediated by nuclear receptor


REV-ERBalpha (Gagnidze et al., 2016).
Karen Bulloch’s group established a collaboration with Nina
Papavasiliou at Rockefeller on the RNA editing enzyme, APOBEC1, and
this has resulted in a remarkable set of findings. Microglia (MG), a
heterogeneous population of phagocytic cells, aid in maintaining
homeostasis by producing anti-inflammatory cytokines and neuro-
trophic factors, supporting myelin production, removing synapses and
cellular debris as well as participating in “cross correction”, a process
that supplies neurons with key factors for executing autophagy-lyso-
somal function. As sentinels for the immune system, MG also detect
“danger” signals (pathogenic or traumatic insult), become activated,
produce pro-inflammatory cytokines, and recruit monocytes and den-
dritic cells to the site of damage through a breached blood brain barrier
or via brain lymphatics. Failure to effectively resolve MG activation can
Fig. 2. Gene expression hippocampus after a bolus of corticosterone compared to the
be problematic and lead to chronic inflammation, a condition proposed
effects of a novel acute forced swim (FST) in naïve, chronically restrained (CRS) and to underlie CNS pathophysiology in heritable brain disorders and age-
recovered (Rec after CRS) showing largely unique gene expression responses, indicating related neurodegenerative and cognitive decline. They have shown that
that the brain is continually changing with experience. There is a set of genes always APOBEC1-mediated RNA editing occurs within microglia and is key to
activated by FST (e.g., immediately early genes). From Gray et al. (2014) with permis- maintaining their resting status (Rayon-Estrada et al., 2017). Like bone
sion.
marrow-derived macrophages, RNA editing in MG leads to overall
changes in the abundance of edited proteins which coordinate and
induced encephalitis initiates distinct and functional CD103+ CD11b+ harmonize the function of multiple cellular pathways. Conversely, mice
brain dendritic cell populations within the olfactory bulb (D'Agostino lacking the APOBEC1 editing function in MG, display evidence of
et al., 2012b) and that the mortality of virally-induced encephalitis dysregulation, with progressive, age-related signs of

Box 8
Non-genomic Mechanisms of Steroid Action.

After the initial focus on cell nuclear actions of steroid hormones in brain, it became evident that there are other mechanisms of steroid
hormone action (Kelly and Levin, 2001; Levin and Hammes, 2016; Liposits and Bohn, 1993; Norman et al., 1999; Wehling, 1997). Besides
the non-genomic actions of estradiol on synapse formation via pAkt and pLIMK, there are non-genomic actions of adrenal steroids that
figure prominently in the findings described herein. For MR and GR, membrane associated localizations of GR and MR are linked to the
direct stimulation and regulation of excitatory amino acid release (Karst et al., 2005; Tang et al., 2011; Treccani et al., 2014). One of the
first examples of a rapid non-genomic action of glucocorticoids was the rapid activation of tryptophan hydroylase for serotonin formation
by Efrain Azmitia, as a graduate student in the McEwen laboratory (Azmitia and McEwen, 1968). As a postdoctoral fellow, Anat Biegon
explored non-genomic mechanisms for estradiol to affect serotonin and noradrenalin receptors in the brain (Biegon et al., 1983) There is
also an important feed-forward cycle between glucocorticoids and trkB, the receptor for BDNF (McEwen, 2015) as summarized in Fig. 3.

17
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Fig. 3. Glucocorticoids produce direct and indirect


genomic actions as well as nongenomic signaling ac-
tions via glucocorticoid (GR) and mineralocorticoid
(MR) receptors. These involve not only direct and in-
direct genomic actions, but also direct stimulation of
glutamate release and stimulation of endocannabinoid
production, which then feed back on glutamate and
GABA release and actions in mitochondria to affect Ca
++ buffering and free radical formation (Popoli et al.,
2012) (Upper Right). As shown in the article by
Arango-Lievano et al. (2015), BDNF, in the presence of
glucocorticoids, phosphorylates the GR at sites that
facilitate its translocation to the cell nucleus for tran-
scriptional actions; this effect is synergistic with the
ability of glucocorticoids to activate, via a genomic
mechanism, the phosphorylation of the Trk B receptor
independently of BDNF, thus creating a positive feed-
back loop. From McEwen (2015) with permission.

neurodegeneration, characterized by clustering of activated microglia, and stress on gene expression in the brain. Recent technological ad-
aberrant myelination, increased inflammation and lysosomal anomalies vances have allowed high-throughput analysis of gene expression
that culminate in behavioral and motor deficiencies, as in so-called changes in response to stress (Rubin et al., 2014). For example, work by
lysosomal storage diseases (Cole et al., 2017). current postdoctoral fellow, Jason Gray, using a microarray analysis of
whole hippocampus after acute stress, chronic stress, and stress re-
14. Gene expression continually changes with experience covery in mice revealed that acute and chronic stress modulate a core
set of genes, but that numerous changes are exclusive to each condition,
As the first extra-hypothalamic brain structure recognized to have highlighting how duration and intensity of stress alters reactivity (Gray
receptors for adrenal steroids (McEwen et al., 1968), the hippocampus et al., 2014). Furthermore, corticosterone injections do not yield the
is an important gateway for understanding the effects of glucocorticoids same expression profile as acute stress, suggesting that in vivo stressors

Box 9
Metabolic Hormones Enter the Brain and Affect Many Functions.

See Fig. 4. Metabolic hormones are transported into the brain and have multiple functions in the healthy brain. Besides glucocorticoids and
excitatory amino acids, a number of protein hormones have been shown to affect the hippocampus. The hippocampus has receptors for IGF-
1 and insulin and it responds to circulating insulin to translocate glucose transporters to cell membranes (Dore et al., 1997; Piroli et al.,
2007). Circulating IGF-1 is a key mediator of the ability of physical activity to increase neurogenesis in the dentate gyrus of the hippo-
campal formation (Aberg et al., 2000; Carro et al., 2001). IGF-1 is taken up into brain via a transport system different from that which
transports insulin, although there is some overlap (Banks et al., 2012; Pulford and Ishii, 2001; Yu et al., 2006). IGF-1 is a member of the
growth hormone family, and growth hormone is implicated in cognitive function and mood regulation (Donahue et al., 2006; Nyberg,
2000). Growth hormone is expressed in the hippocampus where it is up-regulated by acute stress and also, in females, by estradiol
(Donahue et al., 2006). Interestingly, although growth hormone mRNA is expressed in hippocampus (Donahue et al., 2002) and growth
hormone also enters the brain in small amounts from the circulation, although not by a specific transport system (Pan et al., 2005).
Furthermore, circulating ghrelin, a pro-appetitive hormone, has been shown to increase synapse formation in hippocampal pyramidal
neurons and to improve hippocampal-dependent memory (Diano et al., 2006). Ghrelin is transported into brain via a saturable system
(Banks et al., 2002) and receptors for ghrelin are expressed in hippocampus, as well as in other regions of the brain (Zigman et al., 2006).
Another metabolic hormone, leptin, has been found to exert antidepressant effects when infused directly into the hippocampus (Lu et al.,
2006). Leptin is transported into the brain, and both glucose and insulin mediate the ability of fasting to increase leptin transport into the
brain (Kastin and Akerstrom, 2001). Leptin receptors are found in hippocampus among other brain regions and leptin has actions in
hippocampus, via excitatory amino acid receptors, that reduce the probability of seizures and enhance aspects of cognitive function
(Harvey et al., 2005).

18
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Fig. 4. Metabolic hormones are transported into the


brain and have multiple functions in the healthy brain.
From McEwen (2007).

activate a diverse set of pathways independent of glucocorticoid re- 2013).


ceptor (GR) activation (Gray et al., 2014). Finally, characterization of
expression profiles after extended recovery from 21 days of chronic 15. Mitochondria have their special place in plasticity and
stress showed that, despite a normalization of anxiety-related beha- damage and hormone action
viors, recovery did not represent a return to the stress-naïve baseline,
but rather represents a new state in which reactivity to a novel stressor Mitochondria have emerged as major players in steroid hormone
produces a unique expression profile (Gray et al., 2014). actions and containment of excitotoxicity because of their ability to
Studies in rats confirm that gene expression profiles can vary sig- generate free radicals and to sequester Ca++ ions to contain that
nificantly from the immediate end of stress to 24 h later (Wang et al., process, as well as to express genes from its own genome and receive
2010b), and that chronic stress can alter the transcriptional response to nuclear coded genes that, together, regulate a host of important cell
an acute corticosterone injection in dentate gyrus, as showing in a functions (Picard et al., 2014). Mitochondria are also known to respond
collaboration with Dutch scientists, Nicole Datson, and Ron de Kloet to glucocorticoid receptors translocated into them (Moutsatsou et al.,
(Datson et al., 2013). Together, these studies demonstrate that a history 2001). In the McEwen lab this interest began with a collaborative study
of stress exposure can have a lasting impact on future stress reactivity with Richard Hunter in my lab and Jing Du and Husseini Manji at NIMH
and hippocampal function. It seems logical to assume that this gen- that showed a biphasic effect of glucocorticoids on mitochondria
eralizes to all experiences that we have, whether or not we call them function in which physiological levels of glucocorticoids promote Ca+
“stress”. + sequestration, moderate oxidation and maintain mitochondrial
Histone modifications are keys to epigenetic regulation of gene membrane potential while supra-physiological glucocorticoid levels
expression, Besides the acetylation of histones involved in the up-reg- start to do so but fail after 24 h and lead to a failure of all three mea-
ulation of mGlu2 gene expression described above, repressive epige- sures (Du et al., 2009). Hunter went on to more recently locate the site
netic modifications of histones are also evident after acute and chronic on the mitochondrial circular DNA where glucocorticoid receptors exert
stress, as shown by Richard Hunter, now a faculty member at U Mass their transcriptional effects (Hunter et al., 2016).
Boston. Acute stress dramatically increased the levels of H3K9 tri-me- Martin Picard, while at the University of Pennsylvania as a post-
thylation (H3K9me3) in the dentate gyrus (DG) and CA1, while chronic doctoral fellow, introduced us to mitochondrial dysfunction on the
restraint stress (CRS) for 21 days abolished this effect. Treatment with premise that the experience of psychological stress triggers neu-
fluoxetine during CRS reversed the decrease in DG H3K9me3 (Hunter roendocrine, inflammatory, metabolic, and transcriptional perturba-
et al., 2009). To dig deeply into the substantial, regionally specific, tions that ultimately predispose to disease via allostatic load/overload.
increase in hippocampal levels of the repressive histoneH3 lysine 9 To examine the role of mitochondria, using the hypothesis that ab-
trimethylation (H3K9me3), Hunter used ChIP coupled with next gen- normal mitochondrial functions would differentially modulate the or-
eration sequencing (ChIP-Seq) to determine the genomic localization of ganism’s multisystemic response to psychological stress, he uses mice
the H3K9me3 response. We found that acute stress increases in with mutations or deletions of key mitochondrial genes to selectively
H3K9me3 trapped and therefore repressed expression enrichment at impair mitochondrial respiratory chain function, energy exchange, and
transposable element loci and, using RT-PCR, we demonstrate that this mitochondrial redox balance in mice. In a collaborative study we found
effect represses expression of intracisternal- A particle endogenous that mitochondrial dysfunctions altered the hypothalamic– pituitar-
retrovirus elements and B2 short interspersed elements, but it does not y–adrenal axis, sympathetic adrenal–medullary activation and ca-
appear to have a repressive effect on long interspersed element RNA. In techolamine levels, the inflammatory cytokine IL-6, circulating meta-
addition, the histone H3K9-specific methyltransferases Suv39h2 is up- bolites, and hippocampal gene expression responses to stress. Each
regulated by acute stress in the hippocampus, and that this may explain mitochondrial defect generated a distinct whole-body stress response
the hippocampal specificity (Hunter et al., 2012). This response may signature. Thus mitochondria are stress-response modulators, with
represent a genomic stress response aimed at maintaining genomic and implications for understanding the mechanisms of stress pathophy-
transcriptional stability in vulnerable brain regions such as the hippo- siology and mitochondrial diseases (Picard et al., 2015). And it is also
campus, although the transposome might have adaptive functions at the important not to ignore estradiol which regulates mitochondrial oxi-
level of both evolution and the individual organism (Hunter et al., dation, since estradiol receptor beta is also translocated into

19
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

mitochondria and has neuroprotective actions (Rettberg et al., 2014). In estradiol on board and not after ovariectomy (Shansky et al., 2010).
particular, after a surgical menopause, as shown in rhesus monkeys, Shansky, now at Northeastern University, is investigating the con-
circular mitochondria are formed in cortical neurons that are known to sequences of such sex differences for the behavioral mechanistic sex
generate free radicals and oxidative and inflammatory damage (Hara differences in how male and female rats learn and extinguish fear
et al., 2014; Picard and McEwen, 2014), where estradiol treatment may conditioning even though the outcome is similar (Gruene et al., 2015).
be particularly important as a neuroprotective strategy. In the hippocampus of male rats, 21 days of chronic restraint stress
(CRS) causes apical dendrites of CA3 neurons to retract and a loss of
16. Diurnal cycles and circadian disruption ∼30% of the parvalbumin (PARV)-containing neurons in the dentate
gyrus; these changes do not occur following CRS in female rats (Galea
Plasticity of the brain extends to the day-night (diurnal) cycle of et al., 1997; Milner et al., 2013). Moreover, female and male rats show
waking and sleeping and it goes beyond the hippocampus to other brain effects in the opposite direction of chronic stress on hippocampal de-
regions. During his postdoctoral work, Conor Liston, now an Assistant pendent memory, with males showing impairment and females showing
Professor of Psychiatry at Cornell Medical School, found that some, but enhancement or no effect (Bowman et al., 2003; Luine et al., 1996,
not all, synapses in many parts of the cerebral cortex turn over during 1994). Moreover, exposure of male and female rats to restraint plus
the diurnal cycle due to the fluctuation of cortisol and that interfering intermittent tail shock has opposite effects on classical eyeblink con-
with that cycle by elevated cortisol at the wrong time of day interferes ditioning, inhibiting it in females and enhancing it in males; in females,
with motor learning, like learning to play golf (Liston et al., 2013). this effect is abolished by ovariectomy and is therefore estradiol de-
Considering how many ways we modern humans interfere with our pendent (Shors et al., 2001; Wood and Shors, 1998). A morphological
natural day-night rhythm, for example, by turning on a light in the correlate of this in the hippocampus is the finding that acute stress
middle of the night, this is a lesson to all of us to give the “wisdom of inhibits estradiol-depending spine formation in CA1 neurons of the
the body” a better chance to help us. hippocampus, whereas the same acute stressors enhance spine density
Another way we interfere with the natural cycle is through shift in male CA1 neurons, possibly by increasing testosterone secretion
work and jet lag. As a post-doctoral fellow, Ilia Karatsoreos, now an (Shors et al., 2001) upon which spine formation in the male CA1 is
Associate Professor at Washington State University, found that creating dependent (Leranth et al., 2003). Neonatal masculinizaton of females
an animal model of shift work caused dendrites in the prefrontal cortex, made them respond positively, like genetic males, to the shock stressor
the brain region that governs our ability to regulate emotions and im- (Shors, 2016) Moreover, in females, depending on reproductive status
pulses as well as working memory, to shrink and the animal to become and previous experience, the negative stress effect was altered, e.g., it
cognitively rigid when challenged with a memory task that required was absent in mothers and virgin females with experience with infants
changing the rules; moreover, the shift work animals became fatter and (Shors, 2016).
insulin resistant, signs of pre-diabetes and depressive-like behavior Among possible mechanisms for the sex differences, the cortico-
(Karatsoreos et al., 2011). Shift work in our own species is associated trophin releasing factor receptor stands out since in hippocampus as
with greater obesity, diabetes, cardiovascular disease and mental health well as in locus coeruleus, there are sex differences in the association of
problems (Bowles et al., 2017; McEwen and Karatsoreos, 2015). As the CRF receptor (CRF1) with the G protein s and beta-arrestin 2 that
shown by Nicole Bowles working with Karatsoreos, Cecilia Hillard and make females more responsive to acute stress and less able to adapt to
Matt Hill, one of the mediators for this obesity, acting peripherally, are chronic stress as a result of compromised CRF1 internalization
endocannabinoids acting via the CB1 receptor (Bowles et al., 2015). (Valentino et al., 2013) Thus the failure of female rats and mice to show
Nicole Bowles, who is now at Oregon Health Sciences University as a spine loss and dendrite shrinkage (Galea et al., 1997; Pawlak et al.,
postdoctoral fellow, has gone on to begin to relate this story to the 2005) may be related to this sex difference.
health effects of shift work in transit workers (Bowles et al., 2017). Sex differences in stress-induced remodeling of hippocampal, pre-
frontal cortex and amygdala neurons develop during the adolescent
17. Sex hormones and sex differences maturation, and they are quantitatively, but not qualitatively, evident
in adolescent rats subjected to repeated restraint stress for 21d, ac-
As noted at the beginning, we and others found that sex hormones cording to studies carried out by Lisa Eiland and reviewed together with
act on the hippocampus and virtually every extra-hypothalamic brain studies of how stress affects the adolescent brain and body development
region to affect a wide range of neurological and behavioral processes by Eiland and Romeo (Eiland et al., 2012; Eiland and Romeo, 2012).
(McEwen et al., 2012; McEwen and Milner, 2007, 2017). Up to this However, restraint stress in adolescents suppresses neurogenesis in fe-
point in the review, we have not considered how men and women males but not in males (Barha et al., 2011). Then, in adulthood, there
differ. Women are more prone to depression after toxic stress while men are striking qualitative sex differences in stress-induced remodeling, as
show more antisocial behavior as their response (Eme, 2007; Kessler noted above.
et al., 1994, 1993). Over the years until the present, many McEwen lab Sex differences in hormones are also evident in the hippocampus.
members have gone on to study sex differences in how the brain re- Neil MacLusky and colleagues, when Neil was at Yale, have docu-
sponds to stress. Liisa Galea, now at University of British Columbia, mented how male rats do not show estradiol-induced synapse formation
showed that female rats did display the same chronic stress-induced in hippocampus, although androgens induce synapses in male hippo-
dendritic retraction that is evident in males and she did so together with campus (Leranth et al., 2003), although males will respond to estradiol
postdoctoral fellows Bob Spencer and student Firdaus Dhabhar (Galea if sexual differentiation is blocked at birth by aromatase inhibitors
et al., 1997). (Galea has gone on to make seminal contributions re- (Lewis et al., 1995). Moreover, testosterone, but not estradiol, increases
garding estradiol regulation of neurogenesis (Galea et al., 2013) and the survival of dentate gyrus neurons in the male brain (Hamson et al.,
neural and behavioral effect of pregnancy, including post-partum de- 2013; Spritzer and Galea, 2007). Androgen receptors are present in the
pression (Barha et al., 2007; Galea et al., 2001; Pawluski and Galea, nucleus and non-nuclear locations of CA1 pyramidal cells where the
2007; Workman et al., 2016).) synapse formation occurs and in non-nuclear (Kerr et al., 1995; Tabori
Likewise, in collaboration with us, postdoctoral fellow Rebecca et al., 2005). Other sex differences in hormone acton have been docu-
Shansky and John Morrison showed that males and females differ in mented in a special issue of the Journal of Neuroscience Research
response to stress in the medial prefrontal cortex – males show stress- edited by Larry Cahill (Cahill, 2017).
induced shrinkage of dendrite that project cortically whereas females The field of sex differences research now recognizes that men and
do not show that change but, rather, show stress-induced expansion of women typically handle stress in different ways even though we do
dendrites that project to the amygdala but only when there were many of the same things equally well – hence, the “Men are from Mars

