Regarding chapter, we could

advertisement
1
INTRODUCTION:
I. Stress and depression
Depressive disorders are widely regarded as stress-related conditions. While genetic
vulnerability is critical to the development of depression, in the absence of environmental stressors,
the incidence of depressive disorders is very low (Kendler et al. 1995a); and in approximately 75%
of cases of depression there is a precipitating life event (Brown & Harris 1978; Frank et al., 1994).
Living organisms survive by maintaining a complex dynamic equilibrium or homeostasis that is
constantly challenged by intrinsic or extrinsic stressors. These stressors set in motion responses
aimed at preserving homeostasis, including activation of a wide variety of neurotransmitters and
neuromodulators. The hypothalamic pituitary adrenal (HPA) axis is the body's main stress
hormonal system. Corticotropin releasing hormone (CRH), is the principal central effectors of the
stress response (Chrousos and Gold 1992). CRH triggers the release of adrenocorticotropic
hormone (ACTH) from the anterior pituitary corticotrope which, in turn, triggers the release of
adrenal glucocorticoids. The stress response is terminated by glucocorticoid feedback at brain and
pituitary sites.
Depression has been conceptualized as maladaptive, exaggerated responses to stress.
Abnormalities of the hypothalamic-pituitary-adrenal (HPA) axis, as manifested by
hypercortisolemia and disruption of the circadian rhythm of cortisol secretion, are well established
phenomena in depression (Carroll et al. 1976; Sachar et al. 1973). The assumption that the
pathophysiology of depression involves exaggerated responses to stress is supported by evidence
that CRH, is activated in these patients (Altemus et al. 1992; Charney et al. 1987; Butler and
Nemeroff, 1990; Southwick et al, 1993, Bhagwagar et al, 2003; Young et al, 2001a; Raadsheer et
al, 1994)
2
Sex differences in depression:
As well as their association with stress, another striking feature of mood disorders is the
increased prevalence of these conditions in women. Several lines of evidence indicate that sex
steroid hormones play a role in the increased vulnerability of women to anxiety disorders and
depression. Women have an increased incidence of unipolar depression (Weissman and Olfson
1995) which arises at puberty. The immediate postpartum period, in particular, is a time of greatly
increased risk for new onset or recurrence of mood disorders (Altshuler et al. 1998; Dean et al.
1989). In some women, recurrent depressive symptoms occur only during the premenstrual period
and in others, chronic depression is often exacerbated premenstrually (Rubinow and Roy-Byrne
1984). Recent reports indicate that estrogen may be an effective treatment for postpartum (Gregoire
et al. 1996) and perimenopausal depression (Zweifel and O'Brien 1997).
Hypothalamic-Pituitary-Adrenal (HPA) Axis Abnormalities in Depression: Overactivity of the
HPA axis, as manifested by an increase in cortisol secretion, is a well-established phenomenon in
depression (Sachar et al., 1973; Carroll, Curtis et al., 1976). The original studies of Sachar and
colleagues ( Sachar et al., 1973) demonstrated increased cortisol secretory activity in depressed
patients as measured by mean plasma cortisol concentration, the number of cortisol secretory
episodes and the number of minutes of active secretion. Later studies have continued to validate
this hypercortisolemia in depression (Carroll, Curtis et al., 1976, Rubin et al., 1987, Halbreich U,
Asnis GM, Schindledecker R, Zurnoff B, Nathan RS 1985, Pfohl B, Sherman B, Schlecte J, Stone,
R 1985). As many as two-thirds of endogenously depressed patients fail to suppress cortisol, or
show an early escape of cortisol, following overnight administration of 1 mg of dexamethasone,
using a cortisol cut-off of 5 µg/dl to define “escape” (Carroll, Feinberg, 1981). While non-
3
suppression of cortisol to dexamethasone is strongly associated with endogenous depression, this
finding is less robust in outpatients with depression. Although both hypercortisolemia and
feedback abnormalities to dexamethasone are present in depressed patients, they do not necessarily
occur in the same individuals (Halbreich et al., 1985, Carroll, Feinberg et al.,1981). Abnormal
glucocorticoid fast feedback (Young, Haskett et al., 1991) and a blunted ACTH response to oCRF
have also been reported in depressed patients (Gold et al., 1986; Holsboer et al., 1984, Young,
Watson et al., 1990). The blunted response to oCRF appears to be dependent upon increased
baseline cortisol, since blockade of cortisol production with metyrapone normalizes the ACTH
response (von Bardeleben et al., 1988, Young, Akil et al., 1995). It was expected that the increased
cortisol would be accompanied by an increased level of ACTH in plasma, but this expectation has
been difficult to validate. Some studies (Pfohl et al., 1985, Linkowski et al., 1985, Young, Carlson
et al., 2001b) have been able to demonstrate small differences between normal controls and
depressed subjects in their mean 24 hr plasma ACTH levels. The demonstration of enhanced
sensitivity to ACTH 1-24 in depressed patients suggests that increased ACTH secretion is not
necessarily the cause of increased cortisol secretion (Amsterdam et al., 1983). However, other
studies using very low "threshold" doses of ACTH 1-24 have not been able to demonstrate an
increased sensitivity to ACTH in depressed patients (Krishnan KRR et al., 1990), which suggests
that increased cortisol secretion is secondary to increased ACTH secretion. Our 24H studies of
ACTH and cortisol secretion demonstrated that subjects with increased mean cortisol also
demonstrated increased mean ACTH, supporting a central origin of the HPA axis overactivity.
(Young EA et al, 2001b). Finally our studies with metyrapone in major depression support the
presence of increased CNS drive, at least in the evening (Young EA et al, 1994, Young EA et al,
1997). It appears likely that there is increased CRF/ACTH secretion, which is then probably
amplified at the adrenal level leading to increased cortisol. These changes in cortisol secretion are
4
commonly considered to be “state” changes and resolve when the depression resolves. However,
almost all studies examining the HPA axis in major depression in euthymic subjects have
examined patients on tricyclic antidepressants, which exert direct effects on the HPA axis. Two
recent studies of ours as well as a recent report by Bhagwagar and colleagues have found that
saliva cortisol is increased in subjects with lifetime major depression, in subjects who had no
current mood symptoms (Young et al, 2000; Young and Breslau, in press; Bhagwagar et al,
2003). The overall picture in depression if of increased production of activational elements of the
HPA axis together with a reduction in feedback inhibition.
Chronic antidepressant treatment inhibits stress responsive systems at multiple sites, through
reductions in CRH and tyrosine hydroxylase activity, enhanced glucocorticoid receptor activity
(Brady et al.1991; Barden, Reul and Holsboer, 1995), and down regulation of arousal producing ßadrenergic receptors (Heninger and Charney 1987). Chronic treatment with antidepressant agents
also reduces both the behavioral and endocrine responses to stress (Murua and Molina 1992; Reul
et al. 1993; Barden, Reul and Holsboer, 1995). Consequently, antidepressant actions may be
through stress systems as well as classical neurotransmitter systems that affect neurobiological
systems involved in the pathophysiology of depression.
