16
Neuroscienceand BiobehavioralReviews, Vol. 16, pp. 115-130, 1992 0149-7634/92 $5.00 + .00 Printed in the USA. All rights reserved. Copyright© 1992 Pergamon Press Ltd. Mechanisms of Stress: A Dynamic Overview of Hormonal and Behavioral Homeostasis ELIZABETH O. JOHNSON,*t t THEMIS C. KAMILARIS,*t GEORGE P. CHROUSOS* AND PHILIP W. GOLDt *Developmental Endocrinology Branch, National hlstitute of Child Health and Human Development and "~Clinical Neuroendocrinology Branch, National hzstitute of Mental Health, NIH, Bethesda MD 20892 Received 13 December 1990 JOHNSON, E. O., T. C. KAMILARIS, G. P. CHROUSOS AND P. W. GOLD. Mechanisms of stress: A dynamic overview of hormonal and behavioral homeostasis. NEUROSCI BIOBEHAV REV 16(2) 115-130, 1992.-Environmental events, both physi- cal and emotional, can produce stress reactions to widely varying degrees. Stress can affect many aspects of physiology, and levels of stress, emotional status, and means of coping with stress can influence health and disease. The stress system consists of brain elements, of which the main components are the corticotropin-releasing hormone (CRH) and locus ceruleus (LC)-norepi- nephrine (NE)/autonomic systems, as well as their peripheral effectors, the pituitary-adrenal axis and the autonomic system, which function to coordinate the stress response. Activation of the stress system results in behavioral and physical changes which allow the organism to adapt. This system is closely integrated with other central nervous system elements involved in the regulation of behavior and emotion, in addition to the axes responsible for reproduction, growth and immunity. With current trends in stress research which focus on understanding the mechanisms through which the stress-response is adaptive or becomes maladaptive, there is a growing association of stress system dysfunction, characterized by hyperactivity and/or hypoactivity to various patho- physiological states. The purpose of this review is to 11 define the concepts of stress and the stress response from a historical perspective, 2) present a dynamic overview of the biobehavioral mechanisms that participate in the stress response, and 3) exam- ine the consequences of stress on the physiologic and behavioral well-being of the organism by integrating knowledge from appar- ently disparate fields of science. Stress HPA axis Autonomic system Aging Reproductive suppression Growth retardation Immunosuppression Depression DEFINITION OF STRESS A critical problem faced by investigators lies in the defini- tion of stress and related concepts (Table 1). The term "stress" describes a state of threatened "homeostasis" (Greek for "'steady state") or threatened harmony, balance, or equilibrium. The threatening, disturbing forces are defined as "'stressors,'" while the counteracting forces put forth to neutralize the effects of the stressors and reestablish homeostasis are called the "'adaptive response" (42). A major problem in stress biology is confusion regarding the definitions for "stress," "stressor," "'adaptive re- sponses," and "consequences of stress." Historical Development of Stress as a Concept The concept of "homeostasis" goes back to the ancient Greeks (Table 2). The natural philosopher Empedocles consid- ered that all matter was a harmonious mixture of elements and qualities (42,65). This early expression of homeostasis was ex- tended to living beings by Hippocrates, who considered health as the state of harmonious balance, and disease as the state of dysharmony (3). Hippocrates described disturbing forces of na- ture as causes of disease and referred to the healing forces in- herent to the organism as the "healing power of Nature." This was later called by Galen "vis medicatrix Naturae.'" Psychogenic stress was mentioned by Epicurus, who suggested that coping with emotional stressors was a way to improve the "quality" of life (42,65). In the early nineteenth century, the French physiologist Claude Bernard introduced a theory suggesting that, as organisms be- come more independent of their surroundings, they develop more complex ways of stabilizing their internal environments to counter the changes in their external environment. The impor- tance of adaptive mechanisms was thus recognized as the con- stancy of the "milieu interieur,- which would be the condition of a free and independent existence (18). In the early 1900's, Walter Cannon expanded this theory and coined the term "homeostasis." He demonstrated in several seminal experiments that the sympathoadrenal system was re- sponsible for coordinating the "fight or flight" response neces- sary to meet external challenges (27). Cannon was able to show that both physical and emotional disturbances triggered the same response from the organism. In addition, he proposed that there was a "critical" level of stress, in terms of magnitude and du- ration, against which the homeostatic mechanisms fail and the organism perishes. Cannon believed that an individual organ- ism's susceptibility to this critical stress "varied under different tRequests for reprints should be addressed to Elizabeth O. Johnson, Developmental Endocrinology Branch, National Institute of Child Health and Human Development, NIH, Bldg. 10, Room 10N262, Bethesda, MD 20892. 115