20
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

and Women are from Venus” distinction. This is evident in an imaging the ability of neurons to obtain an adequate energy supply, particularly
study on men and women during tests of emotional sensitivity, where in the hippocampus when it is activated by a cognitive challenge
men and women sored equally well but showed marked differences in (Sapolsky, 1992; de Leon et al., 1997).
the brain regions activated by the same tasks (Derntl et al., 2010). In- Insulin receptors in hippocampus and hypothalamus: In order to
deed, although we and others find that there are receptors for both understand the nature of the insulin resistance in the brain, work by
estradiol, androgens and progestins in both the male and female brain former postdoctoral fellows, Claudia Grillo, Gerardo Piroli and Larry
that regulate memory, pain, coordinated movement and other func- Reagan at the University of South Carolina, has led to important dis-
tions, there important developmentally- and genetically-programmed tinctions between hypothalamus and hippocampus. When hypotha-
sex differences in our brains and in how the brain responds to stressors lamic insulin receptors were depleted by a locally applied antisense
and other experiences. (Grillo et al., 2014), systemic features of diabetes appeared and there
These sex differences occur throughout the brain and not just in was also increased depressive- and anxiety-like behavior along with a
brain regions like the hypothalamus that are involved in reproduction depletion of BDNF in the hippocampus and hypothalamus. Indeed, a
(McEwen and Milner, 2017). In fact, new research indicates that, at the short-term corticosterone treatment causes not only systemic insulin
molecular level, male and female responses to stress in the hippo- resistance but also insulin resistance in the hippocampus reflected by
campus, can be strikingly different (Marrocco and McEwen, 2016; decreased GLUT4 levels and reduced insulin-stimulated insulin receptor
Marrocco et al., 2017). For example, RNA sequencing of ribosome- phosphorylation (Piroli et al., 2007). Moreover, the systemic metabolic
bound RNA from a single neuron type, hippocampal CA3 neurons re- state plays a role in the brain, in that fatty Zucker rats have poorer
vealed remarkable qualitative sex differences. For example, female hippocampal dependent memory than lean Zucker rats, as well as im-
mice displayed greater gene expression activation after acute stress paired translocation of an insulin-dependent glucose transporter to
than males and the direction of stress regulation of genes was often in hippocampal membranes (Winocur et al., 2005). Moreover, a diet rich
the opposite direction in males and females. Moreover, heterozygous in fat has been shown to impair hippocampal-dependent memory
BDNF Val66Met mice, a model that is genetically susceptible to stress, (Winocur and Greenwood, 1999) and a combination of a high fat diet
recapitulated the effects of acute stress on mRNA levels in wild-type and a 3 week predator exposure causes retraction of dendrites in the
mice without any applied stress (a “pre-stress” state). However, like CA3 hippocampus, even though neither treatment alone had this effect
wild-type mice, heterozygous BDNF Val66Met mice only displayed (Baran et al., 2005).
activation of immediate early genes when actually stressed. This phe- Indeed systemic metabolism, particularly triglycerides and leptin
notype included the expression of genes related to glutamatergic and resistance in the hippocampus, has turned out to be very important, as a
glucocorticoid pathways. Behaviorally, only heterozygous BDNF mild food restriction paradigm both prevented and reversed the obesity
Val66Met females exhibited spatial memory impairment, regardless of phenotype in the rats in which hypothalamic insulin receptors had been
acute stress, and this effect was not observed in ovariectomized het- inactivated by antisense (Grillo et al., 2014). The food restriction
erozygous BDNF Val66Met females, suggesting that circulating ovarian paradigm restored long-term potentiation (LTP) in the CA1 region and
hormones induce cognitive impairment in Met carriers. Cognitive def- reversed the decreases in the phosphorylated/total ratio of GluA1
icits were not observed in males of either genotype, which instead Ser845 AMPA receptor subunit expression observed in the hippo-
showed impaired memory only after stress. This work sheds further campus of hypo-IRAS rats. Because regulation of GluA1 Ser845 AMPA
light on ways that genes, environment and sex interact to affect the receptor subunit phosphorylation is a key feature of leptin action, the
transcriptome, furthering our understanding of sexually dimorphic impairment of insulin action in hypo-IRAS rats is mediated through the
molecular mechanisms that underlie the response to stress in men and impairment of hippocampal leptin activity (Grillo et al., 2014).
women. However, while glucose uptake is important and impaired leptin
signaling by elevated triglycerides can impair insulin action, it is not
18. Metabolic hormones, glucose and the brain the dominant feature of insulin action and insulin resistance in the
hippocampus (Fadel and Reagan, 2016). Antisense inactivation of
Because of the work of Robert Sapolsky on glucocorticoid en- hippocampal insulin receptors produced impaired long-term potentia-
dangerment of the hippocampus, I was drawn into a collaboration with tion and hippocampal dependent learning without any systemic meta-
Mony de Leon at NYU that showed in Alzheimer’s patients an increase bolic disturbance (Grillo et al., 2015). While antisense inactivation of
in cortisol secretion with a glucose challenge that increased with the insulin receptors is not a likely mechanism in pathophysiology, it has
extent of hippocampal atrophy (DeLeon et al., 1988). A follow up study been shown that excessive stimulation of NMDA receptors by glutamate
among normal individuals showed that a pharmacological dose of and sustained calcium influx stimulates the activity of tyrosine phos-
cortisol, reduced the glucose utilization of the hippocampus, and serum phatases on insulin receptors and attenuates insulin signal transduction
glucose levels increased. This did not occur in Alzheimer’s patients, and this is a process that may lead to a cascade resulting in dementia
indicating that glucocorticoid-mediated regulation of glucose transport (De Felice et al., 2014). Another processes that may be involved in brain
is altered in AD, and that this may underlie both the hippocampal in- insulin resistance is the active transport of IGF-1 into the brain, which is
sensitivity to cortisol and the failure in these patients to mount a per- activated by neural activity (Nishijima et al., 2010); impairment of this
ipheral glucose response (de Leon et al., 1997; DeLeon et al., 1988). has not yet been demonstrated in insulin resistance but the requirement
Indeed an important factor for hippocampal structure and function of neural activity may be one of the reasons that exercise is beneficial
is glucose regulation; and this realization has led to insights about the for cognitive function and against insulin resistance.
ability of metabolic hormones such as insulin, leptin, ghrelin and IGF-1 Role of astrocytic glycogen: While muscle glycogen fuels exercising
to enter and regulate hippocampal health and function. When the muscles to sustain endurance capacity, the brain also stores glycogen in
hippocampus is activated by a cognitive task it needs glucose (McNay astrocytes to produce lactate as an energy source transported to active
et al., 2000) and activation of glucose mobilization occurs by a choli- neurons via the monocarboxylate transporter MCT2. Disrupting the
nergically-mediated pathway to the liver (Uemura et al., 1989). These expression of the neuronal lactate transporter, MCT2, leads to failure of
changes decline with aging (Gold, 1987; McNay and Gold, 2001). long-term memory and amnesia; in particular, glycogenolysis and as-
Consistent with Robert Sapolsky’s glucocorticoid endangerment hy- trocytic lactate transporters are also critical for the molecular changes
pothesis (Sapolsky, 1992), cortisol inhibits glucose uptake in the required for memory formation, including the induction of phospho-
healthy aging brain as a way of preventing damaging glycation of CREB, Arc and phospho-cofilin (Suzuki et al., 2011). A collaboration
proteins by excess glucose, as shown by Larry Reagan (Reagan et al., with Hideyaki Soya at Tsukuba University has shown that MCT2 is also
2000). And, yet, according to Sapolsky, excess glucocorticoids impairs important for the effects of exercise and the feeling of exhaustion

21
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

(Matsui et al., 2017). Using a rat model of prolonged exhaustive ex- significantly lower in individuals with MDD compared to healthy con-
ercise, microwave irradiation of brains, metabolomics, and in- trols. Low LAC levels in individuals with MDD are associated with
tracerebroventricular injection of inhibitors of glycogenolysis and greater severity and earlier onset, and a treatment resistant course of
MCT2 showed that lactate derived from astrocytic glycogen fuels the illness. Consistent with lower LAC in more severe forms of MDD, there
prolonged exercising brain to maintain endurance capacity. Further is an association between low LAC levels and childhood trauma in pa-
studies of this process are needed in relation to hippocampal insulin tients with a treatment resistant course of illness. Together with the
resistance. association between reduced LAC and insulin resistance (IR) in animal
Metabolic factors, depression, dementia and aging: But there is a lot models, LAC is potential a biomarker to delineate, diagnose and treat a
more to this story, as shown by important work of Antonio Convit at novel biologically-defined MDD subtype that may associate LAC with
NYU and Natalie Rasgon at Stanford. Metabolic factors involving glu- insulin resistane.
cose regulation and insulin resistance play a role in hippocampal vo-
lume change in the human hippocampus in mild cognitive impairment 20. Transgenerational influences and brain development
with aging (Convit et al., 2003; Rasgon et al., 2001). Poor glucose
regulation is associated with smaller hippocampal volume and poorer Another important element so far in this discussion of stress is the
memory function in individuals in their 60s and 70s who have “mild influence of events early in life and their epigenetic effects on brain and
cognitive impairment” (MCI) (McIntyre et al., 2010), and both MCI and body development. Michael Meaney, a former postdoctoral fellow and
Type 2, as well as Type 1, diabetes are recognized as risk factors for now Professor at McGill University, has led the way in demonstrating
depression and dementia as will now be described (de Leon et al., 2001; the important role of postnatal maternal care in emotional and cogni-
Haan, 2006; Ott et al., 1996; Rasgon et al., 2001) . tive development. In my lab, Meaney and Robert Sapolsky investigated
Within the life course perspective of epigenetic effects of a pro- ontogeny of glucocorticoid receptors in the neonatal rat brain (Meaney
gression of lifetime experiences and health-related behaviors operating et al., 1985a, 1985b), which led them to later investigate the ability of
on different genotypes (Halfon et al., 2014), there is growing evidence neonatal “handling” of infant rats to slow down brain aging (Meaney
for a relationship between insulin resistance and risk for cognitive de- et al., 1988, 2013). Meaney went on to show that “handling” (Levine
cline and dementia, as first described by Natalie Rasgon who has be- et al., 1967), i.e., separating pups from the mother for 10–20′, increases
come a collaborator and has catalyzed the formation of PALS (https:// maternal care when the pups are returned. Going on to explore the role
www.pals-network.org/) (Rasgon and Jarvik, 2004; Rasgon et al., of maternal care, Meaney and Darlene Francis showed that infant rats
2011). Hippocampal shrinkage is reported in women with insulin re- raised with a nurturing mom develop less emotionality and great ability
sistance who are at risk for Alzheimer’s disease (Rasgon et al., 2011), to explore novel places and things, while pups raised with an anxious
and in men as well as women with Type 2 diabetes (Gold et al., 2007). mom that provides inconsistent care shows the opposite outcome
Hippocampal shrinkage is an index of early Alzheimer’s Disease (de (Francis et al., 1999). This was done by Francis switching embryos in
Leon et al., 2004; Hampel et al., 2008). We have seen that elevated the womb (Francis et al., 2003) and also by cross-fostering pups post-
glucocorticoid levels can also shut off glucose and endanger survival of natally between mothers of two mouse strains by Wayne Brake, Russ
hippocampal neurons (Sapolsky, 1992). As noted, the normal, physio- Romeo and Helen Sisti together with Darlene Francis (Priebe et al.,
logical regulation apparently does not happen in the Alzheimer brain in 2005).
which input and output of hippocampus is damaged (de Leon et al., Later, Akaysha Tang’s group working with Russ Romeo and me
1997; DeLeon et al., 1988). An oxidative stress and inflammatory cas- showed that it was the consistency of maternal care and not the amount
cade has been suggested as a pathway from insulin resistance to Alz- that has the most positive effect (Tang et al., 2014). Indeed, chaos in the
heimer’ pathology (Gold et al., 2013; Manolopoulos et al., 2010) re- nest has negative effects on the offspring as it does in humans (Evans
presenting an example of an allostatic overload. and Wachs, 2010; Molet et al., 2014). Cross-fostering of infants between
Possible interventions including hormone therapy, particularly un- good and bad moms alters the outcome, pointing to what is now re-
opposed estradiol (Rasgon et al., 2014), and PPARγ agonists such as ferred to as epigenetic transgenerational behavioral transmission
pioglitazone (Gold et al., 2013; Lin et al., 2015) along with lifestyle (Francis et al., 1999).
changes leading to increased physical activity. Given the occurrence of John Kral at Downstate Medical Center, Brooklyn, introduced us to
obesity and Type 2 diabetes already in adolescent youth, as shown by other forms of transgenerational transmission of traits: even before
Convit’s group at NYU (Yau et al., 2012), early life intervention is im- conception and during life in the womb, paternal and maternal obesity
perative wherever possible to redirect the progression of the allostatic can affect the child (Donkin et al., 2016; Kral et al., 2006). These may
load towards a healthier path. The involvement of my laboratory and involve “epigenetic” changes of the DNA of the sperm and egg that do
former fellows led unexpectedly to a new direction involving a mole- not alter the genetic code per se, but, rather, how it is read. In the case
cule, carnitine, that was the subject of my first co-authored paper in of parental obesity, this increases the risk that the child will also be-
1959 (Fritz and Mcewen, 1959). come obese. Moreover, adverse early life experience in infancy and
childhood involving poverty, abuse and neglect, affect how genes are
19. Depressive-like behavior and insulin resistance associated expressed and determine how well brain regions such as the hippo-
with acetyl-L-carntine (LAC) deficiency campus, amygdala and prefrontal cortex develop and function during
childhood into young adulthood. Indeed, as we have seen, the brain is
As explained above, the arrival of Carla in our laboratory for the continually changing with experiences, creating memories and alerting
final year of her Ph.D. thesis introduced us to the role of acetyl-L-car- brain architecture via mechanisms that are facilitated in part by cir-
nitine (LAC) in ameliorating depressive-like behavior in animal models culating sex, stress and metabolic hormones and chemicals produced by
(Nasca et al., 2013). In one animal model, the FSL rat, continuing a the immune system.
collaboration with Aleksander Mathe and Per Svenningsson of Kar- This has led to a new view of the epigenetic changes over the life
olinska Institute, Stockholm, LAC deficiency is associated with depres- course that determine trajectories of health and disease and the plas-
sive-like behavior and, more recently, it is accompanied by elevated ticity of the brain offers opportunities for changing the trajectory
insulin, leptin and triglycerides in serum; LAC supplementation not (Halfon et al., 2014). Indeed, we cannot “roll back the clock” and
only reduces the depressive like behavior within 3–5 days, but it also “reverse” the effects of experiences, positive or negative. Rather, we
treats the metabolic dysregulation in a study led by Benedetta Bigio must think of “recovery” and “redirection” and ”resilience”, rather than
(Bigio et al., 2016), In human MDD, Carla Nasca, Natalie Rasgon, “reversal” (Gray et al., 2014). See Fig. 2. We therefore think about
Francis Lee, Jim Kocis and James Murrough find that LAC is “changing trajectories” of function resulting in compensatory changes