Sex Differences In HPA Axis Regulation in Depression
Morning and Evening Cortisol Hypersecretion. We studied baseline cortisol secretion in the
morning in 16 depressed patients and 16 age-and sex-matched control patients and found
predictably increased cortisol secretion in the group as a whole (Young et al. 1991). However,
5
there were also clear sex differences: male patients and their matched controls demonstrated the
same plasma cortisol concentration, whereas female depressed patients demonstrated a
significantly higher mean plasma cortisol concentration (11.3 ± 0.9 µg/dl) than that of the
matched control group (8.1 ±0.95 µg/dl; significant by a two-tailed t-test, p=0.033). Removal of
glucocorticoid negative feedback by metyrapone demonstrated increased central drive in
depressed patients in the evening (Young et al. 1994). The response to metyrapone also showed
sex differences; in that only the female depressed patients manifested rebound -LPH/-end
secretion in comparison to their matched controls (ANOVA, F=8.8, df=1, p=.01); whereas the
males did not.
Dexamethasone Non-Suppression and Menopause.
We also examined the effect of loss of
gonadal steroids at menopause on HPA axis regulation in depressed women (Young et al. 1993).
We conducted studies using a protocol examining baseline and post-dexamethasone secretion of
-LPH/-end and cortisol over the course of the day (8 AM-4 PM). These were carried out on
51 depressed women; 36 of whom were premenopausal and 15 postmenopausal.
The
premenopausal women demonstrated a significantly lower incidence of pituitary (-LPH/-end)
non-suppression (44%; n=36) than the postmenopausal women (Non-Suppressor=81%; n=15).
To determine which of a number of potential variables were associated with -LPH/-end nonsuppression in women, a step-wise regression analysis was used.
Independent variables
included: age, menopausal status, baseline -LPH/-end and cortisol, severity of depression
(Hamilton Depression rating scores), and the number of previous episodes of depression. The
dependent variable was -LPH/-end non-suppression. We found that age had a significant effect
on pituitary non-suppression, but when age and menopausal status were compared, menopausal
6
status showed a stronger correlation and combined with cortisol gave a correlation coefficient of
0.817. This suggests that menopausal status, in conjunction with cortisol hypersecretion, is a
critical variable in the development of HPA dysregulation, as manifested by resistance to
dexamethasone, and accounts for 65% of the variance.
In summary, depressed women demonstrate greater HPA axis activation than depressed men.
Furthermore, this activation is linked to insensitivity to dexamethasone, and thus appears to
reflect the development of glucocorticoid receptor down-regulation following a period of
hypercortisolemia. Menopause is not associated with increases in plasma cortisol concentrations
in depressed women, but it is associated with an increase in dexamethasone resistance.
EFFECTS OF GONADOSTEROIDS ON THE HPA AXIS:
Animal studies
Studies in rodents support the existence of sex differences in several of the elements of the HPA
axis. Female rats appear to have a more robust HPA axis response to stress than do male rats, and
there is evidence that estrogen is at least partly responsible for this sexual dimorphism. For
example, compared with male rats, female rats have a faster onset of corticosterone secretion after
stress and a faster rate of rise of corticosterone (Jones et al. 1972). The increased corticosterone
response is accompanied by a greatly increased ACTH response to stress in female rodents (Young
1996). Furthermore, corticosteroid binding globulin (CBG) is positively regulated by estrogen and
thus higher in female rats; however, estrogen and progesterone have been demonstrated to affect
the HPA axis independent of the effects of CBG (Young, 1996). In addition, chronic estrogen
treatment of ovariectomized female rats enhances their corticosterone response to stress, and slows
their recovery from stress (Burgess and Handa 1992). Studies by Viau and Meaney (1991)
7
demonstrate a greater ACTH and corticosterone stress response in acute estradiol treated rats
compared with ovariectomized female rats, or with estradiol plus progesterone treated female rats,
after short-term (24h) but not long-term (48h) estradiol treatment. However, our studies (Young
and Altemus, 2001) found that two centrally active estradiol antagonists, tamoxifen and CI-628,
led to a greater stress response in intact female rats.
Also, estradiol replacement acted of
ovariectomized rats led to decreased stress responses. Similar inhibitory effects of estradiol on
stress response have been found in sheep and humans. (Komesaroff et al, 1998 and 1999). Studies
showing inhibitory effects of estradiol on the stress response have used lower doses of estradiol
than those showing activation of the axis.
Work by Keller-Wood and colleagues (Keller-Wood et al. 1988) in pregnant ewes and ewes
given progesterone infusions, demonstrate that progesterone can diminish the effectiveness of
cortisol feedback on stress responsiveness in vivo. In addition, progesterone demonstrates antiglucocorticoid effects on feedback in intact rats in vivo and in vitro (Svec 1988; Duncan and
Duncan 1979). Progesterone binds to the glucocorticoid receptor; while it does so with a faster
binding time than glucocorticoid itself, progesterone binding is to a different site on the receptor
than glucocorticoid binding (Svec 1988). Progesterone can also increase the rate of dissociation of
glucocorticoids from the glucocorticoid receptor (Rousseau et al. 1972). In addition, binding
studies with expressed human mineralocorticoid receptor (MR) have demonstrated an affinity of
progesterone for MR receptor in a range similar to that of dexamethasone (Arriza et al. 1987).
Furthermore, there was an increase in MR binding following progesterone treatment of female rats
(Carey et al. 1995). Finally, female rats have a greater number of glucocorticoid receptors in the
hippocampus than male rats (Turner and Weaver 1985), and progesterone modulates
immunoreactive glucocorticoid receptor distribution in the hippocampus of rats (Ahima et al.
8
1992). It should be noted that binding studies do not distinguish agonist effects from antagonist
effects, therefore even increases in number could result from antagonist effects at GR.
Human Studies
Until recently, the lack of a reliable stress test limited studies on sex differences in stress
response in humans. In the Trier Social Stress Test, subjects undergo a mock job interview in front
of a panel of interviewers who are instructed not to provide any verbal or non-verbal feedback; it is
a reliable and robust stressor in normal subjects (Kirschbaum et al. 1995). It has now been shown
that oral contraceptives decrease the free cortisol response to a social stressor in women
(Kirschbaum et al. 1995), while the treatment of normal men with estradiol for 48 hours results in
an enhanced ACTH and cortisol response to a social stressor (Kirschbaum et al. 1996). These data
of estrogen treatment in men are consistent with results of studies in rats (Burgess and Handa 1992;
Viau and Meaney 1991). However, results from studies of oral contraceptives are harder to
interpret because they are synthetic steroids given at constant doses for a prolonged period of time
and may differ from endogenous steroids in their effects. Direct comparison of the ACTH response
to this social stressor in men and women has demonstrated a smaller ACTH response in women
but a similar cortisol response in both sexes (Kirschbaum et al, 1999; Young , Abelson and
Cameron, 2004). These data are in agreement with studies demonstrating that estradiol decreases
stress responsiveness in women (Komesaroff et al, 1999)
With respect to the influence of changes in ovarian hormones across the menstrual cycle in
women, recent studies by Altemus and colleagues (1997) have found increased resistance to
dexamethasone suppression during the luteal phase of the menstrual cycle, compared to during the
follicular phase, a change that may again be related to either increased estradiol or progesterone
during the luteal phase. In addition, ACTH, vasopressin and cortisol responses to stress are
9
enhanced in the luteal phase compared to the follicular phase of the menstrual cycle (Altemus et al.