Mechanisms of Stress

Embed Size (px)

Citation preview

Page 1: Mechanisms of Stress

Neuroscience and Biobehavioral Reviews, Vol. 16, pp. 115-130, 1992 0149-7634/92 $5.00 + .00 Printed in the USA. All rights reserved. Copyright © 1992 Pergamon Press Ltd.

Mechanisms of Stress: A Dynamic Overview of Hormonal and Behavioral Homeostasis

E L I Z A B E T H O. J O H N S O N , * t t T H E M I S C. K A M I L A R I S , * t G E O R G E P. C H R O U S O S * A N D PHILIP W. G O L D t

*Developmental Endocrinology Branch, National hlstitute o f Child Health and Human Development and "~Clinical Neuroendocrinology Branch, National hzstitute o f Mental Health, NIH, Bethesda MD 20892

Rece ived 13 D e c e m b e r 1990

JOHNSON, E. O., T. C. KAMILARIS, G. P. CHROUSOS AND P. W. GOLD. Mechanisms of stress: A dynamic overview of hormonal and behavioral homeostasis. NEUROSCI BIOBEHAV REV 16(2) 115-130, 1992.-Environmental events, both physi- cal and emotional, can produce stress reactions to widely varying degrees. Stress can affect many aspects of physiology, and levels of stress, emotional status, and means of coping with stress can influence health and disease. The stress system consists of brain elements, of which the main components are the corticotropin-releasing hormone (CRH) and locus ceruleus (LC)-norepi- nephrine (NE)/autonomic systems, as well as their peripheral effectors, the pituitary-adrenal axis and the autonomic system, which function to coordinate the stress response. Activation of the stress system results in behavioral and physical changes which allow the organism to adapt. This system is closely integrated with other central nervous system elements involved in the regulation of behavior and emotion, in addition to the axes responsible for reproduction, growth and immunity. With current trends in stress research which focus on understanding the mechanisms through which the stress-response is adaptive or becomes maladaptive, there is a growing association of stress system dysfunction, characterized by hyperactivity and/or hypoactivity to various patho- physiological states. The purpose of this review is to 11 define the concepts of stress and the stress response from a historical perspective, 2) present a dynamic overview of the biobehavioral mechanisms that participate in the stress response, and 3) exam- ine the consequences of stress on the physiologic and behavioral well-being of the organism by integrating knowledge from appar- ently disparate fields of science.

Stress HPA axis Autonomic system Aging Reproductive suppression Growth retardation Immunosuppression Depression

DEFINITION OF STRESS

A critical problem faced by investigators lies in the defini- tion of stress and related concepts (Table 1). The term "st ress" describes a state of threatened "homeostasis" (Greek for "'steady state") or threatened harmony, balance, or equilibrium. The threatening, disturbing forces are defined as "'stressors,'" while the counteracting forces put forth to neutralize the effects of the stressors and reestablish homeostasis are called the "'adaptive response" (42). A major problem in stress biology is confusion regarding the definitions for "s t ress ," "s tressor ," "'adaptive re- sponses," and "consequences of stress."

Historical Development of Stress as a Concept

The concept of "homeostasis" goes back to the ancient Greeks (Table 2). The natural philosopher Empedocles consid- ered that all matter was a harmonious mixture of elements and qualities (42,65). This early expression of homeostasis was ex- tended to living beings by Hippocrates, who considered health as the state of harmonious balance, and disease as the state of dysharmony (3). Hippocrates described disturbing forces of na- ture as causes of disease and referred to the healing forces in- herent to the organism as the "healing power of Nature." This

was later called by Galen "vis medicatrix Naturae.'" Psychogenic stress was mentioned by Epicurus, who suggested that coping with emotional stressors was a way to improve the "quality" of life (42,65).

In the early nineteenth century, the French physiologist Claude Bernard introduced a theory suggesting that, as organisms be- come more independent of their surroundings, they develop more complex ways of stabilizing their internal environments to counter the changes in their external environment. The impor- tance of adaptive mechanisms was thus recognized as the con- stancy of the "milieu interieur,- which would be the condition of a free and independent existence (18).