22
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

in the brain and body over the life course. A pediatrician researcher at science/national-scientific-council-on-the-developing-child/).
UCLA, Neal Halfon, has written and spoken about “life course health
development”, or LCHD as the most up-to-date overview of medicine, 22. Interventions
contrasting that with the view of “magic bullets” like penicillin that
revolutionized treatment of infectious disease but does not apply to Based on this, what are the implications of this line of research for
antidepressants or drugs, like statins, that help but do not “cure” the in interventions that help individuals and societies deal with the stresses
principle preventable diseases of modern life (Halfon et al., 2014). of modern life and their health consequences? We shall consider in-
Going beyond the psychosocial model of how health behaviors and dividuals, first, and then the role of societies and how groups of in-
toxic stress cause those diseases (Engel, 1977), LCHD notes the im- dividuals, differing in income and education are affected, and can be
portance of events pre-conception, prenatally and throughout the life- affected by programs of government and the private sector.
course in which income and education have a huge influence. The de- Personalized medicine: Regarding individuals, the term “persona-
terminants of diabetes, depression and dementia are a good example of lized medicine” has become very popular, but it has generally focused
this (Rasgon and McEwen, 2016). on the individual genetic constitution and not the influence of epige-
netics. However, I was very fortunate to meet and establish a long-
21. Life course health development for an ever-changing brain distance collaboration with Linn Getz Professor in Behavioural Sciences
in Medicine of the Department of Public Health and General Practice at
What does this mean for prevention and treatment? In the spirit of Norwegian University of Science and Technology in Trondheim,
integrative medicine, it seems to me that it is important to let the Norway. In a joint paper (McEwen and Getz, 2013) we wrote: “The aim
“wisdom of the body” prevail and to focus upon strategies that center of personalized medicine is to base medical prevention and therapy on
around the use of targeted behavioral therapies along with treatments, the unique health and disease susceptibility profile of each individual.
including pharmaceutical agents, that “open up windows of plasticity” From a medical perspective, knowledge of a person encompasses both
in the brain and facilitate the efficacy of the behavioral interventions biological and biographical perspectives. The latter includes significant
(McEwen, 2012). This is because a major challenge throughout the life events and experiences throughout the person's lifespan, from concep-
course is to find ways of redirecting future behavior and physiology in tion to the present, in which epigenetic influences play an important
more positive and healthy directions (Halfon et al., 2014). We do not role. In practice, personalized medicine should emphasize the devel-
mean “reversibility” as in “rolling back the developmental clock” but opment and maintenance of a healthy nervous system. The neurobio-
rather “redirection” of those features of a species that can be modified logical processes for that depend heavily on the psychosocial environ-
by experiences. ment, in particular the presence of responsible, caring adults and
Even in adulthood, gene expression continually changes with ex- integration in a reasonably fair society. A healthy brain subsequently
perience (see Fig. 2 (Gray et al., 2014)) and there is loss of resilience promotes good health throughout life, both through direct, favorable
with aging (Bloss et al., 2010) that can be redirected by exercise influences on the body's intrinsic biological pathways, and indirectly by
(Erickson et al., 2011) and possibly by pharmacological intervention enabling the person to engage in supportive relationships, make wise
(Pereira et al., 2014). Even chronic anxiety, possibly resulting from decisions and take good care of him/ herself. From a public health
adverse childhood experiences, can respond to a behavioral interven- perspective, hi-tech personalized medicine based on detailed bio-mo-
tion in adulthood (Holzel et al., 2010). Indeed mindfulness based stress lecular mapping, monitoring and tailored drug interventions holds
reduction and meditation increases functional connectivity within the promise only as part of a wider, socio-culturally informed approach to
brain and benefit fluid intelligence as well as improving function in the person.”
aging (Gard et al., 2014a, 2014b) and meaning and purpose in life also So what can we do to change trajectories and alleviate the many
has benefits for overall health and cognitive function (Carlson et al., stressors in our lives? At the individual level, prevention of early life
2009; Fredrickson et al., 2013). adversity for children and families is key via programs like the Nurse-
The response of the brain to stressors is a complex process involving Family Partnership (http://www.nursefamilypartnership.org/) that
multiple interacting mediators that utilizes both genomic and non- pave the way for strong attachment between mother and child and good
genomic mechanisms from the cell surface to the cytoskeleton to epi- child-rearing practices. Yet, when bad things have happened to a child
genetic regulation via the cell nucleus. Resilience in the face of stress is from adverse prenatal, gestational and early life influences, our in-
a key aspect of a healthy brain, even though gene expression shows a creasing knowledge of the plasticity of the brain gives hope for thera-
brain that continually changes with experience (McEwen et al., 2015a). pies that utilize brain-body interactions while enhancing a sense of self-
Resilience may be thought of as an active process that implies ongoing regulation and control to reduce toxic to at least more tolerable stress.
adaptive plasticity without external intervention (Russo et al., 2012). Along with a healthy diet, positive social interactions and support,
Therefore recovery of stress-induced changes in neural architecture adequate sleep and attention to maintaining a normal sleep waking
after stress is not a “reversal” but a form of neuroplastic adaptation and cycle, regular physical activity is vitally important not only for meta-
resilience that may be impaired in mood disorders, when the brain “gets bolic and cardiovascular function but also for memory, mood and for
stuck” and needs external intervention. Loss of resilience may also prevention of dementia. We think of this as using healthy behaviors to
occur with aging and need external intervention such as exercise (Bloss “open a window” of plasticity and allow the wisdom of the body to
et al., 2010). exert itself as well as to allow targeted behavioral interventions to
On the other hand, resilience is decreased and vulnerability is in- shape brain circuits in a more positive direction. Examples of the tar-
creased by adverse childhood experiences (ACE) and poverty that lead geted interventions include compensatory repair of damage from a
to “biological embedding” of trajectories of response to stressful life stroke by intensive physical therapy (Chollet et al., 2011) and how
events (Shonkoff et al., 2009) throughout the life course (Halfon et al., “lazy eye”, i.e., abnormal development of a visual field that responds
2014) which contribute disproportionately to allostatic overload in the abnormally to the input of both eyes, is ameliorated by binocular visual
form of physical and mental health disorders over the life course and stimulation (Vetencourt et al., 2008).
impaired brain development (Felitti et al., 1998; Hanson et al., 2013). Policy and Practice: At the level of societies, income and education
Evidence from CpG methylation of DNA indicates the embedded in- are hugely important as determinants of longievity and health span as
fluence of early adversity (McGowan et al., 2009). From hereon in this shown in 2 publications from the MacSES Network (see http://www.
review, we move into the domain started for me by the MacArthur macses.ucsf.edu/downloads/Reaching_for_a_Healthier_Life.pdf and
Foundation networks and continued for me in the National Scientific more in depth: http://onlinelibrary.wiley.com/doi/10.1111/nyas.
Council on the Developing Child (http://developingchild.harvard.edu/ 2010.1186.issue-1/issuetoc). As noted at the beginning of this article

23
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

and in Box 1, my experience with MacArthur Foundation Research References


Networks brought me together with social scientists, physicians, and
epidemiologists who were interested in a common problem, namely, Aberg, M.A., Aberg, N.D., Hedbacker, H., Oscarsson, J., Eriksson, P.S., 2000. Peripheral
how to measure “stress” from our social and physical environments and infusion of IGF-1 selectively induces neurogenesis in the adult rat hippocampus. J.
Neurosci. 20, 2896–2903.
how it affects our health behaviors and our disease vulnerability. A Acheson, S.D., 1998. Independent Inquiry into Inequalities in Health Report. The
major focus was and still is the influence of income and education, Stationary Office, London.
referred to as “socioeconomic status”, and how it “gets under the skin”. Ahima, R., Krozowski, Z., Harlan, R., 1991. Type I corticosteroid receptor-like im-
munoreactivity in the rat CNS: distribution and regulation by corticosteroids. J.
In so doing, it provided the impetus and the collegial environment to Comp. Neurol. 313, 522–538.
develop the concepts of allostasis and allostatic load and overload and Ahima, R.S., Harlan, R.E., 1990. Charting of type II glucocorticoid receptor-like im-
begin to measure them and then try to find ways to alleviate toxic stress munoreactivity in the rat central nervous system. Neuroscience 39, 579–604.
Akama, K.T., McEwen, B.S., 2003. Estrogen stimulates postsynaptic density-95 rapid
and reduce allostatic overload. This I have done and in the process protein synthesis via the Akt/protein kinase B pathway. J. Neurosci. 23, 2333–2339.
came to know and admire another colleague, Teresa Seeman, Professor Allfrey, V.G., 1970. Changes in chromosomal proteins at times of gene activation. Fed.
of Epidemiology in Medicine at UCLA, who has played major role in Proc. 29, 1447–1460.
Altman, J., Das, G.D., 1965. Autoradiographic and histological evidence of postnatal
measuring and using allostatic load/overload to predict health out-
hippocampal neurogenesis in rats. J. Comp. Neurol. 124, 319–336.
comes (McEwen and Seeman, 1999; Seeman et al., 2010a, 2010c; Wiley Alves, S.E., Hoskin, E., Lee, S.J., Brake, W.G., Ferguson, D., Luine, V., Allen, P.B.,
et al., 2016). Greengard, P., McEwen, B.S., 2002. Serotonin mediates CA1 spine density but is not
One additional perspective became clear from the MacSES Network: crucial for ovarian steroid regulation of synaptic plasticity in the adult rat dorsal
hippocampus. Synapse 45, 143–151.
the most important contributor to poor adult health is childhood pov- Arango-Lievano, M., Lambert, W.M., Bath, K.G., Garabedian, M.J., Chao, M.V.,
erty as well as abuse, neglect and chaos in the home (Caspi et al., 2016; Jeanneteau, F., 2015. Neurotrophic-priming of glucocorticoid receptor signaling is
Evans and Wachs, 2010). Since the MacSES Network ended, I have essential for neuronal plasticity to stress and antidepressant treatment. Proc. Natl.
Acad. Sci. USA 112, 15737–15742.
continued this interest and contributed my expertise as a member of the Arendt, T., Stieler, J., Strijkstra, A.M., Hut, R.A., Rudiger, J., Van der Zee, E.A., Harkany,
National Scientific Council on the Developing Child, which provides the T., Holzer, M., Hartig, W., 2003. Reversible paired helical filament-like phosphor-
best current scientific knowledge in a digestible form for policy makers ylation of tau is an adaptive process associated with neuronal plasticity in hibernating
animals. J. Neurosci. 23, 6972–6981.
as well as the pediatric community and the general public to both Arnold, A., Breedlove, S., 1985. Organizational and activational effects of sex steroids on
prevent and alleviate the effects of adversity in childhood. The National brain and behavior: a reanalysis. Horm. Behav. 19, 469–498.
Scientific Council web site is very useful (http://developingchild. Azmitia Jr., E.C., McEwen, B.S., 1968. Corticosterone regulation of tryptophan hydro-
xylase in rat midbrain. Science 166, 127401276.
harvard.edu/science/national-scientific-council-on-the-developing- Banks, W.A., Owen, J.B., Erickson, M.A., 2012. Insulin in the brain: there and back again.
child/) . Pharmacol. Ther. 136, 82–93.
One of the most influential studies in this area is that conducted in Banks, W.A., Tschop, M., Robinson, S.M., Heiman, M.L., 2002. Extent and direction of
ghrelin transport across the blood-brain barrier is determined by its unique primary
Dunedin, New Zealand, by Avshalom Caspi and Terrie Moffitt. Early life
structure. J. Pharmacol. Exper. Ther. 302, 822–827.
adversity, including poverty, impair brain development, particularly Baran, S.E., Campbell, A.M., Kleen, J.K., Foltz, C.H., Wright, R.L., Diamond, D.M.,
the development of self-regulatory behaviors that determine childhood Conrad, C.D., 2005. Combination of high fat diet and chronic stress retracts hippo-
self control, and poor self control predicts physical health, substance campal dendrites. NeuroReport 16, 39–43.
Barha, C.K., Brummelte, S., Lieblich, S.E., Galea, L.A., 2011. Chronic restraint stress in
dependence, personal finances, and criminal offending outcomes, fol- adolescence differentially influences hypothalamic-pituitary-adrenal axis function
lowing a gradient of self-control (Moffitt et al., 2011). In this study, and adult hippocampal neurogenesis in male and female rats. Hippocampus 21,
“effects of children’s self-control could be disentangled from their in- 1216–1227.
Barha, C.K., Falck, R.S., Davis, J.C., Nagamatsu, L.S., Liu-Ambrose, T., 2017. Sex differ-
telligence and social class as well as from mistakes they made as ado- ences in aerobic exercise efficacy to improve cognition: A systematic review and
lescents. In another cohort of 500 sibling-pairs, the sibling with lower meta-analysis of studies in older rodents. Front. Neuroendocrinol. 46, 86–105.
self-control had poorer outcomes, despite shared family background. Barha, C.K., Pawluski, J.L., Galea, L.A.M., 2007. Maternal care affects male and female
offspring working memory and stress reactivity. Physiol. Behav. 92, 939–950.
Interventions addressing self-control might reduce a panoply of societal Barker, J.M., Galea, L.A., 2009. Sex and regional differences in estradiol content in the
costs, save taxpayers money, and promote prosperity (Moffitt et al., prefrontal cortex, amygdala and hippocampus of adult male and female rats. Gen.
2011) Going further, the same group wrote in a recent paper (Caspi Comp. Endocrinol. 164, 77–84.
Bas-Hoogendam, J.M., Andela, C.D., van der Werff, S.J., Pannekoek, J.N., van
et al., 2016): “A segment comprising 22% of the cohort accounted for
Steenbergen, H., Meijer, O.C., van Buchem, M.A., Rombouts, S.A., van der Mast, R.C.,
36% of the cohort’s injury insurance claims; 40% of excess obese Biermasz, N.R., van der Wee, N.J., Pereira, A.M., 2015. Altered neural processing of
kilograms; 54% of cigarettes smoked; 57% of hospital nights; 66% of emotional faces in remitted Cushing's disease. Psychoneuroendocrinology 59,
134–146.
welfare benefits; 77% of fatherless child-rearing; 78% of prescription
Bath, K.G., Schilit, A., Lee, F.S., 2013. Stress effects on BDNF expression: effects of age,
fills; and 81% of criminal convictions. Childhood risks, including poor sex, and form of stress. Neuroscience 239, 149–156.
brain health at three years of age, predicted this segment with large Baulieu, E.E., Robel, P., 1990. Neurosteroids: a new brain function? J. Steroid Biochem.
effect sizes. Early-years interventions that are effective for this popu- Mol. Biol. 37, 395–403.
Bavelier, D., Levi, D.M., Li, R.W., Dan, Y., Hensch, T.K., 2010. Removing brakes on adult
lation segment could yield very large returns on investment.” brain plasticity: from molecular to behavioral interventions. J. Neurosci. 30,
Finally, what can be done to improve the outcome? Societies differ 14964–14971.
in the degree to which they promote an environment that encourages Bennett, E., Diamond, M., Krech, D., Rosenzweig, M., 1964. Chemical and anatomical
plasticity of brain. Science 146, 610–619.
equality and harmony or inequality and discord (Wilkinson and Pickett, Bennur, S., Shankaranarayana Rao, B.S., Pawlak, R., Strickland, S., McEwen, B.S.,
2010). Virtually all policies of government and the private sector affect Chattarji, S., 2007. Stress-induced spine loss in the medial amygdala is mediated by
health, whether they deal with housing, transportation, health care, tissue-plasminogen activator. Neuroscience 144, 8–16.
Biegler, R., McGregor, A., Krebs, J.R., Healy, S.D., 2001. A larger hippocampus is asso-
education, flexible working hours, and vacations (Acheson, 1998). ciated with longer-lasting spatial memory. Proc. Natl. Acad. Sci. USA 98, 6941–6944.
Thus, policies that are forward-looking by government and the private Biegon, A., Reches, A., Snyder, L., McEwen, B.S., 1983. Serotonergic and noradrenergic
sector are vitally important to promote better health and to increase receptors in the rat brain: modulation by chronic exposure to ovarian hormones. Life
Sci. 32, 2015–2021.
“healthspan”, as opposed to “lifespan”. They can do so by providing
Bigio, B., Mathe, A.A., Sousa, V.C., Zelli, D., Svenningsson, P., McEwen, B.S., Nasca, C.,
opportunities for individuals to experience better working and living 2016. Epigenetics and energetics in ventral hippocampus mediate rapid anti-
environments and to learn and practice healthy behaviors, recognizing depressant action: Implications for treatment resistance. Proc. Natl. Acad. Sci. USA
113, 7906–7911.
the “the wisdom of the body”. We must give the body and brain, which
Bliss, T.V.P., Lomo, T., 1973. Long-lasting potentiation of synaptic transmission in the
work together and know what to do, a chance to do so!. For business, dentate area of the anaesthetized rabbit following stimulation of the perforant path.
such policies also improve employee morale, loyalty and productivity. J. Physiol. 232, 331–356.
From such policies, there is enormous benefit to society to reduce ex- Bloss, E.B., Janssen, W.G., McEwen, B.S., Morrison, J.H., 2010. Interactive effects of stress
and aging on structural plasticity in the prefrontal cortex. J. Neurosci. 30,
penses for health care and, most importantly, to reduce human misery!