1997; Galliven, et al 1997) suggesting that decreases in glucocorticoid receptors may explain the
decreased response to dexamethasone. In a design which allowed investigators to distinguish the
effects of progesterone from those of estrogen, Roca and coworkers (1998a;1998b. 2003) studied
control women first treated with Lupron, a gonadotrophin-releasing hormone (GnRH) agonist,
which causes suppression of both estrogen and progesterone secretion, and then given sequential
replacement of the two hormones. They examined the response to exercise stress as well as to
dexamethasone feedback, and found that the exercise stress response was increased. Response to
dexamethasone feedback was decreased during the progesterone "add back" phase but not during
the estrogen "add back" phase. Again, these data suggest that progesterone acts as a glucocorticoid
antagonist. Thus, data from human studies suggest that ovarian steroids may modulate stress
responses in opposite directions so that estradiol decreases stress response while progesterone
decreases sensitivity to negative feedback.
II. Genetic Vulnerability to Depression.
While stress is clearly a precipitating factor in the onset of depression, not everyone who
experiences stress develops a depressive episode suggesting an underlying vulnerability
(Kendler, K. S., 1998). It has long been known that depression clusters in families and extensive
evidence has now accumulated that depression is a complex disorder resulting from the
interaction of genetic and environmental factors (Jones, et al, 2002). Twin studies of unipolar
depression have shown variable degrees of heritability depending on the type of sample and the
definitions of depression used. In a recent meta-analysis of twin data, Sullivan (Sullivan et al,
2000) estimated the heritability of depression to be 37% with a significant contribution of
environmental events specific to individuals but only a small contribution from shared familial
10
environmental effects. However, in studies of subjects recruited from psychiatric treatment
centers, rather than community samples, the heritability was much higher (60-70%) (Kendler, et
al, 1995).(McGuffin et al, 1996) indicating a possibly greater heritability of more severe forms of
depression.
Twin studies have examined the genetic risk factors for major depression separately in men and
women with conflicting findings. Two studies have suggested that there is equal heritability for
depression in the two sexes (McGuffin et al, 1996; Kendler et al, 1995). However, in studies
where a broader definition of depression was used, genetic factors accounted for a significantly
greater proportion of the liability to develop depression in women compared with men (Bierut et
al, 1999; Lyons et al, 1998). Although the genes that influence risk for depression in the two
sexes are correlated, they are probably not entirely the same and it is possible that genes
conferring risk for depression differ in men and women (Kendler et al, 2001). Kendler goes on to
suggest that environmental factors may bring out distinct genetic variation in the two sexes or,
alternatively, that biological factors, such as hormonal variations during the menstrual cycle and
pregnancy, could elicit discrete genetic variation in women and men. Genetic influences may also
operate through temperamental dimensions such as neuroticism and through the effect of
different developmental influences such as the onset of puberty marking an increased
vulnerability to depression resulting from negative life events in girls (Rutter, M., 2002).
III. Neuroanatomical Basis of Depression:
Post-mortem studies of depressed patients have often involved patients who die by suicide, thus
conflating depression with suicide, which only occurs in a small minority of patients and may
involve comorbid disorders, particularly alcohol and substance abuse. Furthermore, because of
the nature of post-mortem studies, a priori hypotheses are usually tested rather than new
11
hypotheses about functional systems and their relationship to depression. Post-mortem data have
confirmed low serotonin in the CSF of suicide victims, as well as down-regulation of 5HT1a
terminal receptors in cortical regions (Asberg; Mann and Arrango review). They have also
demonstrated increased CRH and vasopressin in the hypothalamus and down-regulation of CRH
receptor binding in the frontal cortex (Raadsheer et al, 1994) . Structural MRI studies have
found smaller hippocampi, often taken to indicate hypercortisolism associated with depression
(Sheline et al, 1996, 2003). Functional imaging gives us another opportunity to understand the
networks involved in depression. However, abnormalities found on functional imaging may not
necessarily identify the "anatomical lesion" of depression but may also demonstrate
compensatory coping mechanisms.
The majority of functional imaging studies have identified abnormalities in the prefrontal cortex,
particularly dorsolateral and ventrolateral areas as the primary finding in depression (Mayberg,
1994: Mayberg et al, 1994; Ketter et al, 1996; Drevets, et al, 1992; Buchsbaum et al, 1986,
Baxter et al, 1989; George et al, 1994) Studies of emotion in normal subjects find these same key
areas activated in response to sad mood (George et al, 1995) Other areas identified in some
studies include limbic areas such as amygdala, temporal cortex and insula but there is less
consensus for these findings. Mayberg has outlined a functional circuit in depression involving
brainstem nuclei, striatum, and thalamus (Mayberg et al, 2000). Treatment data suggest that
pharmacological therapies target the brainstem, limbic and subcortical areas in addition to frontal
cortical areas, while psychotherapies target the frontal cortical abnormalities (Mayberg, et al,
2000; Mayberg et al 2002).
The issue of sex differences have not yet been addressed in either post-mortem or in vivo imaging
studies.
12
IV. Serotonin and Depression
A fundamental hypothesis of the etiology of depressive disorders is that these disorders may be due
to a relative deficiency of serotonin. Major depression is accompanied by downregulation of
5HT1A receptors and mRNA, in brain and lymphocytes, as seen in post-mortem studies (López et
al, 1997; Arango et al, 2001) or in vivo imaging (Drevets et al, 1999; Drevets et al, 2000) as well
as increased 5HT2a binding (Arango et al, 1990; Hrdina et al, 1993; Yates et al, 1990).
Furthermore, these two serotonin receptors are targets for antidepressant action, particularly
tricyclic antidepressants. Rodent studies have shown that chronic antidepressant administration
results in functional "upregulation" of the postsynaptic 5-HT1a receptor in the hippocampus (Blier
et al, 1994). Some studies have also reported a modest increase in 5-HT1a receptor number in
hippocampus following antidepressant administration to rodents (Welner et al, 1989; Klimek et al,
1994). Studies have reported decreases in 5-HT2a binding in the prefrontal cortex after chronic
antidepressant administration (Peroutka, Snyder 1980). These findings have led some investigators
to propose that postsynaptic 5-HT1a and 5-HT2a receptors have functionally opposing effects
(Schreiber, De Vry, 1993), that a disturbed balance of these receptors may be contributing to the
pathophysiology of depression (Berendsen, 1995), and that restoration of this balance is necessary
for effective antidepressant action (Borsini, 1994).
Both stress and glucocorticoids modulate serotonin transmission. Acute stress levels of
glucorticoids increase serotonin turnover and increase the responsiveness of hippocampal neurons
to 5HT1A receptor stimulation (Meijer and de Kloet, 1998; McEwen, 1995). When elevated levels
of glucocorticoids persist, such as following chronic social stress, downregulation of hippocampal
13
5HT1A receptors occur while 5HT2 receptors in the cerebral cortex are upregulated (McEwen,
1995).