In the early 1900's, Walter Cannon expanded this theory and coined the term "homeostasis ." He demonstrated in several seminal experiments that the sympathoadrenal system was re- sponsible for coordinating the "fight or flight" response neces- sary to meet external challenges (27). Cannon was able to show that both physical and emotional disturbances triggered the same response from the organism. In addition, he proposed that there was a "critical" level of stress, in terms of magnitude and du- ration, against which the homeostatic mechanisms fail and the organism perishes. Cannon believed that an individual organ- ism's susceptibility to this critical stress "varied under different

tRequests for reprints should be addressed to Elizabeth O. Johnson, Developmental Endocrinology Branch, National Institute of Child Health and Human Development, NIH, Bldg. 10, Room 10N262, Bethesda, MD 20892.

115

Page 2: Mechanisms of Stress

116 J O H N S O N , KAMILARIS , C H R O U S O S A N D G O L D

TABLE 1

THE CONCEPT OF HOMEOSTASIS

Disturbing Harmony Counteracting Forces ~ Balance ~: Neutralizing

Equilibrium Forces Homeostasis

(Steady State)

II II II Stressors ~ Threatened ,c Adaptive

Homeostasis Responses (stress)

general conditions and during the normal and pathologic ups and down of existence in an ordinary life-cycle" (27).

In 1936, Selye presented his concept of the "General Adap- tation Syndrome," and attention shifted from the sympathetic nervous system to the adrenal glands (183). Selye defined four stages of the stress reaction: 1) an initial "alarm reaction," characterized by an immediate sympathoadrenomedullary dis- charge; 2) a "stage of resistance," characterized by activation of the hypothalamic-pituitary-adrenal (HPA) axis; 3) a stage of adrenal hypertrophy, gastrointestinal ulceration, and thymic and lymphoid shrinkage, which he called the "General Adaptation Syndrome"; and 4) a final stage of exhaustion and death. Chronic alterations in the hormonal responses and abnormal changes in several tissues were believed to cause the "diseases of adaptation" (184,187).

Hams, a contemporary of Selye's, suggested in 1948 that hypothalamic releasing or inhibiting factors regulate anterior pi- tuitary function (83). Saffran and Schally demonstrated in 1955 that a factor from the hypothalamus was regulating ACTH re- lease from the pituitary (166). They named this principle "corti- cotropin-releasing factor." After intensive investigation, in 1981, Vale and others were able to isolate this factor and characterize its structure as a 41 amino-acid peptide (207). When the chemi- cal structure was identified, the term "factor" was changed to "hormone."

Contemporary theory in stress biology conceptualizes an in- tegrated "stress system" consisting of neuroanatomical and func- tional structures that function to produce the behavioral, physiological and biochemical changes directed toward maintain- ing homeostasis (42). This theory is supported by recent find- ings in neurobiology demonstrating anatomical and functional connections between the hypothaiamus, the arousal center in the pons, and several sympathetic nuclei in the hindbrain (26, 141, 209). In the periphery, the adrenocortical and sympathetic divi- sions of the stress system have additional integrative actions. These include complementary and permissive interactions of glucocorticoids and catecholamines in the regulation and mainte- nance of metabolic and cardiovascular homeostasis (22,157).

Classification of Stressors--The Adaptive Response to Stress

The adaptive response to stress appears to depend upon the quality (physical or emotional), strength, and duration (acute, chronic) of the stimulus, as well as upon the constitution and state of the organism (51). A stressor can be viewed as any per- turbation that disrupts homeostasis. Physical stressors include disturbances of the internal environment (anoxia, hypoglycemia, etc.), external extremes (heat and cold), and multifaceted stres- sors (noxious stimuli and physical strain, such as exercise or in-

TABLE 2

HISTORY OF THE CONCEPT OF STRESS

Empedocles (500--430 BC) Hippocrates (460-375 BC)

Epicurus (341-270 BC) Claude Bernard (1813-1878) Walter Cannon (1871-1945) Hans Seyle 1907-1982)

first written reference to homeostasis.

Health is the state of the harmonious balance of the elements, and disease is the state of dysharmony."Nature heals disease" (Noysvn Fyseiw Iatroi). Coping with emotional stressors improves the quality of life. "Milieu interieur."