24
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

6726–6731. Daniel, J.M., Dohanich, G.P., 2001. Acetylcholine mediates the estrogen-induced increase
Bowles, N.P., Karatsoreos, I.N., Li, X., Vemuri, V.K., Wood, J.A., Li, Z., Tamashiro, K.L., in NMDA receptor binding in CA1 of the hippocampus and the associated improve-
Schwartz, G.J., Makriyannis, A.M., Kunos, G., Hillard, C.J., McEwen, B.S., Hill, M.N., ment in working memory. J. Neurosci. 21, 6949–6956.
2015. A peripheral endocannabinoid mechanism contributes to glucocorticoid- Datson, N.A., van den Oever, J.M., Korobko, O.B., Magarinos, A.M., de Kloet, E.R.,
mediated metabolic syndrome. Proc. Natl. Acad. Sci. USA 112, 285–290. McEwen, B.S., 2013. Previous history of chronic stress changes the transcriptional
Bowles, N.P., McEwen, B.S., Boutin-Foster, C., 2017. Trouble in transit: Organizational response to glucocorticoid challenge in the dentate gyrus region of the male rat
barriers to workers' health. Am. J. Ind. Med. 60, 350–367. hippocampus. Endocrinology 154, 3261–3272.
Bowman, R.E., Beck, K.D., Luine, V.N., 2003. Chronic stress effects on memory: sex dif- De Felice, F.G., Lourenco, M.V., Ferreira, S.T., 2014. How does brain insulin resistance
ferences in performance and monoaminergic activity. Horm. Behav. 43, 48–59. develop in Alzheimer's disease? Alzheimer's Dementia: J Alzheimer's Assoc. 10,
Brake, W.G., Alves, S.E., Dunlop, J.C., Lee, S.J., Bulloch, K., Allen, P.B., Greengard, P., S26–S32.
McEwen, B.S., 2001. Novel target sites for estrogen action in the dorsal hippocampus: de Leon, M.J., Convit, A., Wolf, O.T., Tarshish, C.Y., DeSanti, S., Rusinek, H., Tsui, W.,
an examination of synaptic proteins. Endocrinology 142, 1284–1289. Kandil, E., Scherer, A.J., Roche, A., Imossi, A., Thorn, E., Bobinski, M., Caraos, C.,
Bulloch, K., Miller, M.M., Gal-Toth, J., Milner, T.A., Gottfried-Blackmore, A., Waters, Lesbre, P., Schlyer, D., Poirier, J., Reisberg, B., Fowler, J., 2001. Prediction of cog-
E.M., Kaunzner, U.W., Liu, K., Lindquist, R., Nussenzweig, M.C., Steinman, R.M., nitive decline in normal elderly subjects with 2-[18F]fluoro-2-deoxy-D-glucose/posi-
McEwen, B.S., 2008. CD11c/EYFP transgene illuminates a discrete network of den- tron-emission tomography (FDG/PET). Proc. Natl. Acad. Sci. USA 98, 10966–10971.
dritic cells within the embryonic, neonatal, adult, and injured mouse brain. J. Comp. de Leon, M.J., DeSanti, S., Zinkowski, R., Mehta, P.D., Pratico, D., Segal, S., Clark, C.,
Neurol. 508, 687–710. Kerkman, D., DeBernardis, J., Li, J., Lair, L., Reisberg, B., Tsui, W., Rusinek, H., 2004.
Burger, D.K., Saucier, J.M., Iwaniuk, A.N., Saucier, D.M., 2013. Seasonal and sex differ- MRI and CSF studies in the early diagnosis of Alzheimer's disease. J. Int. Med. 256,
ences in the hippocampus of a wild rodent. Behav. Brain Res. 236, 131–138. 205–223.
Cahill, L., 2017. An issue whose time has come. J. Neurosci. Res. 95, 12–13. de Leon, M.J., McRae, T., Rusinek, H., Convit, A., De Santi, S., Tarshish, C., Golomb, J.,
Cameron, H.A., Gould, E., 1994. Adult neurogenesis is regulated by adrenal steroids in the Volkow, N., Daisley, K., Orentreich, N., McEwen, B., 1997. Cortisol reduces hippo-
dentate gyrus. Neuroscience 61, 203–209. campal glucose metabolism in normal elderly, but not in Alzheimer's disease. J. Clin.
Cameron, H.A., Gould, E., 1996. The control of neuronal birth and survival. In: Shaw, C.A. Endocrinol. Metabol. 82, 3251–3259.
(Ed.), Receptor Dynamics in Neural Development, first ed. CRC Press, New York, pp. DeLeon, M.J., McRae, T., Tsai, J., George, A., Marcus, D., Freedman, M., Wolf, A.,
141–157. McEwen, B.S., 1988. Abnormal cortisol response in Alzheimer's disease linked to
Carlson, M.C., Erickson, K.I., Kramer, A.F., Voss, M.W., Bolea, N., Mielke, M., McGill, S., hippocampal atrophy. Lancet 391–392.
Rebok, G.W., Seeman, T., Fried, L.P., 2009. Evidence for neurocognitive plasticity in Derntl, B., Finkelmeyer, A., Eickhoff, S., Kellermann, T., Falkenberg, D.I., Schneider, F.,
at-risk older adults: the experience corps program. J. Gerontol. Ser. A, Biol. Sci. Med. Habel, U., 2010. Multidimensional assessment of empathic abilities: neural correlates
Sci. 64, 1275–1282. and gender differences. Psychoneuroendocrinology 35, 67–82.
Carro, E., Trejo, J.L., Busiguina, S., Torres-Aleman, I., 2001. Circulating insulin-like DeVoogd, T., Nottebohm, F., 1981. Gonadal hormones induce dendritic growth in the
growth factor 1 mediates the protective effects of physical exercise against brain adult avian brain. Science 214, 202–204.
insults of different etiology and anatomy. J. Neurosci. 21, 5678–5684. Dhabhar, F., Miller, A.H., Stein, M., McEwen, B.S., Spencer, R., 1994. Diurnal and acute
Carruth, L.L., Jones, R.E., Norris, D.O., 2002. Cortisol and Pacific salmon: a new look at stress-induced changes in distribution of peripheral blood leukocyte subpopulations.
the role of stress hormones in olfaction and home-stream migration. Integr. Comp. Brain Behav. Immun. 8, 66–79.
Biol. 42, 574–581. Dhabhar, F.S., Malarkey, W.B., Neri, E., McEwen, B.S., 2012. Stress-induced redistribu-
Caspi, A., Houts, R.M., Belsky, D.W., Harrington, H., Hogan, S., Ramrakha, S., Richie tion of immune cells-From barracks to boulevards to battlefields: a tale of three
Poulton, R., Moffitt, T.E., 2016. Childhood forecasting of a small segment of the hormones - Curt Richter Award Winner. Psychoneuroendocrinology.
population with large economic burden. Nat. Hum. Behav. 1. Dhabhar, F.S., McEwen, B.S., 1997. Acute stress enhances while chronic stress suppresses
Chen, Y., Molet, J., Lauterborn, J.C., Trieu, B.H., Bolton, J.L., Patterson, K.P., Gall, C.M., cell-mediated immunity in vivo: a potential role for leukocyte trafficking. Brain
Lynch, G., Baram, T.Z., 2016. Converging, synergistic actions of multiple stress Behav. Immun. 11, 286–306.
hormones mediate enduring memory impairments after acute simultaneous stresses. Diamond, D.M., Bennett, M.C., Fleshner, M., Rose, G.M., 1992. Inverted-U relationship
J. Neurosci. 36, 11295–11307. between the level of peripheral corticosterone and the magnitude of hippocampal
Chen, Z.Y., Jing, D., Bath, K.G., Ieraci, A., Khan, T., Siao, C.J., Herrera, D.G., Toth, M., primed burst potentiation. Hippocampus 2, 421–430.
Yang, C., McEwen, B.S., Hempstead, B.L., Lee, F.S., 2006. Genetic variant BDNF Diano, S., Farr, S.A., Benoit, S.C., McNay, E.C., da Silva, I., Horvath, B., Gaskin, F.S.,
(Val66Met) polymorphism alters anxiety-related behavior. Science 314, 140–143. Nonaka, N., Jaeger, L.B., Banks, W.A., Morley, J.E., Pinto, S., Sherwin, R.S., Xu, L.,
Cho, K., 2001. Chronic 'jet lag' produces temporal lobe atrophy and spatial cognitive Yamada, K.A., Sleeman, M.W., Tschop, M.H., Horvath, T.L., 2006. Ghrelin controls
deficits. Nat. Neurosci. 4, 567–568. hippocampal spine synapse density and memory performance. Nat. Neurosci. 9,
Chollet, F., Tardy, J., Albucher, J.F., Thalamas, C., Berard, E., Lamy, C., Bejot, Y., Deltour, 381–388.
S., Jaillard, A., Niclot, P., Guillon, B., Moulin, T., Marque, P., Pariente, J., Arnaud, C., Dickens, M., Romero, L.M., Cyr, N.E., Dunn, I.C., Meddle, S.L., 2009. Chronic stress alters
Loubinoux, I., 2011. Fluoxetine for motor recovery after acute ischaemic stroke glucocorticoid receptor and mineralocorticoid receptor mRNA expression in the
(FLAME): a randomised placebo-controlled trial. Lancet Neurol. 10, 123–130. European starling (Sturnus vulgaris) brain. J. Neuroendocrinol. 21, 832–840.
Clayton, N.S., 2001. Hippocampal growth and maintenance depend on food-caching ex- Donahue, C.P., Jensen, R.V., Ochiishi, T., Eisenstein, I., Zhao, M., Shors, T., Kosik, K.S.,
perience in juvenile mountain chickadees (Poecile gambeli). Behav. Neurosci. 115, 2002. Transcriptional profiling reveals regulated genes in the hippocampus during
614–625. memory formation. Hippocampus 12, 821–833.
Colcombe, S.J., Kramer, A.F., Erickson, K.I., Scalf, P., McAuley, E., Cohen, N.J., Webb, A., Donahue, C.P., Kosik, K.S., Shors, T.J., 2006. Growth hormone is produced within the
Jerome, G.J., Marquez, D.X., Elavsky, S., 2004a. Cardiovascular fitness, cortical hippocampus where it responds to age, sex, and stress. Proc. Natl. Acad. Sci. USA 103,
plasticity, and aging. Proc. Natl. Acad. Sci. USA 101, 3316–3321. 6031–6036.
Colcombe, S.J., Kramer, A.F., Erickson, K.I., Scalf, P., McAuley, E., Cohen, N.J., Webb, A., Donkin, I., Versteyhe, S., Ingerslev, L.R., Qian, K., Mechta, M., Nordkap, L., Mortensen, B.,
Jerome, G.J., Marquez, D.X., Elavsky, S., 2004b. Cardiovascular fitness, cortical Appel, E.V., Jorgensen, N., Kristiansen, V.B., Hansen, T., Workman, C.T., Zierath,
plasticity, and aging. Proc. Natl. Acad. Sci. USA 101, 3316–3321. J.R., Barres, R., 2016. Obesity and bariatric surgery drive epigenetic variation of
Cole, D.C., Chung, Y., Gagnidze, K., Hajdarovic, K.H., Rayon-Estrada, V., Harjanto, D., spermatozoa in humans. Cell Metab. 23, 369–378.
Bigio, B., Gal-Toth, J., Milner, T.A., McEwen, B.S., Papavasiliou, F.N., Bulloch, K., Dore, S., Kar, S., Rowe, W., Quirion, R., 1997. Distribution and levels of [125I]IGF-I, [125I]
2017. Loss of APOBEC1 RNA-editing function inmicroglia exacerbates age-related IGF-II and [125I]insulin receptor binding sites in the hippocampus of aged memory-
CNS pathophysiology. Proc. Natl. Acad. Sci. USA. www.pnas.org/cgi/doi/10.1073/ unimpaired and -impaired rats. Neuroscience 80, 1033–1040.
pnas.171049311. Drevets, W.C., Price, J.L., Simpson Jr, J.R., Todd, R.D., Reich, T., Vannier, M., Raichle,
Conrad, C.D., Galea, L.A.M., Kuroda, Y., McEwen, B.S., 1996. Chronic stress impairs rat M.E., 1997. Subgenual prefrontal cortex abnormalities in mood disorders. Nature
spatial memory on the Y-Maze and this effect is blocked by tianeptine pre-treatment. 386, 824–827.
Behav. Neurosci. 110, 1321–1334. Driessen, M., Hermann, J., Stahl, K., Zwaan, M., Meier, S., Hill, A., Osterheider, M.,
Conrad, C.D., Lupien, S.J., McEwen, B.S., 1999a. Support for a bimodal role for type II Petersen, D., 2000. Magnetic resonance imaging volumes of the hippocampus and the
adrenal steroid receptors in spatial memory. Neurobiol. Learning Memory 72, 39–46. amygdala in women with borderline personality disorder and early traumatization.
Conrad, C.D., Magarinos, A.M., LeDoux, J.E., McEwen, B.S., 1999b. Repeated restraint Arch. Gen. Psychiat. 57, 1115–1122.
stress facilitates fear conditioning independently of causing hippocampal CA3 den- Du, J., Wang, Y., Hunter, R., Wei, Y., Blumenthal, R., Falke, C., Khairova, R., Zhou, R.,
dritic atrophy. Behav. Neurosci. 113, 902–913. Yuan, P., Machado-Vieira, R., McEwen, B.S., Manji, H.K., 2009. Dynamic regulation
Conrad, C.D., McLaughlin, K.J., Harman, J.S., Foltz, C., Wieczorek, L., Lightner, E., of mitochondrial function by glucocorticoids. Proc. Natl. Acad. Sci. USA 106,
Wright, R.L., 2007. Chronic glucocorticoids increase hippocampal vulnerability to 3543–3548.
neurotoxicity under conditions that produce CA3 dendritic retraction but fail to Duman, R.S., Heninger, G.R., Nestler, E.J., 1997. A molecular and cellular theory of de-
impair spatial recognition memory. J. Neurosci. 27, 8278–8285. pression. Arch. Gen. Psychiat. 54, 597–606.
Convit, A., Wolf, O.T., Tarshish, C., de Leon, M.J., 2003. Reduced glucose tolerance is Dumitriu, D., Rapp, P.R., McEwen, B.S., Morrison, J.H., 2010. Estrogen and the aging
associated with poor memory performance and hippocampal atrophy among normal brain: an elixir for the weary cortical network. Ann. N.Y. Acad. Sci. 1204, 104–112.
elderly. Proc. Natl. Acad. Sci. USA 100, 2019–2022. Eiland, L., Ramroop, J., Hill, M.N., Manley, J., McEwen, B.S., 2012. Chronic juvenile
D'Agostino, P.M., Gottfried-Blackmore, A., Anandasabapathy, N., Bulloch, K., 2012a. stress produces corticolimbic dendritic architectural remodeling and modulates
Brain dendritic cells: biology and pathology. Acta Neuropathol. 124, 599–614. emotional behavior in male and female rats. Psychoneuroendocrinology 37, 39–47.
D'Agostino, P.M., Kwak, C., Vecchiarelli, H.A., Toth, J.G., Miller, J.M., Masheeb, Z., Eiland, L., Romeo, R.D., 2012. Stress and the developing adolescent brain. Neuroscience.
McEwen, B.S., Bulloch, K., 2012b. Viral-induced encephalitis initiates distinct and Eme, R.F., 2007. Sex differences in child-onset, life-course-persistent conduct disorder. a
functional CD103+ CD11b+ brain dendritic cell populations within the olfactory review of biological influences. Clin. Psychol. Rev. 27, 607–627.
bulb. Proc. Natl. Acad. Sci. USA 109, 6175–6180. Engel, G.L., 1977. The need for a new medical model: a challenge for biomedicine.