In addition, 5HT2C receptors are increased following corticosterone adrenectomy, and
normalize following corticosterone replacement (Meijer and de Kloet, 1998). Animal studies
demonstrate that chronic treatment with high doses of glucocorticoids lead to decreased serotonin
receptor mediated responses, similar to the picture observed in depressed patients, although the
exact mechanism of this hypofunctional serotonin state is unclear (Meijer and de Kloet, 1998).
This hypofunctional serotonin state may have further consequences for glucocorticoid secretion,
since serotonin appears to be an important regulator of glucocorticoid feedback. Antidepressants
which increase serotonin cause increases in glucocorticoid receptor number and can reverse the
increased glucocorticoid secretion seen in depressed humans and in transgenic mice who have been
genetically altered to demonstrate reduced glucocorticoid receptors and increased glucocorticoid
secretion (partial GR knockout) (Barden et al, 1995). Lesions of the serotonergic input to the
hippocampus, an important site in inhibiting glucocorticoid secretion, does produce decreased
glucocorticoid receptor expression and increased glucocorticoid secretion (Seckl and Fink, 1991).
Sex differences in serotonin systems:
Gonadal steroids appear to modulate mood, at least in part, through effects on serotonergic
systems. Overall, the literature suggests that estrogen enhances the efficiency of serotonergic
neurotransmission. Basic science studies indicate that there are clear sex differences in brain
serotonin systems some of which may depend upon estrogen and others upon testosterone. The
serotonin content and uptake in multiple areas of forebrain, hypothalamus and limbic system are
higher in females then males (Haleem et al, 1990, Borisova et al, 1996, Carlsson & Carlsson,
1988). 5HT1C and 5HT2 receptor binding in the dentate gyrus or CA4 regions of the
hippocampus is similar in males and females in rats; however, the 5HT1A receptor binding in CA1
14
region of hippocampus is higher in female rats and ovariectomy had no effect on this sex
difference (Mendelson & McEwen 1991). Stress has been shown to cause greater increases in
serotonin in female rats in multiple areas of brain (Heinsbroek et al, 1990). More recent studies
have found that estradiol increased serotonin transporter binding in female rat brains (McQueen et
al, 1997), as well as stimulating an increase in 5HT2A binding sites in limbic cortex (Fink et al
1996). Limited evidence suggests that like estrogen, progesterone may upregulate 5HT2 receptors
(Biegon et al. 1983), and increase serotonin content (Pecins-Thompson et al., 1996). However, no
consistent effects of progesterone administration on serotonin function have been identified. There
also is evidence that testosterone has opposing effects to estrogen on serotonergic activity.
Reductions in androgenic steroid have been associated with enhancement of central serotonergic
activity (Bonson et al. 1994; Fishette et al. 1984; Matsuda et al. 1991). In addition, administration
of testosterone has been associated with reductions in central serotonergic activity (MartinezConde et al. 1985; Mendelson and McEwen 1990).
Studies of sex differences in serotonin systems in humans are more limited.
Sex
differences in the prolactin and cortisol responses to serotonin agonists have been reported, with
women showing greater response to the serotonergic challenges (Monteleone et al, 1997; Ryan et
al, 1992, Lerer et al, 1996, Gelfin et al, 1995). However, estrogen regulates prolactin synthesis as
well as cortisol secretion so greater responses in women do not necessarily indicate serotonin
receptor differences. One study examining 5HT2 receptors on platelets in children, found a
suggestion of increased binding in teenage girls after the age of 14, but the study was clearly
limited by a small sample size of post pubertal adolescents (Biegon and Greuner, 1992).
Incubation of human platelets with sex steroids had no direct effects on serotonin uptake
(Ehrenkranz, 1976). Depressed women have a higher density of 5HT2 receptors on platelets than
male depressed patients (Hrdina et al, 1995). No sex differences have been found in serotonin
15
metabolites in cerebrospinal fluid of normal subjects (Leckman, 1994; Yoshino, 1982). One
postmortem study examining serotonin binding in human brain found no sex differences
(Marcusson et al, 1984); while another reported increased 5HT2 binding in frontal cortex in
women (Arato et al, 1991). One PET imaging study found a sex differences in 5HT2A in brain
with men showing more 5HT receptors than women. (Biver et al, 1996). It should be noted that
many of the human studies were conducted before an understanding of multiple serotonin receptors
existed, so many of the compounds used to detect serotonin receptors were non-specific. It is
likely that there are sex differences in serotonin systems in humans and the differences may be
larger in depressed women compared to depressed men than the differences seen in normal
subjects. Of note, the two illnesses shown to have a specific response to serotonergic
antidepressants, premenstrual syndrome (Eriksson et al. 1995) and obsessive-compulsive disorder
(Greist et al. 1995) appear to be particularly sensitive to changes in gonadal steroids.
CONCLUSIONS
The fact that women have much greater and repetitive fluxes in reproductive hormones
over the lifespan may enhance the potential for dysregulation of a wide variety of brain
neurochemical systems. In addition, as noted above, organizational differences between male and
female brains result fromexposure to high levels of gonadal steroids during the pre- and peri-natal
periods. The interactions of these organizational effects in females with cyclical gonadal steroid
hormone changes following puberty, then followed by menopause when there isloss of these same
steroids, suggest that stress responsiveness and susceptibility to stress related disorders could vary
16
substantially over the lifetime of women. There is certainly evidence that women's increased
vulnerability to depression arises at puberty, when gonadal steroids could further enhance HPA
axis responsiveness (Kessler et al. 1993). Additionally, the evidence linking stress and
glucocorticoids to hippocampal damage and subsequent memory problems (Issa et al. 1990), and
the important role that gonadal steroids may play in protection from these effects in premenopausal
women, imply that further research is needed into the interaction of stress, menopause and memory
impairment.
References
Ahima, R.S., Lawson, A.N.L., Osei, S.Y.S., & Harlan, R.E. (1992). Sexual dimorphism in regulation of
type II corticosteroid receptor immunoreactivity in the rat hippocampus. Endocrinology, 131:14091416.
Altemus, M., Pigott, T., Kalogeras, K.T., et al. (1992). Abnormalities in the regulation of vasopressin
and corticotropin releasing factor secretion in obsessive-compulsive disorder. Archives of General
Psychiatry, 49: 9-20.
Altemus, M., Redwine, L., Yung-Mei, L., Yoshikawa, T., Yehuda, R., Detera-Wadleigh, S. &
Murphy, D. (1997). Reduced sensitivity to glucocorticoid feedback and reduced glucocorticoid
receptor mRna expression in the luteal phase of the menstrual cycle. Neurosychopharmacology,
17(2):100-109.
17
Altshuler, L.L., Hendrick, V., Cohen, L.S. (1998). Course of mood and anxiety disorders during
pregnancy and the postpartum period. Journal of Clinical Psychiatry, 59 Suppl 2: 29-33.
Amsterdam JC, Winokur A, Abelman E, Lucki I, Richels K (1983). Co-syntropin (ACTH a1-24)
stimulation test in depressed patients and healthy subjects. Am J Psychiatry 140:907-909.
Arango V., Ernsberger P., Marzuk PM, Chen JS, Tierney H, Stanley M, Reiss DJ, Mann JJ.
Autoradiographic demonstration of increased serotonin 5-HT2 and b-adrenergic receptor binding sites in
the brain of suicide victims. Arch Gen Psychiatry. 1990;47:1038-1047.