"Fight or flight" reaction; "homeostasis."

The "General Adaptation Syndrome."

jury). Psychological stressors are stimuli that affect emotion and result in fear, anxiety, or frustration, and are among the most potent activators of the HPA axis (113, 124, 185). It should be noted that stressors may be mixed and act in combination.

The adaptive response to stress includes the behavioral and physiological processes that the organism consistently musters in its attempt to reestablish homeostasis in the face of a wide range of stressors (183,186). In general, the adaptive response to stress involves a redirection of both behavior and energy (43) (Table 3). Behavioral adaptation is viewed as the facilitation of adap- tive, and inhibition of nonadaptive, neural pathways which en- able the organism to cope more successfully with the stressful stimulus. These behavioral responses include altered cognitive and sensory thresholds, increased alertness, selective memory enhancement (27), stress-induced analgesia (204), and suppres- sion of feeding and reproductive behavior (28,191). Peripheral adaptation is viewed as provision of the energy necessary to overcome stressors and involves both a shift of energy substrates from storage sites to the bloodstream, as well as appropriate cardiovascular changes. Glucocorticoids, epinephrine (E), and norepinephrine (NE) act to inhibit glucose uptake, fatty acid storage, and protein synthesis at storage sites and stimulate the release of energy substrates, including glucose, amino acids, and free fatty acids, from muscle, fat tissue and liver (133,218). These changes in energy availability are paralleled by the stimu- lation of cardiovascular and pulmonary function, which include increased heart rate, blood pressure and respiration (218). Simul- taneously, anabolic processes, such as digestion, growth, repro- duction and immune function, are suppressed (105).

It appears that the ability to appropriately regulate the stress response may be as important as the ability to initiate it. Con- tainment of the stress response is crucial to avoid the behavioral and physical consequences of the mobilization of behaviors and resources. Chronic activation of the catabolic processes of the stress response can ultimately become destructive and patho- genic. Thus metabolic (myopathy, fatigue, changes in glycemia) and cardiovascular consequences (hypertension), compromised growth and tissue repair, peptic ulceration, reproductive suppres- sion (impotence and amenorrhea), as well as consequences of immunosuppression (increased susceptibility to infection and can- cer) can occur when the state of stress is unduly prolonged (105). In addition to elimination of, or habituation to, the stres- sor, the containment of the stress response is assisted by various stress-induced substances. Thus, for example, glucocorticoids and opioids suppress both the HPA axis and the central sympa-

Page 3: Mechanisms of Stress

M E C H A N I S M S O F STRESS 117

TABLE 3 BEHAVIORAL AND PHYSIOLOGICAL ADAPTATION

DURING STRESS

Behavioral Adaptation • Altered cognition and attention span level • Increased alertness • Altered sensory threshold • Sharpened memory and sensation • Stress-induced analgesia • Suppression of feeding behavior • Suppression of reproductive behavior

Peripheral Adaptation • Oxygen and nutrients directed to the CNS and stressed body sites • Detoxification from toxic products • Altered cardiovascular tone • Containment of the stress-response

thetic system, while centrally produced norepinephrine inhibits the sympathetic system via local a2-adrenergic receptor inhibi- tion (203).

Although there is a stereotypic consistency of the responses that the body activates during stress, all stressors do not result in identical profiles of behavioral and peripheral responses. These differences appear to be related to genetic constitution, as well as to early-life experiences (112, 163,200). Moreover, not all stressors should be construed as noxious or injurious. Indeed, many stressors promote essential differentiation, growth, and enhanced physiological and behavioral competence, which would be markedly impaired in a relatively stress-free environment. On the other hand, in the context of constant or inescapable stress, or in an organism in which the usual counterregulatory elements of the stress response are relatively inoperative, the effectors of the generalized stress response could produce secondary changes that interfere with adaptation, rather than promote it (181, 182, 213 ,214) . An analogy to this situation of an unrestrained or ex- cessive stress response is that of the spectrum of autoimmune diseases that occur as a consequence of excessive stimulation of the immune response or its escape from its usual counterregula- tory elements (194-196).

Homeostasis and the Components of the Stress Response

The HPA axis and the sympathetic system are important reg- ulators of an animal 's homeostatic functions (205). Thus the or- ganism's response to stress is composed of behavioral, endocrine, and autonomic components, coordinated to neutralize the dis- rupting effects of the "s t ressors" on homeostasis (42, 73, 74). Below, we describe the mechanisms by which the various com- ponents of the stress system restore homeostasis.