25
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Science 196, 129–136. Gold, S.M., Dziobek, I., Sweat, V., Tirsi, A., Rogers, K., Bruehl, H., Tsui, W., Richardson,
Erickson, K.I., Voss, M.W., Prakash, R.S., Basak, C., Szabo, A., Chaddock, L., Kim, J.S., S., Javier, E., Convit, A., 2007. Hippocampal damage and memory impairments as
Heo, S., Alves, H., White, S.M., Wojcicki, T.R., Mailey, E., Vieira, V.J., Martin, S.A., possible early brain complications of type 2 diabetes. Diabetologia 50, 711–719.
Pence, B.D., Woods, J.A., McAuley, E., Kramer, A.F., 2011. Exercise training increases Gould, E., Cameron, H.A., Daniels, D.C., Woolley, C.S., McEwen, B.S., 1992. Adrenal
size of hippocampus and improves memory. Proc. Natl. Acad. Sci. USA 108, hormones suppress cell division in the adult rat dentate gyrus. J. Neurosci. 12,
3017–3022. 3642–3650.
Evans, G.W., Wachs, T.D., 2010. Chaos and Its Influence on Children's Development: An Gould, E., McEwen, B.S., 1993. Neuronal birth and death. Curr. Opin. Neurobiol. 3,
Ecological Perspective, first ed. American Psychological Association, 676–682.
Washington, DC. Gould, E., McEwen, B.S., Tanapat, P., Galea, L.A.M., Fuchs, E., 1997. Neurogenesis in the
Fadel, J.R., Reagan, L.P., 2016. Stop signs in hippocampal insulin signaling: the role of dentate gyrus of the adult tree shrew is regulated by psychosocial stress and NMDA
insulin resistance in structural, functional and behavioral deficits. Curr. Opin. Behav. receptor activation. J. Neurosci. 17, 2492–2498.
Sci. 9, 47–54. Gould, E., Woolley, C., Frankfurt, M., McEwen, B.S., 1990. Gonadal steroids regulate
Felger, J.C., Abe, T., Kaunzner, U.W., Gottfried-Blackmore, A., Gal-Toth, J., McEwen, B.S., dendritic spine density in hippocampal pyramidal cells in adulthood. J. Neurosci. 10,
Iadecola, C., Bulloch, K., 2010. Brain dendritic cells in ischemic stroke: time course, 1286–1291.
activation state, and origin. Brain Behav. Immun. 24, 724–737. Govindarajan, A., Rao, B.S.S., Nair, D., Trinh, M., Mawjee, N., Tonegawa, S., Chattarji, S.,
Felitti, V.J., Anda, R.F., Nordenberg, D., Williamson, D.F., Spitz, A.M., Edwards, V., Koss, 2006. Transgenic brain-derived neurotrophic factor expression causes both anxio-
M.P., Marks, J.S., 1998. Relationship of childhood abuse and household dysfunction genic and antidepressant effects. Proc. Natl. Acad. Sci. USA 103, 13208–13213.
to many of the leading causes of death in adults. The adverse childhood experiences Gray, J.D., Rubin, T.G., Hunter, R.G., McEwen, B.S., 2014. Hippocampal gene expression
(ACE) study. Am. J. Prev. Med. 14, 245–258. changes underlying stress sensitization and recovery. Mol. Psychiat. 19, 1171–1178.
Fischer, A.K., von Rosenstiel, P., Fuchs, E., Goula, D., Almeida, O.F., Czeh, B., 2002. The Greenough, W.T., Volkmar, F.R., 1973. Pattern of dendritic branching in occipital cortex
prototypic mineralocorticoid receptor agonist aldosterone influences neurogenesis in of rats reared in complex environments. Exp. Neurol. 40, 491–504.
the dentate gyrus of the adrenalectomized rat. Brain Res. 947, 290–293. Greenough, W.T., Volkmar, F.R., Juraska, J.M., 1973. Effects of rearing complexity on
Fraga, M.F., Ballestar, E., Paz, M.F., Ropero, S., Setien, F., Ballestar, M.L., Heine-Suner, D., dendritic branching in frontolateral and temporal cortex of the rat. Exp. Neurol. 41,
Cigudosa, J.C., Urioste, M., Benitez, J., Boix-Chornet, M., Sanchez-Aguilera, A., ling, 371–378.
C., Carlsson, E., Poulsen, P., Vaag, A., Stephan, Z., Spector, T.D., Wu, Y.-Z., Plass, C., Griffiths, B.B., Hunter, R.G., 2014. Neuroepigenetics of stress. Neuroscience 275,
Esteller, M., 2005. Epigenetic differences arise during the lifetime of monozygotic 420–435.
twins. Proc. Natl. Acad. Sci. USA 102, 10604–10609. Grillo, C.A., Mulder, P., Macht, V.A., Kaigler, K.F., Wilson, S.P., Wilson, M.A., Reagan,
Francis, D., Diorio, J., Liu, D., Meaney, M.J., 1999. Nongenomic transmission across L.P., 2014. Dietary restriction reverses obesity-induced anhedonia. Physiol. Behav.
generations of maternal behavior and stress responses in the rat. Science 286, 128, 126–132.
1155–1158. Grillo, C.A., Piroli, G.G., Lawrence, R.C., Wrighten, S.A., Green, A.J., Wilson, S.P., Sakai,
Francis, D.D., Szegda, K., Campbell, G., Martin, W.D., Insel, T.R., 2003. Epigenetic sources R.R., Kelly, S.J., Wilson, M.A., Mott, D.D., Reagan, L.P., 2015. Hippocampal insulin
of behavioral differences in mice. Nat. Neurosci. 6, 445–446. resistance impairs spatial learning and synaptic plasticity. Diabetes 64, 3927–3936.
Frankfurt, M., Gould, E., Wolley, C., McEwen, B.S., 1990. Gonadal steroids modify den- Gruene, T.M., Roberts, E., Thomas, V., Ronzio, A., Shansky, R.M., 2015. Sex-specific
dritic spine density in ventromedial hypothalamic neurons: a Golgi study in the adult neuroanatomical correlates of fear expression in prefrontal-amygdala circuits. Biol.
rat. Neuroendocrinology 51, 530–535. Psychiat. 78, 186–193.
Fredrickson, B.L., Grewen, K.M., Coffey, K.A., Algoe, S.B., Firestine, A.M., Arevalo, J.M., Guillemin, R., 1978. Peptides in the brain: the new endocrinology of the neuron. Science
Ma, J., Cole, S.W., 2013. A functional genomic perspective on human well-being. 202, 390–402.
Proc. Natl. Acad. Sci. USA 110, 13684–13689. Gunduz-Cinar, O., Hill, M.N., McEwen, B.S., Holmes, A., 2013. Amygdala FAAH and
Friedman, E.M., Hayney, M.S., Love, G.D., Urry, H.L., Rosenkranz, M.A., Davidson, R.J., anandamide: mediating protection and recovery from stress. Trends Pharmacol. Sci.
Singer, B.H., Ryff, C.D., 2005. Social relationships, sleep quality, and interleukin-6 in 34, 637–644.
aging women. Proc. Natl. Acad. Sci. USA 102, 18757–18762. Haan, M.N., 2006. Therapy insight: type 2 diabetes mellitus and the risk of late-onset
Fritz, I.B., Mcewen, B., 1959. Effects of carnitine on fatty-acid oxidation by muscle. Alzheimer's disease. Nat. Clin. Pract. Neurol. 2, 159–166.
Science 129, 334 334-334. Halfon, N., Larson, K., Lu, M., Tullis, E., Russ, S., 2014. Lifecourse health development:
Frodl, T., Meisenzahl, E.M., Zetzsche, T., Born, C., Jager, M., Groll, C., Bottlender, R., past, present and future. Maternal Child Health J. 18, 344–365.
Leinsinger, G., Moller, H.-J., 2003. Larger amygdala volumes in first depressive Hampel, H., Burger, K., Teipel, S.J., Bokde, A.L., Zetterberg, H., Blennow, K., 2008. Core
episode as compared to recurrent major depression and healthy control subjects. Biol. candidate neurochemical and imaging biomarkers of Alzheimer's disease. Alzheimer's
Psychiat. 53, 338–344. Dementia: J. Alzheimer's Assoc. 4, 38–48.
Gagnidze, K., Hajdarovic, K.H., Moskalenko, M., Karatsoreos, I.N., McEwen, B.S., Bulloch, Hamson, D.K., Wainwright, S.R., Taylor, J.R., Jones, B.A., Watson, N.V., Galea, L.A.,
K., 2016. Nuclear receptor REV-ERBalpha mediates circadian sensitivity to mortality 2013. Androgens increase survival of adult-born neurons in the dentate gyrus by an
in murine vesicular stomatitis virus-induced encephalitis. Proc. Natl. Acad. Sci. USA androgen receptor-dependent mechanism in male rats. Endocrinology 154,
113, 5730–5735. 3294–3304.
Galea, L.A., Wainwright, S.R., Roes, M.M., Duarte-Guterman, P., Chow, C., Hamson, D.K., Hanson, J.L., Hair, N., Shen, D.G., Shi, F., Gilmore, J.H., Wolfe, B.L., Pollak, S.D., 2013.
2013. Sex, hormones and neurogenesis in the hippocampus: hormonal modulation of Family poverty affects the rate of human infant brain growth. PLoS One 8, e80954.
neurogenesis and potential functional implications. J. Neuroendocrinol. 25, Hara, Y., Waters, E.M., McEwen, B.S., Morrison, J.H., 2015. Estrogen effects on cognitive
1039–1061. and synaptic health over the lifecourse. Physiol. Rev. 95, 785–807.
Galea, L.A., Wide, J.K., Barr, A.M., 2001. Estradiol alleviates depressive-like symptoms in Hara, Y., Yuk, F., Puri, R., Janssen, W.G., Rapp, P.R., Morrison, J.H., 2014. Presynaptic
a novel animal model of post-partum depression. Behav. Brain Res. 122, 1–9. mitochondrial morphology in monkey prefrontal cortex correlates with working
Galea, L.A.M., McEwen, B.S., Tanapat, P., Deak, T., Spencer, R.L., Dhabhar, F.S., 1997. memory and is improved with estrogen treatment. Proc. Natl. Acad. Sci. USA 111,
Sex differences in dendritic atrophy of CA3 pyramidal neurons in response to chronic 486–491.
restraint stress. Neuroscience 81, 689–697. Hara, Y., Yuk, F., Puri, R., Janssen, W.G., Rapp, P.R., Morrison, J.H., 2016. Estrogen
Gard, T., Holzel, B.K., Lazar, S.W., 2014a. The potential effects of meditation on age- restores multisynaptic boutons in the dorsolateral prefrontal cortex while promoting
related cognitive decline: a systematic review. Ann. N.Y. Acad. Sci. 1307, 89–103. working memory in aged rhesus monkeys. J. Neurosci. 36, 901–910.
Gard, T., Taquet, M., Dixit, R., Holzel, B.K., de Montjoye, Y.A., Brach, N., Salat, D.H., Harris, G.W., 1970. Effects of the nervous system on the pituitary-adrenal activity. Prog.
Dickerson, B.C., Gray, J.R., Lazar, S.W., 2014b. Fluid intelligence and brain func- Brain Res. 32, 86–88.
tional organization in aging yoga and meditation practitioners. Front. Aging Harvey, J., Shanley, L.J., O'Malley, D., Irving, A.J., 2005. Leptin: a potential cognitive
Neurosci. 6, 76. enhancer? Biochem. Soc. Trans. 33, 1029–1032.
Gee, K., Chang, W.-C., Brinton, R., McEwen, B.S., 1987. GABA-dependent modulation of Hebb, D.O., 1949. The Organization of Behavior; A Neuropsychological Theory. Wiley,
C1 ionophore by steroids in rat brain. Eur. J. Pharm. 136, 419–423. New York.
Gerlach, J., McEwen, B.S., 1972. Rat brain binds adrenal steroid hormone: radio- Hill, M.N., Kumar, S.A., Filipski, S.B., Iverson, M., Stuhr, K.L., Keith, J.M., Cravatt, B.F.,
autography of hippocampus with corticosterone. Science 175, 1133–1136. Hillard, C.J., Chattarji, S., McEwen, B.S., 2013. Disruption of fatty acid amide hy-
Gerlach, J., McEwen, B.S., Pfaff, D.W., Moskovitz, S., Ferin, M., Carmel, P., Zimmerman, drolase activity prevents the effects of chronic stress on anxiety and amygdalar mi-
E., 1976. Cells in regions of rhesus monkey brain and pituitary retain radioactive crostructure. Mol. Psychiat. 18, 1125–1135.
estradiol, corticosterone and cortisol differently. Brain Res. 103, 603–612. Hill, M.N., McEwen, B.S., 2010. Involvement of the endocannabinoid system in the
Gianaros, P.J., Jennings, J.R., Sheu, L.K., Greer, P.J., Kuller, L.H., Matthews, K.A., 2007. neurobehavioural effects of stress and glucocorticoids. Prog. Neuropsychopharmacol.
Prospective reports of chronic life stress predict decreased grey matter volume in the Biol. Psychiat. 34, 791–797.
hippocampus. NeuroImage 35, 795–803. Hill, M.N., McLaughlin, R.J., Bingham, B., Shrestha, L., Lee, T.T., Gray, J.M., Hillard, C.J.,
Gibbs, R.B., Hashash, A., Johnson, D.A., 1997. Effect of estrogen on potassium-stimulated Gorzalka, B.B., Viau, V., 2010. Endogenous cannabinoid signaling is essential for
acetylcholine release in the hippocampus and overlying cortex of adult rats. Brain stress adaptation. Proc. Natl. Acad. Sci. USA 107, 9406–9411.
Res. 749, 143–146. Hoekzema, E., Barba-Muller, E., Pozzobon, C., Picado, M., Lucco, F., Garcia-Garcia, D.,
Glennon, E., Kaunzner, U.W., Gagnidze, K., McEwen, B.S., Bulloch, K., 2015. Pituitary Soliva, J.C., Tobena, A., Desco, M., Crone, E.A., Ballesteros, A., Carmona, S.,
dendritic cells communicate immune pathogenic signals. Brain Behav. Immun. 50, Vilarroya, O., 2017. Pregnancy leads to long-lasting changes in human brain struc-
232–240. ture. Nat. Neurosci. 20, 287–296.
Gold, P.E., 1987. Sweet memories. Am. Sci. 75, 151–155. Hojo, Y., Hattori, T.-A., Enami, T., Furukawa, A., Suzuki, K., Ishii, H.-T., Mukai, H.,
Gold, P.W., Licinio, J., Pavlatou, M.G., 2013. Pathological parainflammation and en- Morrison, J.H., Janssen, W.G.M., Kominami, S., Harada, N., Kimoto, T., Kawato, S.,
doplasmic reticulum stress in depression: potential translational targets through the 2003. Adult male rat hippocampus synthesizes estradiol from pregnenolone by cy-
CNS insulin, klotho and PPAR-gamma systems. Mol. Psychiat. 18, 154–165. tochromes P45017a and P450 aromatase localized in neurons. Proc. Natl. Acad. Sci.