Arango V, Underwood MD, Boldrini M, Tamir H, Kassir SA, Hsiung S, Chen JJ, Mann JJ. Serotonin
1A Receptors, Serotonin Transporter Binding and Serotonin Transporter mRNA Expression in the
Brainstem of Depressed Suicide Victims. Neuropsychopharmacology. 2001;25:892-903.
Arato, M., Frecska, E., Tekes, K., MacCrimmon, D.J. (1991). Serotonergic interhemispheric
asymmetry: gender difference in the orbital cortex. Acta Psychiatrica Scandinavica, 84(1):110-1.
Arriza, J.L., Weinberger, C., Cerelli, G., Glaser, T.M., Handelin, B.L., Housman, D.E., & Evans, R.M.
(1987). Cloning of human mineralocorticoid receptor complementary DNA: Structural and functional
kinship with the glucocorticoid receptor. Science, 237:268-275.
Barden, N., Reul, J.M.H.M. & Holsboer, F. (1995). Do antidepressants stabilize mood through actions
on the hypothalamic-pituitary-adrenocortical system? Trends in Neuroscience, 18:6-10.
18
Baxter LR Jr, Schwartz JM, Phelps ME, et al. (1989)Reduction of prefrontal cortex glucose metabolism
common to three types of depression. Arch Gen Psychiatry. 46:243-250
Berendsen HH. Interactions between 5-hydroxytryptamine receptor subtypes: is a disturbed receptor
balance contributing to the symptomatology of depression in humans? Pharmacol Ther. 1995;66:17-3
Bhagwagar Z, Hafizi S, Cowen PJ. (2003) Increase in concentration of waking salivary cortisol in
recovered patients with depression. Am J Psychiatry. 160:1890-1
Biegon, A., & Greuner, N. (1992). Age-related changes in serotonin 5HT2 receptors on human blood
platelets. Psychopharmacology, 108(1-2):210-212.
Bierut LJ, Heath AC, Bucholz KK, Dinwiddie SH, Madden PAF, Statham DJ et al. Major
Depressive Disorder in a Community-Based Twin Sample. Are There Different Genetic and
Environmental Contributions for Men and Women? Arch.Gen.Psychiatry 1999;56:557-63.
Biver F, Lotstra F, Monclus M, Wikler D, Damhaut P, Mendlewicz J, Goldman S. (1996) Sex
difference in 5HT2 receptor in the living human brain. Neurosci Lett. 204: 25-8.
Blier P, de Montigny C. Current advances and trends in the treatment of depression. Trends Pharmacol
Sci. 1994;15:220-226.
19
Bonson, K.R., Johnson ,R.G., Fiorella, D., et al. (1994). Serotonergic control of androgen-induced
dominance. Pharmacol Biochem Behavior, 49: 313-322.
Borisova, N.A., Proshlyakova, E.V., Sapronova, A.Y., Ugrumov, M.V. (1996). Androgen-dependent
sex differences in the hypothalamic serotoninergic system. European Journal of Endocrinology,
134(2):232-235.
Borsini F. Balance between cortical 5-HT1A and 5-HT2 receptor function: hypothesis for a faster
antidepressant action. Pharmacol Res. 1994;30:1-11.
Brady, L., Whitfield, H.J., Fox, R.J., et al. (1991). Long-term antidepressant administration alters
corticotropin releasing hormone, tyrosine hydroxylase and mineralocorticoid receptor gene expression in
rat brain. Therapeutic implications. Journal of Clinical Investigations, 87: 831-837.
Brown, G.W. & Harris. T. (1978). Social origins of depression: a study of psychiatric disorder
in women. New York, The Free Press.
Buchsbaum MS, Wu J, DeLisi LE, et al. (1986) Frontal cortex and basal ganglia metabolic rates
assessed by positron emission tomography with FDG in affective illness. Aff Disord. 10:137-152
Burgess, L.H., Handa, R.J. (1992). Chronic estrogen-induced alterations in adrenocorticotropin and
corticosterone secretion, and glucocorticoid receptor-mediated functions in female rats. Endocrinology,
131: 1261-1269.
20
Butler, P.D., Nemeroff, C.B. (1990). Corticotropin releasing factor as a possible cause of comorbidity in
anxiety and depressive disorders. In Comorbidity of Mood and Anxiety Disorders (JD Maser, CR
Cloninger, Eds.) American Psychiatric Press, Washington DC.
Carey, M.P., Deterd, C.H., de Koning, J., Helmerhorst, DeKloet, E.R. (1995). The influence of
ovarian steroids on hypothalamic-pituitary-adrenal regulation in the femal rat.
Journal of
Endocrinology, 144:311-332.
Carlsson, M. & Carlsson, A. (1988). A regional study in sex differences in rat brain serotonin.
Progress in Neuro-Psychopharmacology & Biological Psychiatry, 12(1):53-61.
Carroll, B.J., Curtis, G.C., Mendels, J. (1976). Neuroendocrine regulation in depression I. Limbic
system-adrenocortical dysfunction. Archives of General Psychiatry, 33:1039-1044.
Carroll BJ, Feinberg M, Greden JF, Tarika J, Albala AA, Haskett RF, James N, Kronfol Z, Lohr N,
Steiner M, DeVigne JP, Young EA (1981). A specific laboratory test for the diagnosis of melancholia.
Arch Gen Psychiatry 38:15-22.
Charney, D., Woods, S., Goodman, W., et al. (1987). Neurobiological mechanisms of panic anxiety:
biochemical and behavioral correlates of yohimbine-induced panic attacks. American Journal of
Psychiatry, 144: 1030-1036.
Chrousos, G.P., Gold, P.W. (1992). The concepts of stress and stress system disorders. Overview of
physical and behavioral homeostasis. JAMA, 267: 1244-1252.
21
Dean, C., Williams, R.J., Brockington, I.F. (1989). Is puerperal psychosis the same as bipolar maicdepressive disorder? a family study. Psychological Medicine, 19: 637-647.
Drevets WC, Frank E, Price JC, Kupfer DJ, Greer PJ, Mathis C. Serotonin type-1A receptor imaging in
depression. Nucl Med Biol. 2000;27:499-507.
Drevets WC, Frank E, Price JC, Kupfer DJ, Holt D, Greer PJ, Huang Y, Gautier C, Mathis C. PET
imaging of serotonin 1A receptor binding in depression. Biol Psychiatry. 1999;46:1375-1387.
Drevets WC, Videen TO, Price JL, et al. (1992) A functional anatomical study of unipolar depression. J
Neurosci. 12: 3628-3641
Duncan, M.R. & Duncan, G.R. (1979). An in vivo study of the action of antiglucocorticoids on thymus
weight ratio, antibody titre and the adrenal-pituitary-hypothalamus axis. Journal of Steroid
Biochemistry, 10:245-259.
Ehrenkranz, J.R. (1976). Effects of sex steroids on serotonin uptake in blood platelets. Acta
Endocrinologica, 83(2):420-428.
Eriksson, E., Hedberg, M.A., Andersch, B., et al. (1995). The serotonin reuptake inhibiotr paroxetine is
superior to the noradrenaline reuptke inhibitor maprotiline in the treatment of premenstrual syndrome.