MECHANISMS OF THE STRESS RESPONSE

Hypothalamic-Pituitary-Adrenal (HPA) A.ris

The function of the HPA axis has been the subject of intense basic and clinical research which has attempted to understand why glucocorticoids are critical for life. The response to stress is associated with increases in the levels of plasma glucocorti- coids. The secretion of glucocorticoids from the adrenal cortex is under the control of ACTH, which in turn is released from the anterior lobe of the pituitary. ACTH secretion is regulated by corticotropin-releasing hormone (CRH) and other secreta-

Psychogenic/ Emotional Diurnal Stimuli Rhythms

Traumatic ~ / Pressure.Sensitiv e Stimuli ~ \ / //Baroreceptor Signals

/ / / Cytokines/ / ,/ J Mediators of

ACT_.H ~ / / / n ^ nflammation -~.//E ( ~ I - 1 3 - E P " "/6 H /~ ~'-'-''" N E/N PY

~ ~ C R H ~ ~.,~"~ 5HT

f - I / ,,~ " GABA/BZD

I / I /

/ Pituitary \ / /

\ ~\ ACTH

Glucocor ico // Adrenal

FIG. 1. Schematic representation of the putative regulation of the hypo- thalamic-pituitary-adrenal (HPA) axis. The corticotropin-releasing hor- mone (CRH) system is the principal central biologic effector which facilitates a characteristic behavioral and peripheral response to stress. CRH stimulates secretion of both hypothalamic and pituitary POMC gene-derived peptides, the latter resulting in glucocorticoid secretion. The activation of the CRH neurons appears to be regulated by central stimulatory and inhibitory inputs and by multiple negative feedback loops. (Solid lines represent stimulatory effects; broken lines represent inhibitory effects.) The latter include an ultrashort CRH-mediated loop, a short hypothalamic POMC gene-derived peptide loop, including both ACTH and [3-endorphin, and a long glucocorticoid-mediated feedback loop.

gogues from the hypothalamus (99, 133, 218) (Fig. 1). Glucocorticoids exert many different effects, including effects

on cardiovascular function, metabolism, muscle function, behav- ior and the immune system (43). These effects can be grouped into two categories defined as permissive and regulatory (88). "Permiss ive" effects of glucocorticoids function to "permi t" other hormones or factors to accomplish their function at a nor- mal level, and are observed primarily in the resting state and may span the resting and stress states. The permissive role of glucocorticoids is crucial for maintenance of homeostasis at the basal state (31, 103, 134, 157, 206). "Regulatory" effects of glucocorticoids are exerted only by stress-induced levels of these hormones. It has been hypothesized that the regulatory effects of stress-related elevations of glucocorticoids may be necessary

Page 4: Mechanisms of Stress

118 JOHNSON, KAMILARIS, CHROUSOS AND GOLD

to prevent overreaction of the central stress, immune and other systems, which, if unchecked, lead to injury (133).

Several closed feedback mechanisms regulate the secretion of glucocorticoids (99, 133, 218) (Fig. 1). There is a major feed- back loop between glucocorticoids and the hypothalamic-pitu- itary axis. Thus circulating glucocorticoids act on the pituitary directly to inhibit ACTH secretion, and on the hypothalamus to suppress secretion of CRH (99). Additional feedback loops in- clude the inhibitory effects of ACTH, [3-endorphin ([3-EP) and CRH on the hypothalamic CRH neuron (34). These mechanisms enable the organism to maintain a stable blood level of gluco- corticoids at all times, while simultaneously providing an emer- gency override, via the central nervous system (CNS), to respond to stressors.

Corticotropin-Releasing Hormone ( CRH )

It has long been recognized that the endocrine response to stress is coordinated at the CNS. CRH is a 41 amino-acid pep- tide secreted from the hypothalamus and has a putative role in regulating the normal response to stress. The chemical isolation and description of CRH by Vale provided an important missing link in the system (207). The genes encoding the CRH molecule in the human, rat and sheep have been characterized (157, 178, 189, 207). These genes have been highly conserved throughout evolution. In fact, the CRH peptides from humans and rats are identical in amino acid sequence (157,189). The existence of CRH has been known since the 1950's, but synthetic CRH only became available recently; the availability of synthetic CRH has opened new experimental avenues.