26
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

USA. Allen, P.B., Greengard, P., Luine, V., McEwen, B.S., 2004. Estrogen alters hippo-
Holzel, B.K., Carmody, J., Evans, K.C., Hoge, E.A., Dusek, J.A., Morgan, L., Pitman, R.K., campal dendritic spine shape and enhances synaptic protein immunoreactivity and
Lazar, S.W., 2010. Stress reduction correlates with structural changes in the amyg- spatial memory in female mice. Proc. Natl. Acad. Sci. USA 101, 2185–2190.
dala. Soc. Cogn. Affect. Neurosci. 5, 11–17. Lin, K.W., Wroolie, T.E., Robakis, T., Rasgon, N.L., 2015. Adjuvant pioglitazone for un-
Hunter, R.G., McCarthy, K.J., Milne, T.A., Pfaff, D.W., McEwen, B.S., 2009. Regulation of remitted depression: clinical correlates of treatment response. Psychiat. Res. 230,
hippocampal H3 histone methylation by acute and chronic stress. Proc. Natl. Acad. 846–852.
Sci. USA 106, 20912–20917. Liposits, Z., Bohn, M.C., 1993. Association of glucocorticoid receptor immunoreactivity
Hunter, R.G., McEwen, B.S., Pfaff, D.W., 2013. Environmental stress and transposon with cell membrane and transport vesicles in hippocampal and hypothalamic neurons
transcription in the mammalian brain. Mobile Gen. Elem. 3, e24555. of the rat. J. Neurosci. Res. 35, 14–19.
Hunter, R.G., Murakami, G., Dewell, S., Seligsohn, M., Baker, M.E., Datson, N.A., Liston, C., Cichon, J.M., Jeanneteau, F., Jia, Z., Chao, M.V., Gan, W.B., 2013. Circadian
McEwen, B.S., Pfaff, D.W., 2012. Acute stress and hippocampal histone H3 lysine 9 glucocorticoid oscillations promote learning-dependent synapse formation and
trimethylation, a retrotransposon silencing response. Proc. Natl. Acad. Sci. USA 109, maintenance. Nat. Neurosci. 16, 698–705.
17657–17662. Liston, C., Gan, W.B., 2011. Glucocorticoids are critical regulators of dendritic spine
Hunter, R.G., Seligsohn, M., Rubin, T.G., Griffiths, B.B., Ozdemir, Y., Pfaff, D.W., Datson, development and plasticity in vivo. Proc. Natl. Acad. Sci. USA 108, 16074–16079.
N.A., McEwen, B.S., 2016. Stress and corticosteroids regulate rat hippocampal mi- Liston, C., McEwen, B.S., Casey, B.J., 2009. Psychosocial stress reversibly disrupts pre-
tochondrial DNA gene expression via the glucocorticoid receptor. Proc. Natl. Acad. frontal processing and attentional control. Proc. Natl. Acad. Sci. USA 106, 912–917.
Sci. USA 113, 9099–9104. Liston, C., Miller, M.M., Goldwater, D.S., Radley, J.J., Rocher, A.B., Hof, P.R., Morrison,
Jensen, E., Jacobson, H., 1962. Basic guides to the mechanism of estrogen action. Rec. J.H., McEwen, B.S., 2006. Stress-induced alterations in prefrontal cortical dendritic
Prog. Horm. Res. 18, 387–408. morphology predict selective impairments in perceptual attentional set-shifting. J.
Joels, M., 2006. Corticosteroid effects in the brain: U-shape it. Trends Pharmacol. Sci. 27, Neurosci. 26, 7870–7874.
244–250. Lowy, M.T., Gault, L., Yamamoto, B.K., 1993. Adrenalectomy attenuates stress-induced
Kagan, J., 2016. An overly permissive extension. Perspect. Psychol. Sci.: J. Assoc. Psychol. elevations in extracellular glutamate concentrations in the hippocampus. J.
Sci. 11, 442–450. Neurochem. 61, 1957–1960.
Kaplan, M.S., 2001. Environment complexity stimulates visual cortex neurogenesis: death Lu, X.-Y., Kim, C.S., Frazer, A., Zhang, W., 2006. Leptin: a potential novel antidepressant.
of a dogma and a research career. Trends Neurosci. 24, 617–620. Proc. Natl. Acad. Sci. USA 103, 1593–1598.
Kaplan, M.S., Bell, D.H., 1983. Neuronal proliferation in the 9-month-old rodent-radio- Luine, V., Martinez, C., Villegas, M., Magarinos, A.M., McEwen, B.S., 1996. Restraint
autographic study of granule cells in the hippocampus. Exp. Brain Res. 52, 1–5. stress reversibly enhances spatial memory performance. Physiol. Behav. 59, 27–32.
Karatsoreos, I.N., Bhagat, S., Bloss, E.B., Morrison, J.H., McEwen, B.S., 2011. Disruption Luine, V., Villegas, M., Martinez, C., McEwen, B.S., 1994. Repeated stress causes re-
of circadian clocks has ramifications for metabolism, brain, and behavior. Proc. Natl. versible impairments of spatial memory performance. Brain Res. 639, 167–170.
Acad. Sci. USA 108, 1657–1662. Luine, V.N., Khylchevcskaya, R.I., McEwen, B.S., 1975. Effect of gonadal steroids on
Karst, H., Berger, S., Turiault, M., Tronche, F., Schutz, G., Joels, M., 2005. activities of monoamine oxidase and choline acetylase in rat brain. Brain Res. 86,
Mineralocorticoid receptors are indispensable for nongenomic modulation of hip- 293–306.
pocampal glutamate transmission by corticosterone. Proc. Natl. Acad. Sci. USA 102, Lupien, S.J., de Leon, M., de Santi, S., Convit, A., Tarshish, C., Nair, N.P.V., Thakur, M.,
19204–19207. McEwen, B.S., Hauger, R.L., Meaney, M.J., 1998. Cortisol levels during human aging
Kastin, A.J., Akerstrom, V., 2001. Glucose and insulin increase the transport of leptin predict hippocampal atrophy and memory deficits. Nat. Neurosci. 1, 69–73.
through the blood-brain barrier in normal mice but not in streptozotocin-diabetic Lupien, S.J., Parent, S., Evans, A.C., Tremblay, R.E., Zelazo, P.D., Corbo, V., Pruessner,
mice. Neuroendocrinology 73, 237–242. J.C., Seguin, J.R., 2011. Larger amygdala but no change in hippocampal volume in
Kaunzner, U.W., Miller, M.M., Gottfried-Blackmore, A., Gal-Toth, J., Felger, J.C., 10-year-old children exposed to maternal depressive symptomatology since birth.
McEwen, B.S., Bulloch, K., 2012. Accumulation of resident and peripheral dendritic Proc. Natl. Acad. Sci. USA 108, 14324–14329.
cells in the aging CNS. Neurobiol. Aging 33 (681–693), e681. MacLusky, N., McEwen, B.S., 1978. Oestrogen modulates progestin receptor concentra-
Kelly, M.J., Levin, E.R., 2001. Rapid actions of plasma membrane estrogen receptors. tions in some rat brain regions but not in others. Nature 274, 276–278.
Trends Endo. Metab. 12, 152–156. MacQueen, G.M., Campbell, S., McEwen, B.S., Macdonald, K., Amano, S., Joffe, R.T.,
Kempermann, G., Gage, F.H., 1999. New nerve cells for the adult brain. Sci. Am. 280, Nahmias, C., Young, L.T., 2003. Course of illness, hippocampal function, and hip-
48–53. pocampal volume in major depression. Proc. Natl. Acad. Sci. USA 100, 1387–1392.
Kempermann, G., Kuhn, H.G., Gage, F.H., 1997. More hippocampal neurons in adult mice MacQueen, G.M., Yucel, K., Taylor, V.H., Macdonald, K., Joffe, R., 2008. Posterior hip-
living in an enriched environment. Nature 586, 493–495. pocampal volumes are associated with remission rates in patients with major de-
Kerr, J.E., Allore, R.J., Beck, S.G., Handa, R.J., 1995. Distribution and hormonal reg- pressive disorder. Biol. Psychiat.
ulation of androgen receptor (AR) and AR messenger ribonucleic acid in the rat Magarinos, A.M., McEwen, B.S., 1995. Stress-induced atrophy of apical dendrites of
hippocampus. Endocrinology 136, 3213–3221. hippocampal CA3c neurons: involvement of glucocorticoid secretion and excitatory
Kessler, R.C., McGonagle, K.A., Nelson, C.B., Hughes, M., Swartz, M., Blazer, D.G., 1994. amino acid receptors. Neuroscience 69, 89–98.
Sex and depression in the National Comorbidity Survey. II: Cohort effects. J. Affect. Maguire, E.A., Frackowiak, R.S.J., Frith, C.D., 1997. Recalling routes around London:
Disord. 30, 15–26. activation of the right hippocampus in taxi drivers. J. Neurosci. 17, 7103–7110.
Kessler, R.C., McGonagle, K.A., Swartz, M., Blazer, D.G., Nelson, C.B., 1993. Sex and Manolopoulos, K.N., Klotz, L.O., Korsten, P., Bornstein, S.R., Barthel, A., 2010. Linking
depression in the National Comorbidity Survey. I: Lifetime prevalence, chronicity and Alzheimer's disease to insulin resistance: the FoxO response to oxidative stress. Mol.
recurrence. J. Affect. Disord. 29, 85–96. Psychiat. 15, 1046–1052.
Kinoshita, Y., Hunter, R.G., Gray, J.D., Mesias, R., McEwen, B.S., Benson, D.L., Kohtz, Marrocco, J., McEwen, B.S., 2016. Sex in the brain: hormones and sex differences.
D.S., 2014. Role for NUP62 depletion and PYK2 redistribution in dendritic retraction Dialogues Clin. Neurosci. 18, 373–383.
resulting from chronic stress. Proc. Natl. Acad. Sci. USA 111, 16130–16135. Marrocco, J., Petty, G.H., Ríos, M.B., Gray, J.D., Kogan, J.F., Waters, E.M., Schmidt, E.F.,
Kovacs, E.G., MacLusky, N.J., Leranth, C., 2003. Effects of testosterone on hippocampal Lee, F.S., McEwen, B.S., 2017. A sexually dimorphic pre-stressed translational sig-
CA1 spine synaptic density in the male rat are inhibited by fimbria/fornix transec- nature in CA3 pyramidal neurons of BDNF Val66Met mice. Nat. Commun. 8 (1), 808.
tion. Neuroscience 122, 807–810. http://dx.doi.org/10.1038/s41467-017-01014-4.
Kral, J.G., Biron, S., Simard, S., Hould, F.-S., Lebel, S., Marceau, S., Marceau, P., 2006. Marsland, A.L., Gianaros, P.J., Abramowitch, S.M., Manuck, S.B., Hariri, A.R., 2008.
Large maternal weight loss from obesity surgery prevents transmission of obesity to Interleukin-6 covaries inversely with hippocampal grey matter volume in middle-
children who were followed for 2 to 18 years. Pediatrics 118, 1644–1649. aged adults. Biol. Psychiat. 64, 484–490.
Kramer, A.F., Hahn, S., Cohen, N.J., Banich, M.T., McAuley, E., Harrison, C.R., Chason, J., Martin, K.P., Wellman, C.L., 2011. NMDA receptor blockade alters stress-induced den-
Vakil, E., Bardell, L., Boileau, R.A., Colcombe, A., 1999. Ageing, fitness and neuro- dritic remodeling in medial prefrontal cortex. Cereb. Cortex 21, 2366–2373.
cognitive function. Nature 400, 418–419. Matsui, T., Omuro, H., Liu, Y.F., Soya, M., Shima, T., McEwen, B.S., Soya, H., 2017.
Labombarda, F., Jure, I., Gonzalez, S., Lima, A., Roig, P., Guennoun, R., Schumacher, M., Astrocytic glycogen-derived lactate fuels the brain during exhaustive exercise to
De Nicola, A.F., 2015. A functional progesterone receptor is required for im- maintain endurance capacity. Proc. Natl. Acad. Sci. USA.
munomodulation, reduction of reactive gliosis and survival of oligodendrocyte pre- Matys, T., Pawlak, R., Matys, E., Pavlides, C., McEwen, B.S., Strickland, S., 2004. Tissue
cursors in the injured spinal cord. J. Steroid Biochem. Mol. Biol. 154, 274–284. plasminogen activator promotes the effects of corticotropin releasing factor on the
Lee, S.J., Romeo, R.D., Svenningsson, P., Campomanes, C.R., Allen, P.B., Greengard, P., amygdala and anxiety-like behavior. Proc. Natl. Acad. Sci. USA 101, 16345–16350.
mcEwen, B.S., 2004. Estradiol affects spinophilin protein differently in gonadecto- McCall, T., Weil, Z.M., Nacher, J., Bloss, E.B., El Maarouf, A., Rutishauser, U., McEwen,
mized males and females. Neuroscience 127, 983–988. B.S., 2013. Depletion of polysialic acid from neural cell adhesion molecule (PSA-
Leranth, C., Petnehazy, O., MacLusky, N.J., 2003. Gonadal hormones affect spine synaptic NCAM) increases CA3 dendritic arborization and increases vulnerability to ex-
density in the CA1 hippocampal subfield of male rats. J. Neurosci. 23, 1588–1592. citotoxicity. Exp. Neurol. 241, 5–12.
Leuner, B., Gould, E., Shors, T.J., 2006. Is there a link between adult neurogenesis and McCullough, L.D., Blizzard, K., Simpson, E.R., Oz, O.K., Hurn, P.D., 2003. Aromatase
learning? Hippocampus 26, 216–224. cytochrome P450 and extragonadal estrogen play a role in ischemic neuroprotection.
Levin, E.R., Hammes, S.R., 2016. Nuclear receptors outside the nucleus: extranuclear J. Neurosci. 23, 8701–8705.
signalling by steroid receptors. Nat. Rev. Mol. Cell Biol. 17, 783–797. McEwen, B.S., 1976. Steroid hormone receptors in developing and mature brain tissue. In:
Levine, S., Haltmeyer, G.C., Karas, G.G., Denenberg, V.H., 1967. Physiological and be- Snyder, S., McEwen, B.S. (Eds.), Neurotransmitters, Hormones and Receptors: Novel
havioral effects of infantile stimulation. Physiol. Behav. 2, 55–59. Approaches. Society of Neuroscience, pp. 50–66.
Lewis, C., McEwen, B.S., Frankfurt, M., 1995. Estrogen-induction of dendritic spines in McEwen, B.S., 1998. Protective and Damaging Effects of Stress Mediators. N. Engl. J.
ventromedial hypothalamus and hippocampus: effects of neonatal aromatase Med. 338, 171–179.
blockade and adult castration. Dev. Brain Res. 87, 91–95. McEwen, B.S., 1999. Stress and hippocampal plasticity. Annu. Rev. Neurosci. 22,
Li, C., Brake, W.G., Romeo, R.D., Dunlop, J.C., Gordon, M., Buzescu, R., Magarinos, A.M., 105–122.