Neuropsychopharmacology, 12: 167-176.
22
Fink, G., Sumner, B.E., Rosie, R., Grace, O., & Quinn J.P. (1996). Estrogen control of central
neurotransmission: effect on mood, mental state, and memory. Cellular & Molecular Neurobiology,
16(3):325-344.
Fishette, C.T., Biegon, A., McEwen, B.S. (1984). Sex steroid modulation of the serotonin behavioral
syndrome. Life Sciences, 35: 1197-1206.
Frank, E., Anderson, B., Reynolds, C., Ritenour, A., Kupfer, D.J. (1994). Life events and the
research diagnostic criteria endogenous subtype: a confirmation of the distinction using the
Bedford College methods. Archives of General Psychiatry, 51:519-524.
Galliven EA, Singh A, Michelson D, Bina S, Gold PW, Deuster PA (1997) Hormonal and
metabolic responses to exercise across time of day and menstrual cycle phase. Journal of Applied
Physiology 6:1822-1831.
Gelfin, Y. Lerer, B., Lesch, K.P., Gorfine, M., Allolio, B. (1995). Complex effects of age and
gender on hypothermic, adrenocorticotrophic hormone and cortisol responses to ipsapirone
challenge in normal subjects. Psychopharmacology. 120(3):356-364.
George MS, Ketter TA, Post RM. (1994) Prefrontal cortex dysfunction in clinical depression.
Depression. 2: 59-72
George MS, Ketter TA, Parekh PI, Horwitz B, Herscovitch P, Post RM. (1995) Brain activity
during transient sadness and happiness in healthy women. Am J Psychiatry. 152: 341-51.
23
Gold PW, Loriaux DL, Roy A, Kling MA, Calabrese JR, Kellner CH, Nielman LK, Post RM, Pickar D,
Gallucci W, Avgerinos P, Paul S, Oldfield EH, Cutler GB, Chrousos GP (1986). Response to
corticotropin-releasing hormone in the hypercortisolism of depression and Cushing's disease. N Engl J
Med 314:1329-35.
Gregoire, A., Kumar, R., Everitt, B., et al. (1996). Transdermal oestrogen for treatment of severe
postnatal depression. Lancet, 347: 930-933.
Greist, J.H., Jefferson, J.W., Kobak, K.A., et al. (1995). Efficacy and tolerability of serotonin transport
inhibitors in obsessive-compulsive disorder. Archives of General Psychiatry, 52: 53-60.
Halbreich U, Asnis GM, Schindledecker R, Zurnoff B, Nathan RS. (1985). Cortisol secretion in
endogenous depression I. Basal plasma levels. Arch Gen Psychiatry 42:909-914.
Haleem, D.J., Kennett, G.A. & Curzon, G. (1990). Hippocampal 5-hydroxytryptamine synthesis is
greater in female rats than in males and more decreased by the 5-HT1A agonist 8-OH-DPAT. Journal of
Neural Transmission, 79(1-2):93-101.
Heinsbroek, R.P., van Haaren, F., Feenstra, M.G., van Galen, H., Boer, G., van de Pool, N.E. (1990).
Sex differences in the effects of inescapable footshock on central catecholaminergic and serotonergic
activity. Pharmacology, Biochemistry & Behavior, 37(3):539-550.
24
Heninger, G.R., Charney, D.S. (1987). Mechanism of action of antidepressant treatment: Implications
for the etiology and treatment of depressive disorders, in Psychopharmacology: The Third Generation of
Progress. Edited by H. Y. Meltzer. (pp. 535-544). New York, Raven Press.
Holsboer F, Bardeleden U, Gerken A, Stalla G, Muller O (1984). Blunted corticotropin and normal
cortisol response to human corticotropin-releasing factor in depression. N Engl J Med 311:1127.
Hrdina, P.D., Bakish, D., Chudzik, J., Ravindran, A., Lapierre, Y.D. (1995). Serotonergic markers in
platelets of patients with major depression: upregulation of 5-HT2 receptors. Journal of Psychiatry &
Neuroscience. 20(1):11-19.
Hrdina PD, Demeter E, Vu TB, Sotonyi P, Palkovits M. 5-HT uptake sites and 5-HT2 receptors in brain
of antidepressant-free suicide victims/depressives: Increase in 5-HT2 sites in cortex and amygdala.
Brain Res. 1993;614:37-44.
Issa, A.M., Rowe, W. & Meaney, M.J. (1990). Hypothalamic-pituitary-adrenal activity in aged,
cognitively impaired and cognitively unimpaired rats. Journal of Neuroscience, 10:3247-3254.
Jones I, Kent L, Craddock N. Genetics of Affective Disorders. In McGuffin P, Owen MJ,
Gottesman II, eds. Psychatric Genetics & Genomics, pp 211-45. Oxford: Oxford University
Press, 2002.
25
Jones, M.T., Brush, F.R., & Neame, R.L.B. (1972). Characteristics of fast feedback control of
corticotrophin release by corticosteroids. Journal of Endocrinology, 55:489.
Keller-Wood, M., Silbiger, J. & Wood, C.E. (1988). Progesterone attenuates the inhibition of
adrenocorticotropin responses by cortisol in nonpregnant ewes. Endocrinology, 123:647-651.
Kendler KS. Major Depression and the Environment: A Psychiatric Genetic Perspective.
Pharmacopsychiat. 1998;31:5-9.
Kendler KS, Gardner CO, Neale MC, Prescott CA. Genetic risk factors for major depression in
men and women: similar or different heritabilities and same or partly distinct genes?
Psychol.Med. 2001;31:605-16.
Kendler, K.S., Kessler, R.C., Walters, E.E., MacLean, C., Neale, M.C., Heath, A.C., & Eaves,
L.J. (1995a). Stressful life events, genetic liability and onset of an episode of major depression
in women. American Journal of Psychiatry, 152:833-842.
Kendler KS, Pedersen NL, Neale MC, Mathe AA. A pilot Swedish twin study of affective
disorders including hospital- and population-ascertained subsamples: results of model fitting.
Behav.Genet. 1995;25:217-32.
Kessler, R.C., McGonagle, K.A., Swartz, M., Blazer, D.G., Nelson, C.B. (1993). Sex and
depression in the National Comorbidity Survey I: lifetime prevalence, chronicity and recurrence.
Journal of Affective Disorders, 29:85-96.
26
Ketter TA, George MS, Kimbrell TA, et al. (1996)Functional Brain Imaging, Limbic Function,
and Affective Disorders. The Neuroscientist.; 2: 55-65
Kirschbaum C, Kudielka BM, Gaab J, Schommer NC, Hellhammer DH (1999): Impact of
gender, menstrual cycle phase and oral contraceptives on the hypothalamic-pituitary-adrenal axis.
Psychosomatic Medicine 64:154-162
Kirschbaum, C., Pirke, K-M, & Hellhammer, D.H. (1995). Preliminary evidence for reduced
cortisol responsivity to psychological stress in women using oral contraceptive medication.
Psychoneuroendocrinology, 20:509-514.