Anatomical distribution. CRH, localized by immunocyto- chemistry, is found in neuronal cell bodies in the paraventricular nucleus (PVN) (26,141). CRH neurons project to the median eminence, where they terminate on the capillaries of the hy- pophyseal portal vessels. These vessels function as a direct short vascular pathway from the hypothalamus to the anterior pitu- itary. Thus CRH, which is released into the hypophyseal portal system, is transported to the anterior pituitary, where it stimu- lates pituitary corticotrophs to both synthesize and secrete ACTH. A different set of PVN CRH neurons send projections to the hindbrain, where they stimulate the electrical activity of their target neurons in the arousal and sympathetic centers (208).

There also are extrahypothalamic CRH neurons. Through im- munocytochemical, receptor studies and the detection of CRH mRNA, CRH has been demonstrated in the brainstem, midbrain, striatum, hippocampus, cerebral cortex, spinal cord, sympathetic ganglia and adrenal gland (47,199). The broad distribution of CRH and its receptors in the CNS provides a substrate for the wide-ranging behavioral effects of this peptide.

Receptors. The CRH receptor is highly concentrated in the brain, anterior pituitary, adrenal medulla and sympathetic gan- glia of the rat and several primates (45,209). CRH receptors in pituitary corticotrophs appear to be sensitive to circulating levels of glucocorticoids, as these receptors decrease shortly after adre- nalectomy or during chronic stress (6,217). These conditions are associated with increased hypothalamic CRH and vasopressin (AVP) secretion, and decreased responsiveness to physiologic increases in plasma glucocorticoids, respectively.

Regulation. CRH secretion can be affected by stimuli such as emotion, pain and changes in blood pressure. The PVN has connections with various components of the limbic system, im- plicated in emotions, such as fear, and anger. Pain pathways are believed to be primarily located in the spinothalamic tracts which project via the reticular formation of the brainstem to the PVN. Changes in blood pressure also affect CRH release. Blood pres-

sure is regulated by receptors in the carotid sinus, the aortic arch, major chest veins, and both atria. When blood pressure increases, impulses originating from these receptors travel to the nucleus of the tractus solitarius in the medulla and then to the PVN, where CRH secretion is inhibited. Conversely, CRH se- cretion is increased when blood pressure decreases. The latter has been associated with a decrease in the number of impulses reaching the PVN via the nucleus tractus solitarius (63).

Several neurotransmitter systems regulate PVN CRH release (Fig. 1). Both NE and E stimulate CRH release. It appears that the former stimulates CRH release via the a~-adrenergic receptor (35). Acetylcholine and serotonin are also excitatory mediators participating in both the circadian rhythm and stress-induced re- lease of CRH, whereas gamma-aminobutyric acid (GABA), the opioid peptide system, ACTH, and glucocorticoids are inhibitory (35). Several products of the immune system such as several cy- tokines, including Interleukin-1 (IL-I), Interleukin-2 (IL-2), In- terleukin-6 (IL-6), or inflammatory mediators, such as platelet activating factor (PAF) and tumor necrosis factor (TNF) appear to stimulate secretion of hypothalamic CRH in vitro and in vivo (19, 194, 216).

Behavioral effects. The distribution of CRH within and be- yond the hypothalamus provides an anatomical context for the observation that CRH can simultaneously activate and coordinate metabolic, circulatory and behavioral responses during adaptive situations (59, 64, 202). CRH injected directly into the cerebro- ventricular system produces a number of effects that are remi- niscent of the stress response (28,29). These include neuronal activation, electroencephalographic arousal, and pronounced gen- eral behavioral activation. Behavioral changes are dependent on the situation and dose (102). For example, when CRH is injected ICV in rats, it produces dose-dependent locomotor activation in familiar environments and the "'freeze" posture in foreign envi- ronments (202). At low doses, CRH-induced activation is char- acterized by increased locomotion, sniffing, grooming and rearing. These changes are believed to be consistent with "'general be- havioral arousal." High doses of CRH, on the other hand, pro- duce bizarre behaviors, including repetitive locomotion, irritability, or demonstrations of aggression (102). High doses of CRH ICV have been shown to decrease sexual behaviors (191). Finally, CRH decreascs food intake in the home cage and inhibits in- creases in food intake produced by NE and insulin, implicating CRH as a mediator of stress-related suppression of appetite or food intake (132).