27
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

McEwen, B.S., 2006. Protective and damaging effects of stress mediators: central role of estrogen binding sites in the rat forebrain and autonomic medullary areas.
the brain. Dialogues Clin. Neurosci. 8, 367–381. Endocrinology 149, 3306–3312.
McEwen, B.S., 2007. Physiology and neurobiology of stress and adaptation: central role of Milner, T.A., McEwen, B.S., Hayashi, S., Li, C.J., Reagen, L., Alves, S.E., 2001.
the brain. Physiol. Rev. 87, 873–904. Ultrastructural evidence that hippocampal alpha estrogen receptors are located at
McEwen, B.S., 2010. Stress, sex, and neural adaptation to a changing environment: me- extranuclear sites. J. Comp. Neurol. 429, 355–371.
chanisms of neuronal remodeling. Ann. N.Y. Acad. Sci. 1204 (Suppl.), E38–E59. Mitra, R., Jadhav, S., McEwen, B.S., Vyas, A., Chattarji, S., 2005. Stress duration mod-
McEwen, B.S., 2012. Brain on stress: how the social environment gets under the skin. ulates the spatiotemporal patterns of spine formation in the basolateral amygdala.
Proc. Natl. Acad. Sci. USA 109 (Suppl. 2), 17180–17185. Proc. Natl. Acad. Sci. USA 102, 9371–9376.
McEwen, B.S., 2015. Preserving neuroplasticity: role of glucocorticoids and neuro- Mitterling, K.L., Spencer, J.L., Dziedzic, N., Shenoy, S., McCarthy, K., Waters, E.M.,
trophins via phosphorylation. Proc. Natl. Acad. Sci. USA 112, 15544–15545. McEwen, B.S., Milner, T.A., 2010. Cellular and subcellular localization of estrogen
McEwen, B.S., 2016. Stress-induced remodeling of hippocampal CA3 pyramidal neurons. and progestin receptor immunoreactivities in the mouse hippocampus. J. Comp.
Brain Res. 1645, 50–54. Neurol. 518, 2729–2743.
McEwen, B.S., 2017. Allostasis and the epigenetics of brain and body health over the life Moffitt, T.E., Arseneault, L., Belsky, D., Dickson, N., Hancox, R.J., Harrington, H., Houts,
course: the brain on stress. JAMA Psychiat. R., Poulton, R., Roberts, B.W., Ross, S., Sears, M.R., Thomson, W.M., Caspi, A., 2011.
McEwen, B.S., Akama, K.T., Spencer-Segal, J.L., Milner, T.A., Waters, E.M., 2012. A gradient of childhood self-control predicts health, wealth, and public safety. Proc.
Estrogen effects on the brain: actions beyond the hypothalamus via novel mechan- Natl. Acad. Sci. USA 108, 2693–2698.
isms. Behav. Neurosci. 126, 4–16. Molet, J., Maras, P.M., Avishai-Eliner, S., Baram, T.Z., 2014. Naturalistic rodent models of
McEwen, B.S., Getz, L., 2013. Lifetime experiences, the brain and personalized medicine: chronic early-life stress. Dev. Psychobiol. 56, 1675–1688.
an integrative perspective. Metabolism 62 (Suppl. 1), S20–S26. Moon, H.Y., Becke, A., Berron, D., Becker, B., Sah, N., Benoni, G., Janke, E., Lubejko, S.T.,
McEwen, B.S., Gray, J., Nasca, C., 2015a. Recognizing resilience: learning from the effects Greig, N.H., Mattison, J.A., Duzel, E., van Praag, H., 2016. Running-induced systemic
of stress on the brain. Neurobiol. Stress 1, 1–11. cathepsin B secretion is associated with memory function. Cell Metab. 24, 332–340.
McEwen, B.S., Gray, J.D., Nasca, C., 2015b. 60 years of neuroendocrinology: redefining Moriceau, S., Sullivan, R.M., 2004. Corticosterone influences on mammalian neonatal
neuroendocrinology: stress, sex and cognitive and emotional regulation. J. sensitive-period learning. Behav. Neurosci. 118, 274–281.
Endocrinol. 226, T67–T83. Moutsatsou, P., Psarra, A.-M.G., Tsiapara, A., Paraskevakou, H., Davaris, P., Sekeris, C.E.,
McEwen, B.S., Karatsoreos, I.N., 2015. Sleep deprivation and circadian disruption: stress, 2001. Localization of the glucocorticoid receptor in rat brain mitochondria. Arch.
allostasis, and allostatic load. Sleep Med. Clin. 10, 1–10. Biochem. Biophys. 386, 69–78.
McEwen, B.S., McEwen, C.A., 2016. Response to Jerome Kagan's Essay on Stress (2016). Mucha, M., Skrzypiec, A.E., Schiavon, E., Attwood, B.K., Kucerova, E., Pawlak, R., 2011.
Perspect. Psychol. Sci.: J. Assoc. Psychol. Sci. 11, 451–455. Lipocalin-2 controls neuronal excitability and anxiety by regulating dendritic spine
McEwen, B.S., McKittrick, C.R., Tamashiro, K.L., Sakai, R.R., 2015c. The brain on stress: formation and maturation. Proc. Natl. Acad. Sci. USA 108, 18436–18441.
insight from studies using the Visible Burrow System. Physiol. Behav. 146, 47–56. Murphy, B.E.P., 1991. Treatment of major depression with steroid suppressive drugs. J.
McEwen, B.S., Milner, T.A., 2007. Hippocampal formation: shedding light on the influ- Steroid Biochem. Mol. Biol. 39, 239–244.
ence of sex and stress on the brain. Brain Res. Rev. 55, 343–355. Nacher, J., Guirado, R., Castillo-Gomez, E., 2013. Structural plasticity of interneurons in
McEwen, B.S., Milner, T.A., 2017. Understanding the broad influence of sex hormones the adult brain: role of PSA-NCAM and implications for psychiatric disorders.
and sex differences in the brain. J. Neurosci. Res. 95, 24–39. Neurochem. Res. 38, 1122–1133.
McEwen, B.S., Plapinger, L., 1970. Association of corticosterone-1,2 3H with macro- Nasca, C., Xenos, D., Barone, Y., Caruso, A., Scaccianoce, S., Matrisciano, F., Battaglia, G.,
molecules extracted from brain cell nuclei. Nature 226, 263–264. Mathe, A.A., Pittaluga, A., Lionetto, L., Simmaco, M., Nicoletti, F., 2013. L-acet-
McEwen, B.S., Sakai, R.R., Spencer, R.L., 1993. Adrenal steroid effects on the brain: ylcarnitine causes rapid antidepressant effects through the epigenetic induction of
versatile hormones with good and bad effects. In: Schulkin, J. (Ed.), Hormonally- mGlu2 receptors. Proc. Natl. Acad. Sci. USA 110, 4804–4809.
Induced Changes in Mind and Brain. Academic Press, San Diego, pp. 157–189. Nasca, C., Zelli, D., Bigio, B., Piccinin, S., Scaccianoce, S., Nistico, R., McEwen, B.S., 2015.
McEwen, B.S., Seeman, T., 1999. Protective and damaging effects of mediators of stress: Stress dynamically regulates behavior and glutamatergic gene expression in hippo-
elaborating and testing the concepts of allostasis and allostatic load. Ann. N.Y. Acad. campus by opening a window of epigenetic plasticity. Proc. Natl. Acad. Sci. USA 112,
Sci. 896, 30–47. 14960–14965.
McEwen, B.S., Stellar, E., 1993. Stress and the individual. Mechanisms leading to disease. Nishijima, T., Piriz, J., Duflot, S., Fernandez, A.M., Gaitan, G., Gomez-Pinedo, U.,
Arch. Intern. Med. 153, 2093–2101. Verdugo, J.M., Leroy, F., Soya, H., Nunez, A., Torres-Aleman, I., 2010. Neuronal
McEwen, B.S., Weiss, J., Schwartz, L., 1968. Selective retention of corticosterone by activity drives localized blood-brain-barrier transport of serum insulin-like growth
limbic structures in rat brain. Nature 220, 911–912. factor-I into the CNS. Neuron 67, 834–846.
McEwen, B.S., Wingfield, J.C., 2003. The concept of allostasis in biology and biomedicine. Norman, A.W., Song, X.D., Zanello, L., Bula, C., Okamura, W.H., 1999. Rapid and
Horm. Behav. 43, 2–15. genomic biological responses are mediated by different shapes of the agonist steroid
McEwen, B.S., Wingfield, J.C., 2010. What is in a name? Integrating homeostasis, allos- hormone, 1á,25(OH)2 vitamin D3. Steroids 64, 120–128.
tasis and stress. Horm. Behav. 57, 105–111. Nottebohm, F., 2002. Why are some neurons replaced in adult brain? J. Neurosci. 22,
McGowan, P.O., Sasaki, A., D'Alessio, A.C., Dymov, S., Labonte, B., Szyf, M., Turecki, G., 624–628.
Meaney, M.J., 2009. Epigenetic regulation of the glucocorticoid receptor in human Nyberg, F., 2000. Growth hormone in the brain: characteristics of specific brain targets
brain associates with childhood abuse. Nat. Neurosci. 12, 241–243. for the hormone and their functional significance. Front. Neuroendocrin. 21,
McIntyre, R.S., Kenna, H.A., Nguyen, H.T., Law, C.W., Sultan, F., Woldeyohannes, H.O., 330–348.
Adams, A.K., Cheng, J.S., Lourenco, M., Kennedy, S.H., Rasgon, N.L., 2010. Brain Okamoto, M., Hojo, Y., Inoue, K., Matsui, T., Kawato, S., McEwen, B.S., Soya, H., 2012.
volume abnormalities and neurocognitive deficits in diabetes mellitus: points of pa- Mild exercise increases dihydrotestosterone in hippocampus providing evidence for
thophysiological commonality with mood disorders? Adv. Ther. 27, 63–80. androgenic mediation of neurogenesis. Proc. Natl. Acad. Sci. USA 109, 13100–13105.
McNay, E.C., Fries, T.M., Gold, P.E., 2000. Decreases in rat extracellular hippocampal Ott, A., Stolk, R.P., Hofman, A., van Harskamp, F., Grobbee, D.E., Breteler, M.M.B., 1996.
glucose concentration associated with cognitive demand during a spatial task. Proc. Association of diabetes mellitus and dementia: the Rotterdam study. Diabetologia 39,
Natl. Acad. Sci. USA 97, 2881–2885. 1392–1397.
McNay, E.C., Gold, P.E., 2001. Age-related differences in hippocampal extracellular fluid Pan, W., Yu, Y., Cain, C.M., Nyberg, F., Couraud, P.O., Kastin, A.J., 2005. Permeation of
glucose concentration during behavioral testing and following systemic glucose ad- growth hormone across the blood-brain barrier. Endocrinology 146, 4898–4904.
ministration. J. Gerontol. 56A, B66–B71. Parnavelas, J., Lynch, G., Brecha, N., Cotman, C., GLobus, A., 1974. Spine loss and re-
Meaney, M., Aitken, D., Berkel, H., Bhatnagar, S., Sapolsky, R., 1988. Effect of neonatal growth in hippocampus following deafferentation. Nature 248, 71–73.
handling of age-related impairments associated with the hippocampus. Science 239, Pattwell, S.S., Bath, K.G., Casey, B.J., Ninan, I., Lee, F.S., 2011. Selective early-acquired
766–768. fear memories undergo temporary suppression during adolescence. Proc. Natl. Acad.
Meaney, M.J., Aitken, D.H., Bodnoff, S.R., Iny, L.J., Tatarewicz, J.E., Sapolsky, R.M., Sci. USA 108, 1182–1187.
2013. Early postnatal handling alters glucocorticoid receptor concentrations in se- Pattwell, S.S., Liston, C., Jing, D., Ninan, I., Yang, R.R., Witztum, J., Murdock, M.H.,
lected brain regions. Behav. Neurosci. 127, 637–641. Dincheva, I., Bath, K.G., Casey, B.J., Deisseroth, K., Lee, F.S., 2016. Dynamic changes
Meaney, M.J., Sapolsky, R.M., McEwen, B.S., 1985a. The development of the gluco- in neural circuitry during adolescence are associated with persistent attenuation of
corticoid receptor system in the rat limbic brain. I. Ontogeny and autoregulation. fear memories. Nat. Commun. 7, 11475.
Brain Res. 350, 159–164. Pavlides, C., Watanabe, Y., Magarinos, A.M., McEwen, B.S., 1995. Opposing role of
Meaney, M.J., Sapolsky, R.M., McEwen, B.S., 1985b. The development of the gluco- adrenal steroid Type I and Type II receptors in hippocampal long-term potentiation.
corticoid receptor system in the rat limbic brain. II. An autoradiographic study. Brain Neuroscience 68, 387–394.
Res. 350, 165–168. Pawlak, R., Magarinos, A.M., Melchor, J., McEwen, B., Strickland, S., 2003. Tissue
Mehler, M.F., 2008. Epigenetic principles and mechanisms underlying nervous system plasminogen activator in the amygdala is critical for stress-induced anxiety-like be-
functions in health and disease. Prog. Neurobiol. 86, 305–341. havior. Nat. Neurosci. 6, 168–174.
Mehler, M.F., Mattick, J.S., 2007. Noncoding RNAs and RNA editing in brain develop- Pawlak, R., Rao, B.S.S., Melchor, J.P., Chattarji, S., McEwen, B., Strickland, S., 2005.
ment, functional diversification, and neurological disease. Physiol. Rev. 87, 799–823. Tissue plasminogen activator and plasminogen mediate stress-induced decline of
Merzenich, M., 1998. Long-term change of mind. Science 282, 1062–1063. neuronal and cognitive functions in the mouse hippocampus. Proc. Natl. Acad. Sci.
Milner, T.A., Burstein, S.R., Marrone, G.F., Khalid, S., Gonzalez, A.D., Williams, T.J., USA 102, 18201–18206.
Schierberl, K.C., Torres-Reveron, A., Gonzales, K.L., McEwen, B.S., Waters, E.M., Pawluski, J.L., Galea, L.A.M., 2007. Reproductive experience alters hippocampal neuro-
2013. Stress differentially alters mu opioid receptor density and trafficking in par- genesis during the postpartum period in the dam. Neuroscience 149, 53–67.
valbumin-containing interneurons in the female and male rat hippocampus. Synapse Pereira, A.C., Gray, J.D., Kogan, J.F., Davidson, R.L., Rubin, T.G., Okamoto, M., Morrison,
67, 757–772. J.H., McEwen, B.S., 2016. Age and Alzheimer's disease gene expression profiles re-
Milner, T.A., Lubbers, L.S., Alves, S.E., McEwen, B.S., 2008. Nuclear and extranuclear versed by the glutamate modulator riluzole. Mol. Psychiat.

28
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

Pereira, A.C., Lambert, H.K., Grossman, Y.S., Dumitriu, D., Waldman, R., Jannetty, S.K., 23, 4479–4490.
Calakos, K., Janssen, W.G., McEwen, B.S., Morrison, J.H., 2014. Glutamatergic reg- Russo, S.J., Murrough, J.W., Han, M.H., Charney, D.S., Nestler, E.J., 2012. Neurobiology
ulation prevents hippocampal-dependent age-related cognitive decline through of resilience. Nat. Neurosci. 15, 1475–1484.
dendritic spine clustering. Proc. Natl. Acad. Sci. USA 111, 18733–18738. Rutishauser, U., 2008. Polysialic acid in the plasticity of the developing and adult ver-
Pereira, A.C., Mao, X., Jiang, C.S., Kang, G., Milrad, S., McEwen, B.S., Krieger, A.C., tebrate nervous system. Nat. Rev. Neurosci. 9, 26–35.
Shungu, D.C., 2017. Dorsolateral prefrontal cortex GABA deficit in older adults with Sachar, E.J., Hellman, L., Roffwarg, H.P., Halpern, F.S., Fukushima, D.K., Gallagher, T.F.,
sleep-disordered breathing. Proc. Natl. Acad. Sci. USA 114 (38), 10250–10255. 1973. Disrupted 24-hour patterns of cortisol secretion in psychotic depression. Arch.
http://dx.doi.org/10.1073/pnas.1700177114. Gen. Psychiarty 28, 19–24.
Pfaff, D.W., 1980. Estrogens and Brain Function. Springer-Verlag, New York. Sandi, C., 2004. Stress, cognitive impairment and cell adhesion molecules. Nat. Rev./
Pfaff, D.W., Keiner, M., 1973. Atlas of estradiol-concentrating cells in the central nervous Neurosci. 5, 917–930.
system of the female rat. J. Comp. Neurol. 151, 121–158. Sandstrom, N.J., Williams, C.L., 2001. Memory retention is modulated by acute estradiol
Pham, K., Nacher, J., Hof, P.R., McEwen, B.S., 2003. Repeated restraint stress suppresses and progesterone replacement. Behav. Neurosci. 115, 384–393.
neurogenesis and induces biphasic PSA-NCAM expression in the adult rat dentate Sapolsky, R., 1992. Stress, the Aging Brain and the Mechanisms of Neuron Death, vol. 1.
gyrus. Eur. J. Neurosci. 17, 879–886. Cambridge MIT Press, p. 423.
Picard, M., Juster, R.P., McEwen, B.S., 2014. Mitochondrial allostatic load puts the 'gluc' Sapolsky, R.M., Krey, L.C., McEwen, B.S., 1986. The neuroendocrinology of stress and
back in glucocorticoids. Nat. Rev. Endocrinol. 10, 303–310. aging: the glucocorticoid cascade hypothesis. Endocr. Rev. 7, 284–301.
Picard, M., McEwen, B.S., 2014. Mitochondria impact brain function and cognition. Proc. Schally, A.V., Arimura, A., Kastin, A.J., 1973. Hypothalamic regulatory hormones.
Natl. Acad. Sci. USA 111, 7–8. Science 179, 341–350.
Picard, M., McManus, M.J., Gray, J.D., Nasca, C., Moffat, C., Kopinski, P.K., Seifert, E.L., Schelling, G., Kilger, E., Roozendaal, B., de Quervain, D.J.-F., Briegel, J., Dagge, A.,
McEwen, B.S., Wallace, D.C., 2015. Mitochondrial functions modulate neuroendo- Rothenhausler, H.-B., Krauseneck, T., Nollert, G., Kapfhammer, H.-P., 2004. Stress
crine, metabolic, inflammatory, and transcriptional responses to acute psychological doses of hydrocortisone, traumatic memories, and symptoms of posttraumatic stress
stress. Proc. Natl. Acad. Sci. USA 112, E6614–E6623. disorder in patients after cardiac surgery: a randomized study. Biol. Psychiat. 55,
Piroli, G.G., Grillo, C.A., Reznikov, L.R., Adams, S., McEwen, B.S., Charron, M.J., Reagan, 627–633.
L.P., 2007. Corticosterone impairs insulin-stimulated translocation of GLUT4 in the Schumacher, M., Hussain, R., Gago, N., Oudinet, J.P., Mattern, C., Ghoumari, A.M., 2012.
rat hippocampus. Neuroendocrinology. Progesterone synthesis in the nervous system: implications for myelination and
Pitman, R.K., 2001. Hippocampal diminution in PTSD: more (or less?) than meets the eye. myelin repair. Front. Neurosci. 6, 10.
Hippocampus 11, 73–74. Seeman, T., Epel, E., Gruenewald, T., Karlamangla, A., McEwen, B.S., 2010a. Socio-eco-
Popoli, M., Yan, Z., McEwen, B.S., Sanacora, G., 2012. The stressed synapse: the impact of nomic differentials in peripheral biology: cumulative allostatic load. Ann. NY Acad.
stress and glucocorticoids on glutamate transmission. Nat. Rev. Neurosci. 13, 22–37. Sci. 1186, 223–239.
Poskitt, D.C., Barnett, J., Duffey, K., Lee, A.K., Kimpton, W.G., Muller, H.K., 1984. Stress Seeman, T., Epel, E., Gruenewald, T., Karlamangla, A., McEwen, B.S., 2010b. Socio-
related involution of lymphoid tissues in Australian marsupial mice. Immunobiology economic differentials in peripheral biology: cumulative allostatic load. Ann. N.Y.
166, 286–295. Acad. Sci. 1186, 223–239.
Priebe, K., Romeo, R.D., Francis, D.D., Sisti, H.M., Mueller, A., McEwen, B.S., Brake, W.G., Seeman, T., Gruenewald, T., Karlamangla, A., Sidney, S., Liu, K., McEwen, B., Schwartz,
2005. Maternal influences on adult stress and anxiety-like behavior in C57BL/6J and J., 2010c. Modeling multisystem biological risk in young adults: the Coronary Artery
BALB/CJ mice: a cross-fostering study. Dev. Psychobiol. 47, 398–407 (Wiley Risk Development in Young Adults Study. Am. J. Hum. Biol. 22, 463–472.
Periodicals, Inc.). Shansky, R.M., Hamo, C., Hof, P.R., Lou, W., McEwen, B.S., Morrison, J.H., 2010.
Pulford, B.E., Ishii, D.N., 2001. Uptake of circulating insulin-like growth factors (IGFs) Estrogen promotes stress sensitivity in a prefrontal cortex-amygdala pathway. Cereb.
into cerebrospinal fluid appears to be independent of the IGF receptors as well as IGF- Cortex 20, 2560–2567.
binding proteins. Endocrinology 142, 213–220. Sheline, Y.I., 2003. Neuroimaging studies of mood disorder effects on the brain. Biol.
Radley, J.J., Rocher, A.B., Janssen, W.G.M., Hof, P.R., McEwen, B.S., Morrison, J.H., Psychiat. 54, 338–352.
2005. Reversibility of apical dendritic retraction in the rat medial prefrontal cortex Sheline, Y.I., Gado, M.H., Price, J.L., 1998. Amygdala core nuclei volumes are decreased
following repeated stress. Exp. Neurol. 196, 199–203. in recurrent major depression. NeuroReport 9, 2023–2028.
Rao, R.P., Anilkumar, S., McEwen, B.S., Chattarji, S., 2012. Glucocorticoids protect Sheline, Y.I., Sanghavi, M., Mintun, M.A., Gado, M.H., 1999. Depression duration but not
against the delayed behavioral and cellular effects of acute stress on the amygdala. age predicts hippocampal volume loss in medically healthy women with recurrent
Biol. Psychiat. 72, 466–475. major depression. J. Neurosci. 19, 5034–5043.
Rasgon, N., Jarvik, G.P., Jarvik, L., 2001. Affective disorders and Alzheimer disease: a Shonkoff, J.P., Boyce, W.T., McEwen, B.S., 2009. Neuroscience, molecular biology, and
missing-link hypothesis. Am. J. Geriatric Psychiat.: Off. J. Am. Assoc. Geriatric the childhood roots of health disparities. JAMA 301, 2252–2259.
Psychiat. 9, 444–445. Shors, T.J., 2016. A trip down memory lane about sex differences in the brain. Philos.
Rasgon, N., Jarvik, L., 2004. Insulin resistance, affective disorders, and Alzheimer's dis- Trans. R. Soc. Lond. Ser. B, Biol. Sci. 371.
ease: review and hypothesis. J. Gerontol. 59A, 178–183. Shors, T.J., Chua, C., Falduto, J., 2001. Sex differences and opposite effects of stress on
Rasgon, N.L., Geist, C.L., Kenna, H.A., Wroolie, T.E., Williams, K.E., Silverman, D.H., dendritic spine density in the male versus female hippocampus. J. Neurosci. 21,
2014. Prospective randomized trial to assess effects of continuing hormone therapy 6292–6297.
on cerebral function in postmenopausal women at risk for dementia. PLoS One 9, Sierra, A., Gottfried-Blackmore, A.C., McEwen, B.S., Bulloch, K., 2007. Microglia derived
e89095. from aging mice exhibit an altered inflammatory profile. Glia 55, 412–424.
Rasgon, N.L., Kenna, H.A., Wroolie, T.E., Kelley, R., Silverman, D., Brooks, J., Williams, Skrzypiec, A.E., Shah, R.S., Schiavon, E., Baker, E., Skene, N., Pawlak, R., Mucha, M.,
K.E., Powers, B.N., Hallmayer, J., Reiss, A., 2011. Insulin resistance and hippocampal 2013. Stress-induced lipocalin-2 controls dendritic spine formation and neuronal
volume in women at risk for Alzheimer's disease. Neurobiol. Aging 32, 1942–1948. activity in the amygdala. PLoS One 8, e61046.
Rasgon, N.L., McEwen, B.S., 2016. Insulin resistance-a missing link no more. Mol. Spencer, J.L., Waters, E.M., Romeo, R.D., Wood, G.E., Milner, T.A., McEwen, B.S., 2008.
Psychiat. 21, 1648–1652. Uncovering the mechanisms of estrogen effects on hippocampal function. Front.
Rayon-Estrada, V., Harjanto, D., Hamilton, C.E., Berchiche, Y.A., Gantman, E.C., Sakmar, Neuroendocrinol. 29, 219–237.
T.P., Bulloch, K., Gagnidze, K., Harroch, S., McEwen, B.S., Papavasiliou, F.N., 2017. Spritzer, M.D., Galea, L.A., 2007. Testosterone and dihydrotestosterone, but not estradiol,
Epitranscriptomic profiling across cell types reveals associations between APOBEC1- enhance survival of new hippocampal neurons in adult male rats. Dev. Neurobiol. 67,
mediated RNA editing, gene expression outcomes, and cellular function. Proc. Natl. 1321–1333.
Acad. Sci. USA. www.pnas.org/cgi/doi/10.107/pnas.1714227114. Starkman, M.N., Gebarski, S.S., Berent, S., Schteingart, D.E., 1992. Hippocampal forma-
Reagan, L.P., Magarinos, A.M., Yee, D.K., Swzeda, L.I., Van Bueren, A., McCall, A.L., tion volume, memory dysfunction, and cortisol levels in partiens with Cushing's
McEwen, B.S., 2000. Oxidative stress and HNE conjugation of GLUT3 are increased in syndrome. Biol. Psychiat. 32, 756–765.
the hippocampus of diabetic rats subjected to stress. Brain Res. 862, 292–300. Starkman, M.N., Schteingart, D.E., 1981. Neuropsychiatric manifestations of patients with
Rettberg, J.R., Yao, J., Brinton, R.D., 2014. Estrogen: a master regulator of bioenergetic Cushing's syndrome. Arch. Interm. Med. 141, 215–219.
systems in the brain and body. Front. Neuroendocrinol. 35, 8–30. Stavreva, D.A., Wiench, M., John, S., Conway-Campbell, B.L., McKenna, M.A., Pooley,
Reul, J.M., DeKloet, E.R., 1985. Two receptor systems for corticosterone in rat brain: J.R., Johnson, T.A., Voss, T.C., Lightman, S.L., Hager, G.L., 2009. Ultradian hormone
microdistribution and differential occupation. Endocrinology 117, 2505–2511. stimulation induces glucocorticoid receptor-mediated pulses of gene transcription.
Romeo, R.D., 2017. The impact of stress on the structure of the adolescent brain: im- Nat. Cell Biol. 11, 1093–1102.
plications for adolescent mental health. Brain Res. 1654, 185–191. Sterling, P., Eyer, J., 1988. Allostasis: a new paradigm to explain arousal pathology. In:
Romeo, R.D., Bellani, R., Karatsoreos, I.N., Chhua, N., Vernov, M., Conrad, C.D., McEwen, Fisher, S., Reason, J. (Eds.), Handbook of Life Stress, Cognition and Health. John
B.S., 2006. Stress history and pubertal development interact to shape hypothalamic- Wiley & Sons, New York, pp. 629–649.
pituitary-adrenal axis plasticity. Endocrinology 147, 1664–1674. Stumpf, W., 1971. Autoradiographic techniques and the localization of estrogen, an-
Romeo, R.D., Staub, D., Jasnow, A.M., Karatsoreos, I.N., Thornton, J.E., McEwen, B.S., drogen and glucocorticoid in the pituitary and brain. Am. Zool. 11, 725–739.
2005. Dihydrotestosterone increases hippocampal N-methyl-D-aspartate binding but Suzuki, A., Stern, S.A., Bozdagi, O., Huntley, G.W., Walker, R.H., Magistretti, P.J.,
does not affect choline acetyltransferase cell number in the forebrain or choline Alberini, C.M., 2011. Astrocyte-neuron lactate transport is required for long-term
transporter levels in the CA1 region of adult male rats. Endocrinology 146, memory formation. Cell 144, 810–823.
2091–2097. Szyf, M., McGowan, P., Meaney, M.J., 2008. The social environment and the epigenome.
Rubin, T.G., Gray, J.D., McEwen, B.S., 2014. Experience and the ever-changing brain: Environ. Mol. Mutagen. 49, 46–60.
what the transcriptome can reveal. BioEssays: News Rev. Mol. Cell. Dev. Biol. 36, Tabori, N.E., Stewart, L.S., Znamensky, V., Romeo, R.D., Alves, S.E., McEwen, B.S.,
1072–1081. Milner, T.A., 2005. Ultrastructural evidence that androgen receptors are located at
Rudick, C.N., Gibbs, R.B., Woolley, C.S., 2003. A role for the basal forebrain cholinergic extranuclear sites in the rat hippocampal formation. Neuroscience 130, 151–163.
system in estrogen-induced disinhibition of hippocampal pyramidal cells. J. Neurosci. Tang, A.C., Reeb-Sutherland, B.C., Romeo, R.D., McEwen, B.S., 2014. On the causes of