Kirschbaum, C., Schommer, N., Federenko, I., Gaab, J., Neumann, O., Oellers, M., Rohleder, N.,
Untiedt, A., Hanker, J., Pirke, K., Hellhammer, D. (1996). Short-term estradiol treatment enhances
pituitary-adrenal axis and sympathetic responses to psychosocial stress in healthy young men. JCEM,
81:3639-3643.
Klimek V, Zak-Knapik J, Mackowiak M. Effects of repeated treatment with fluoxetine and citalopram,
5-HT uptake inhibitors, on 5-HT1A and 5-HT2 receptors in the rat brain. J Psychiatry Neurosci.
1994;19:63-67.
27
Komesaroff PA, Esler M, Clarke IJ, Fullerton MJ, Funder JW.
Effects
of
estrogen
and
estrous cycle on glucocorticoid and catecholamine responses to stress in sheep. Am J Physiol.
1998;275:E671-8
Komesaroff PA, Esler MD, Sudhir K. Estrogen supplementation attenuates glucocorticoid and
catecholamine responses to mental stress in perimenopausal women. J Clin Endocrinol Metab.
1999 ;84:606-10
Krishnan KRR, Ritchie JC, Saunders WB, Nemeroff CB, Carroll BJ (1990). Adrenocortical sensitivity
to low-dose ACTH administration in depressed patients. Biol Psychiatry 27:930-933.
Leckman, J.F., Goodwin, W.K., North, W.G., et al. (1994). The role of central oxytocin in obssessivecompulsive disorder and related normal behavior. Psychoneuroendocrinology, 19:723-749.
Lerer, B., Gillon, D., Lichtenberg, P., Gorfine, M., Gelfin, Y., Shapira, B. (1996). Interrelationship of
age, depression, and central serotonergic function: evidence from fenfluramine challenge studies.
International Psychogeriatrics, 8(1):83-102.
López JF, Vázquez DM, Chalmers DT, Akil H, Watson SJ: Regulation of 5-HT receptors and
the Hypothalamic-Pituitary-Adrenal axis: Implications for the neurobiology of suicide. Ann NY
Acad Sci. 1997;836:106-134
Lyons MJ, Eisen SA, Goldberg J, True W, Lin N, Meyer JM et al. A Registry-Based Twin Study of
Depression in Men. Arch.Gen.Psychiatry 1998;55:468-72.
28
Marcusson J, Oreland L and Winblad B 1984 Effect of age on human brain serotonin
(S-1) binding sites. J Neurochem 43(6):1699-1705.
Martinez-Conde E, Leret ML, Diaz S: The influence of testosterone in the brain of the male rat on levels
of serotonin (5-HT) and 5-hydroxyindoleacetic acid (5-HIAA). Comp Biochem Physiol 80: 411-414,
1985
Matsuda T, Nakano Y, Kanda T, et al.: Gonadal hormones affect the hypothermia induced by
serotonin1A(5HT1A) receptor activation. Life Sci 48: 1627-1632, 1991
Mayberg HS. (1994) Frontal lobe dysfunction in secondary depression. J Neuropsych Clin Neurosci.
6:428-442
Mayberg HS, Lewis PJ, Regenold W, et al. (1994) Paralimbic Hypoperfusion in Unipolar
Depression. J Nuc Med. ; 35: 929-934.
Mayberg H, Brannan S, Mahurin R, et al. (1997) Cingulate function in depression: A potential
predictor of treatment response. NeuroReport. 8:1057-1061
Mayberg HS, Brannan SK Mahurin RK, et al. (2000) Regional Metabolic Effects of Fluoxetine in Major
Depression: Serial Changes and Relationship to Clinical Response. Biol Psychiatry. 48:830-843.
29
Mayberg HS, Silva JA, Brannan SK, et al. (2002) The Functional Neuroanatomy of the Placebo Effect,
Am J Psych. 159:728-737.
Mc Ewen BS: Adrenal steroid action on brain: Dissecting the fine line between protection and damage.
In Neurobiological and Clinical Consequences of Stress: From Normal Adaptation to PTSD (MJ
Friedman, DS Charney, AY Deutch, Eds.), Lippincott-Raven, Philadelphia, PA 1995.
McGuffin P, Katz R, Watkins S, Rutherford J. A hospital-based twin register of the heritability
of DSM-IV unipolar depression. Arch.Gen.Psychiatry 1996;53:129-36.
McQueen JK, Wilson H and Fink G 1997 Estradiol-17 beta increases serotonin transporter
(SERT) mRNA levels and the density of SERT-binding sites in female rat brain. Brain Research.
Molecular Brain Research 45(1):13-23.
Meijer OC and de Kloet R 1998 Corticosterone and serotonergic neurotransmission in the hippocampus:
functional implications of central corticosteroid receptor diversity. Critical Rev in Neurobiology
12(1&2):1-20.
Mendelson SD, McEwen BA: Testosterone increases the concentration of (3H)8-hydroxy-2-(di-npropylamino)tetralin binding at 5-HT1A receptors in the medial preoptic nucleus of the castrated male
rat. Eur J Pharmacol 181: 329-331, 1990
30
Mendelson SD and McEwen BS 1991 Autoradiographic analyses of the effects of restraint-induced
stress on 5-HT1A, 5-HT1C and 5-HT2 receptors in the dorsal hippocampus of male and female rats.
Neuroendocrinology 54(5):454-461.
Monteleone P. Catapano F. Tortorella A. Maj M. Cortisol response to d-fenfluramine in patients with
obsessive-compulsive disorder and in healthy subjects: evidence for a gender-related effect.
Neuropsychobiology. 36(1):8-12, 1997
Murua VS, Molina VA: Effects of chronic variable stress and antidepressant drugs on behavioral
inactivity during an uncontrollable stress: interaction beteen both treatments. Behav Neural Biol 57: 879, 1992
Pecins-Thompson M, Brown NA, Kohama SG, et al.: Ovarian steroid regulation of tryptophan
hydroxylase mRNA expression in rhesus macaques. J Neuroscience 16: 7021-7029, 1996
Peroutka SJ, Snyder SH. Regulation of serotonin2 (5-HT2) receptors labeled with [3H]spiroperidol by
chronic treatment with the antidepressant amitriptyline. J Pharmacol Exp Ther. 1980;215:582-587.
Pfohl B, Sherman B, Schlecte J, Stone, R (1985). Pituitary/adrenal axis rhythm disturbances in
psychiatric patients. Arch Gen Psychiatry 42:897-903.
Raadsheer FC, Hoogendijk WJ, Stam FC, Tilders FJ, Swaab DF. Increased numbers of
corticotropin-releasing hormone expressing neurons in the hypothalamic paraventricular nucleus
of depressed patientsNeuroendocrinology. 1994 Oct;60(4):436-44.
31
Reul JM, Stec I, Soder M, et al.: Chronic treatment of rats with the antidepressant amitriptyline
attenuates the activity of the hypothalamic-pituitary adrenocortical system. Endocrinology 133:
312-20, 1993
Roca CA, Altemus M, Galliven E, Schmidt PJ, Deuster P, Gold P, Murphy D, and Rubinow D:
Effect of reproductive hormones on the hypothalamic-pituitary-adrenal axis response to stress.