29
B.S. McEwen Frontiers in Neuroendocrinology 49 (2018) 8–30

early life experience effects: evaluating the role of mom. Front. Neuroendocrinol. 35, distinctive changes of gene expression in the CNS in response to different moderate
245–251. restraint stress. J. Neurochem. 113, 1436–1446.
Tang, A.C., Reeb-Sutherland, B.C., Yang, Z., Romeo, R.D., McEwen, B.S., 2011. Neonatal Watanabe, Y., Gould, E., McEwen, B.S., 1992. Stress induces atrophy of apical dendrites of
novelty-induced persistent enhancement in offspring spatial memory and the mod- hippocampus CA3 pyramidal neurons. Brain Res. 588, 341–344.
ulatory role of maternal self-stress regulation. J. Neurosci. 31, 5348–5352. Waters, E.M., Thompson, L.I., Patel, P., Gonzales, A.D., Ye, H.Z., Filardo, E.J., Clegg, D.J.,
Toft, D., Gorski, J., 1966. A receptor molecule for estrogens: isolation from the rat uterus Gorecka, J., Akama, K.T., McEwen, B.S., Milner, T.A., 2015. G-protein-coupled es-
and preliminary characterization. Proc. Natl. Acad. Sci. 55, 1574–1581. trogen receptor 1 is anatomically positioned to modulate synaptic plasticity in the
Tomasdottir, M.O., Sigurdsson, J.A., Petursson, H., Kirkengen, A.L., Krokstad, S., mouse hippocampus. J. Neurosci. 35, 2384–2397.
McEwen, B., Hetlevik, I., Getz, L., 2015. Self reported childhood difficulties, adult Wehling, M., 1997. Specific, nongenomic actions of steroid hormones. Annu. Rev.
multimorbidity and allostatic load. A cross-sectional analysis of the Norwegian HUNT Physiol. 59, 365–393.
study. PLoS One 10, e0130591. Weiland, N.G., 1992. Estradiol selectively regulates agonist binding sites on the N-methyl-
Towart, L.A., Alves, S.E., Znamensky, V., Hayashi, S., McEwen, B.S., Milner, T.A., 2003. D-aspartate receptor complex in the CA1 region of the hippocampus. Endocrinology
Subcellular relationships between cholinergic terminals and estrogen receptor-a in 131, 662–668.
the dorsal hippocampus. J. Comp. Neurol. 463, 390–401. Wiley, J.F., Gruenewald, T.L., Karlamangla, A.S., Seeman, T.E., 2016. Modeling multi-
Treccani, G., Musazzi, L., Perego, C., Milanese, M., Nava, N., Bonifacino, T., Lamanna, J., system physiological dysregulation. Psychosom. Med. 78, 290–301.
Malgaroli, A., Drago, F., Racagni, G., Nyengaard, J.R., Wegener, G., Bonanno, G., Wilkinson, R.G., Pickett, K., 2010. The Spirit Level: Why Greater Equality Makes Societies
Popoli, M., 2014. Stress and corticosterone increase the readily releasable pool of Stronger. Bloomsbury Press, New York.
glutamate vesicles in synaptic terminals of prefrontal and frontal cortex. Mol. Winocur, G., Greenwood, C.E., 1999. The effects of high fat diets and environment in-
Psychiatr. 19, 433–443. fluences on cognitive performance in rats. Behav. Brain Res. 101, 153–161.
Trejo, J.L., Carro, E., Torres-Aleman, I., 2001. Circulating insulin-like growth factor I Winocur, G., Piroli, G.G., Grillo, C., Reagan, L.P., Greenwood, c.E., Reznikov, L.R.,
mediates exercise-induced increases in the number of new neurons in the adult McEwen, B.S., 2005. Memory impairment in obese Zucker rats: an investigation of
hippocampus. J. Neurosci. 21, 1628–1634. cognitive function in an animal model of insulin resistance and obesity. Behav.
Uemura, K., Iguchi, A., Yatomi, A., Miura, H., Honmura, A., Yanase, M., Sakamoto, N., Neurosci. 119, 1389–1395.
1989. Involvement of the hippocampus in central nervous system-mediated glucor- Wood, G.E., Shors, T.J., 1998. Stress facilitates classical conditioning in males, but im-
egulation in rats. Endocrinology 124, 2449–2455. pairs classical conditioning in females through activational effects of ovarian hor-
Vale, W., Spiess, J., Rivier, C., Rivier, J., 1981. Characterization of a 41-residue ovine mones. Proc. Natl. Acad. Sci. USA 95, 4066–4071.
hypothalamic peptide that stimulates secretion of corticotropin and beta-endorphin. Woolley, C., McEwen, B.S., 1992. Estradiol mediates fluctuation in hippocampal synapse
Science 213, 1394–1397. density during the estrous cycle in the adult rat. J. Neurosci. 12, 2549–2554.
Valentino, R.J., Bangasser, D., Van Bockstaele, E.J., 2013. Sex-biased stress signaling: the Woolley, C., McEwen, B.S., 1993. Roles of estradiol and progesterone in regulation of
corticotropin-releasing factor receptor as a model. Mol. Pharmacol. 83, 737–745. hippocampal dendritic spine density during the estrous cycle in the rat. J. Comp.
van der Kooij, M.A., Fantin, M., Kraev, I., Korshunova, I., Grosse, J., Zanoletti, O., Neurol. 336, 293–306.
Guirado, R., Garcia-Mompo, C., Nacher, J., Stewart, M.G., Berezin, V., Sandi, C., Woolley, C., McEwen, B.S., 1994. Estradiol regulates hippocampal dendritic spine density
2014a. Impaired hippocampal neuroligin-2 function by chronic stress or synthetic via an N-methyl-D-aspartate receptor dependent mechanism. J. Neurosci. 14,
peptide treatment is linked to social deficits and increased aggression. 7680–7687.
Neuropsychopharmacology 39, 1148–1158. Woolley, C.S., 1999. Effects of estrogen in the CNS. Curr. Opin. Neurobiol. 9, 349–354.
van der Kooij, M.A., Fantin, M., Rejmak, E., Grosse, J., Zanoletti, O., Fournier, C., Woolley, C.S., Weiland, N.G., McEwen, B.S., Schwartzkroin, P.A., 1997. Estradiol in-
Ganguly, K., Kalita, K., Kaczmarek, L., Sandi, C., 2014b. Role for MMP-9 in stress- creases the sensitivity of hippocampal CA1 pyramidal cells to NMDA receptor-
induced downregulation of nectin-3 in hippocampal CA1 and associated behavioural mediated synaptic input: correlation with dendritic spine density. J. Neurosci. 17,
alterations. Nat. Commun. 5, 4995. 1848–1859.
van der Werff, S.J., Pannekoek, J.N., Andela, C.D., Meijer, O.C., van Buchem, M.A., Workman, J.L., Gobinath, A.R., Kitay, N.F., Chow, C., Brummelte, S., Galea, L.A.M., 2016.
Rombouts, S.A., van der Mast, R.C., Biermasz, N.R., Pereira, A.M., van der Wee, N.J., Parity modifies the effects of fluoxetine and corticosterone on behavior, stress re-
2015. Resting-state functional connectivity in patients with long-term remission of activity, and hippocampal neurogenesis. Neuropharmacology 105, 443–453.
Cushing's disease. Neuropsychopharmacology 40, 1888–1898. Wright, R.L., Lightner, E.N., Harman, J.S., Meijer, O.C., Conrad, C.D., 2006. Attenuating
van Praag, H., Kempermann, G., Gage, F.H., 1999. Running increases cell proliferation corticosterone levels on the day of memory assessment prevents chronic stress-in-
and neurogenesis in the adult mouse dentate gyrus. Nat. Neurosci. 2, 266–270. duced impairments in spatial memory. Eur. J. Neurosci. 24, 595–605.
van Praag, H., Shubert, T., Zhao, C., Gage, F.H., 2005. Exercise enhances learning and Yau, P.L., Castro, M.G., Tagani, A., Tsui, W.H., Convit, A., 2012. Obesity and metabolic
hippocampal neurogenesis in aged mice. J. Neurosci. 25, 8680–8685. syndrome and functional and structural brain impairments in adolescence. Pediatrics
Vetencourt, J.F.M., Sale, A., Viegi, A., Baroncelli, L., De Pasquale, R., O'Leary, O.F., 130, e856–e864.
Castren, E., Maffei, L., 2008. The antidepressant fluoxetine restores plasticity in the Yu, Y., Kastin, A.J., Pan, W., 2006. Reciprocal interactions of insulin and insulin-like
adult visual cortex. Science 320, 385–388. growth factor I in receptor-mediated transport across the blood-brain barrier.
Vyas, A., Mitra, R., Rao, B.S.S., Chattarji, S., 2002. Chronic stress induces contrasting Endocrinology 147, 2611–2615.
patterns of dendritic remodeling in hippocampal and amygdaloid neurons. J. Yuen, G.S., McEwen, B.S., Akama, K.T., 2011. LIM kinase mediates estrogen action on the
Neurosci. 22, 6810–6818. actin depolymerization factor Cofilin. Brain Res. 1379, 44–52.
Vythilingam, M., Heim, C., Newport, J., Miller, A.H., Anderson, E., Bronen, R., Brummer, Zigman, J.M., Jones, J.E., Lee, C.E., Saper, C.B., Elmquist, J.K., 2006. Expression of
M., Staib, L., Vermetten, E., Charney, D.S., Nemeroff, C.B., Bremner, J.D., 2002. ghrelin receptor mRNA in the rat and the mouse brain. J. Comp. Neurol. 494,
Childhood trauma associated with smaller hippocampal volume in women with 528–548.
major depression. Am. J. Psychiat. 159, 2072–2080. Zigmond, R., McEwen, B.S., 1970. Selective retention of oestradiol by cell nuclei in
Waddington, C.H., 1942. The epigenotype. Endeavoour 1, 18–20. specific brain regions of the ovariectomized rats. J. Neurochem. 17, 889–899.
Wang, J., Rao, H., Wetmore, G.S., Furlan, P.M., Korczykowski, M., Dinges, D.F., Detre, Znamensky, V., Akama, K.T., McEwen, B.S., Milner, T.A., 2003. Estrogen levels regulate
J.A., 2005. Perfusion functional MRI reveals cerebral blood flow pattern under psy- the subcellular distribution of phosphorylated Akt in hippocampal Ca1 dendrites. J.
chological stress. Proc. Natl. Acad. Sci. USA 102, 17804–17809. Neurosci. 23, 2340–2347.
Wang, J.M., Singh, C., Liu, L., Irwin, R.W., Chen, S., Chung, E.J., Thompson, R.F., Brinton, Zohar, J., Yahalom, H., Kozlovsky, N., Cwikel-Hamzany, S., Matar, M.A., Kaplan, Z.,
R.D., 2010a. Allopregnanolone reverses neurogenic and cognitive deficits in mouse Yehuda, R., Cohen, H., 2011. High dose hydrocortisone immediately after trauma
model of Alzheimer's disease. Proc. Natl. Acad. Sci. USA 107, 6498–6503. may alter the trajectory of PTSD: interplay between clinical and animal studies. Eur.
Wang, K., Xiang, X.H., He, F., Lin, L.B., Zhang, R., Ping, X.J., Han, J.S., Guo, N., Zhang, Neuropsychopharmacol. 21, 796–809.
Q.H., Cui, C.L., Zhao, G.P., 2010b. Transcriptome profiling analysis reveals region-

30

You might also like