Biological Psychiatry 43: 6S, 1998a
Roca CA, Schmidt PJ, Altemus M, Dananceau M and Rubinow D. Effects of reproductive
steroids on the Hypothalamic-pituitary-adrenal axis response to low dose dexamethasone.
Abstract at Neuroendocrine Workshop on Stress, June 21-23, 1998b, New Orleans.
Roca, C.A., P.J. Schmidt, M. Altemus, P. Deuster, M.A. Danaceau, K. Putnam & D.R. Rubinow.
2003. Differential menstrual cycle regulation of hypothalamic-pituitary-adrenal axis in women
with premenstrual syndrome and controls. J. Clin. Endocrinol. Metab. 88: 3057-3063.
Rousseau GG, Baxter JD and Tomkins GM: Glucocorticoid receptors: relations between steroid binding
and biological effects. Mol. Biol. 67:99-115, 1972
Rubin RT, Poland RE, Lesser IM, Winston RA, Blodgett N (1987). Neuroendocrine aspects of primary
endogenous depression I. Cortisol secretory dynamics in patients and matched controls. Arch. Gen.
Psychiatry 44:328-336.
32
Rubinow DR, Roy-Byrne PP: Premenstrual syndromes: overviews from a methodologic perspective.
American Journal of Psychiatry 141: 163-172, 1984
Rutter M. The Interplay of Nature, Nurture, and Developmental Influences. Arch.Gen.Psychiatry
2002;59:996-1000.
Ryan N. Birmaher B. Perel JM. Dahl RE. Meyer V. Al-Shabbout M. Iyengar S. Puig-Antich J.
Neuroencocrine response to L-5-hydroxytryptophan challenge in prepubertal major depression.
Archives of General Psychiatry. 49:(11)843-851, 1992
Sachar, E.J., Hellman, L., Roffwarg, H.P., Halpern, F.S., Fukush, D.K., Gallagher, T.F. (1973).
Disrupted 24 hour patterns of cortisol secretion in psychotic depressives. Arch. Gen. Psychiatry
28:19-24.
Schreiber R, De Vry J. Neuronal circuits involved in the anxiolytic effects of the 5-HT1A receptor
agonists 8-OH-DPAT ipsapirone and buspirone in the rat. Eur J Pharmacol. 1993; 249:341-351.
Seckl JR, Fink G 1992 Use of in situ hybridization to investigate the regulation of hippocampal
corticosteroid receptors by monoamines. J Steroid Biochem & Mol Biology 40(4-6):685-688.
Sheline YI, Gado MH, Kraemer HC. Untreated depression and hippocampal volume loss. Am J
Psychiatry. 2003 Aug;160(8):1516-8
33
Sheline YI, Wang PW, Gado MH, Csernansky JG, Vannier MW. 1996 Hippocampal atrophy in
recurrent major depression. Proc Natl Acad Sci USA. 1996;93:3908-13
Southwick S, Krystal J, Morgan C: Abnormal noradrenergic function in posttraumatic stress disorder.
Arch Gen Psychiatry 50: 266-274, 1993
Sullivan PF, Neale MC, Kendler KS. Genetic epidemiology of major depression: review and
meta-analysis. Am.J.Psychiatry 2000;157:1552-62.
Svec F: Differences in the interaction of RU 486 and ketoconazole with the second binding site of the
glucocorticoid receptor. Endocrinology 123:1902-06, 1988
Turner BB and Weaver DA: Sexual dimorphism of glucocorticoid binding in rat brain. Brain Res.
343:16-23, 1985
Viau V and Meaney MJ Variations in the hypothalamic-pituitary-adrenal response to stress
during the estrous cycle in the rat. Endocrinology 129:2503-2511, 1991
von Bardeleben U, Stalla GK, Mueller OA, Holsboer F (1988). Blunting of ACTH response to CRH in
depressed patients is avoided by metyrapone pretreatment. Biol Psychiatry 24:782-786.
Weissman MM, Olfson M: Depression in women: implications for health care research. Science 269:
799-801, 1995
34
Welner SA, De Montigny C, Desroches J, Desjardins P, Suranyi-Cadotte BE. Autoradiographic
quantification of serotonin1A receptors in rat brain following antidepressant drug treatment. Synapse.
1989;4:347-352.
Yates M, Leake A, Candy JM, Fairbairn AF, McKeith IG, Ferrier IN. 5HT2 receptor changes in major
depression. Biol Psychiatry. 1990;27: 489-496.
Yoshino K 1982 [Concentrations of monoamines and monoamine metabolites in cerebrospinal
fluid determined by high-performance liquid chromatography with electrochemical detection].
Brain & Nerve 34(11):1099-1106.
Young EA (1996). Sex differences in response to exogenous corticosterone. Molecular
Psychiatry 1: 313-319.
Young EA, Abelson JL and Cameron OG. (2004) Effect of Comorbid Anxiety Disorders on the
HPA Axis Response to a Social Stressor in Major Depression, under revsion, Biological
Psychiatry
Young EA, Aggen SH, Prescott CA, Kendler KS (2000). Similarity in saliva cortisol measures in
monozygotic twins and the influence of past major depression. Biological Psychiatry, 48:70-74.
Young EA, Altemus M, Parkison V and Shastry S. Effects of Estrogen Antagonists and Agonists
on the ACTH response to restraint stress. Neuropsychopharmacology, 25:881-891, 2001a
35
Young EA and Breslau N. (in press) Cortisol and Catecholamines in Posttraumatic Stress
Disorder: A Community Study, Archives General Psychiatry, in press. .
Young EA, Carlson NE, Brown MB (2001b). 24 Hour ACTH and Cortisol Pulsatility in
Depressed Women, Neuropsychpoharmacology, 25:267-276.
Young EA, Haskett RF, Grunhaus L, Pande A, Weinberg VM, Watson SJ, Akil H (1994).
Increased circadian activation of the hypothalamic pituitary adrenal axis in depressed patients in
the evening. Arch Gen Psychiatry 51:701-707.
Young E.A, Haskett RF, Watson SJ and Akil H:
Loss of Glucocorticoid Fast Feedback in
Depression. Archives of General Psychiatry 48:693-699, 1991
Young EA, Kotun J, Haskett RF, Grunhaus L, Greden JF, Watson SJ, Akil H: Dissociation
between pituitary and adrenal suppression to dexamethasone in depression.
Arch. Gen.
Psychiatry 50:395-403, 1993
Young EA, Lopez JF, Murphy-Weinberg V, Watson SJ and Akil H (1997). Normal Pituitary
Response to Metyrapone in the Morning in Depressed Patients: Implications for Circadian
Regulation of CRH Secretion, Biological Psychiatry, 41: 1149-1155.
Young EA and Vazquez D: Hypercortisolemia, hippocampal glucocorticoid receptors and fast
feedback. Molecular Psychiatry 1:149-159, 1996
36
Young EA, Watson SJ, Kotun J, Haskett RF, Grunhaus L, Murphy-Weinberg V, Vale W, Rivier
J, Akil H (1990). Response to low dose oCRH in endogenous depression: role of cortisol
feedback. Arch Gen Psychiatry 47:449-57.
Zweifel JE, O'Brien WH: A meta-analysis of the effect of hormone replacement therapy upon depressed
mood. Psychoneuroendocrinology 22: 189-212, 1997
Download