ACTH and Endorphins

Anatomical distribution. Proopiomelanocortin (POMC) is the prohormone for ACTH (122). POMC is synthesized in the brain (arcuate nucleus of the hypothalamus, zona incerta, lateral sep- tum, nucleus accumbens, periventricular thalamus, periaqueduc- tal gray, locus coeruleus, nucleus tractus solitarius, reticular formation, stria terminalis and medial amygdala), pituitary gland, gastrointestinal tract and reproductive organs, and is cleaved into different biologically active peptides (79, 84, 106, 137, 176). In the anterior pituitary, POMC is broken down into ACTH, a 39 amino-acid fragment, and 13-1ipotropin, a 92 amino-acid frag- ment; in turn, 13-1ipoprotein is broken further into other smaller active fragments, including [3-endorphin (Fig. 2). The particular POMC fragments produced in a given tissue or cell type are be- lieved to be specific and determined by local enzymes and pH.

ACTH is transported via the systemic circulation to the adre- nal gland, where it stimulates synthesis and secretion of gluco- corticoids, aldosterone and adrenal androgens. ACTH also has a trophic or sensitizing effect on the adrenal cortex, enhancing the

Page 5: Mechanisms of Stress

MECHANISMS OF STRESS 119

NH2 COOH I I

ACTH (1-391 ~°LPH 11-gl) I I I !

13-MSH 11-131 CLIP 118-391 I'-LPH 11-5111 l~-Enclorphln 161-gl1 I i

~-MSH 137-S8) y-Endorphln (81-77 I I

Enl(ephlllln 161-115)

FIG. 2. Schematic representation of proopiomelanocortin (POMC), the large molecular weight precursor of ACTH, 13-1ipotropic hormone (13- LPH), and [3-endorphin. CLIP represents corticotropin-like intermediate lobe peptide, et-MSH, 13-MSH, and a- and [3-melanocyte-stimulating hormones, respectively.

response to subsequent stimulation. Thus, if an animal is ex- posed to regular, fixed doses of ACTH, the glucocorticoid re- sponse to the dose increases over time, primarily as a result of adrenocortical hypertrophy. The same occurs during chronic stress. Conversely, if ACTH secretion is decreased or elimi- nated, the glucocorticoid response to ACTH is reduced, most probably as a result of adrenal atrophy. Interestingly, ACTH may also have neurotrophic effects. For example, ACTH can fa- cilitate recovery after a sciatic nerve crush (51,63).

Receptors. The ACTH receptor follows a pattern of receptor regulation and homeostasis similar to that observed in most en- docrine systems. When ACTH concentrations are high, recep- tors are desensitized and are downregulated or disappear. Conversely, when ACTH concentrations are low, the number or sensitivity of the receptors increases. Although the ACTH recep- tor system appears to follow a normal pattern, the glucocorti- coid response to ACTH does not. Rather, elevations in ACTH result in elevations in glucocorticoid secretion. Thus it appears that the glucocorticoid responsivity to ACTH may be regulated at the postreceptor level (109).

Regulation. CRH is the most potent stimulator of ACTH/13- endorphin secretion from the anterior pituitary corticotrophs. ACTH response to stress, however, is partially regulated by peptides other than CRH. These include arginine vasopressin (AVP), oxytocin (OT), angiotensin II, vasoactive intestinal poly- peptide (VIP), serotonin, and, in the rat, E and NE (8, 148, 157). The physiologic role of these peptides in the regulation of ACTH secretion is still not clear. AVP secreted by parvocellular regions of the PVN into the hypophyseal portal circulation stim- ulates ACTH release synergistically with CRH (177). Glucocor- ticoids inhibit stimulated ACTH secretion both in vivo and in vitro (99). The inhibitory effects in vivo exerted at least at two levels, via inhibition of the secretion of hypothalamic CRH and direct suppression of ACTH release at the pituitary level (34).

Behavior. ACTH, along with vasopressin, is believed to en- hance attention, motivation, learning and memory retention (50,5 i). It appears that only a small part of the ACTH molecule, amino acids 4-7 of the N terminal, is needed to produce these effects (50). Besides these positive effects, ACTH also acts as an opi- ate antagonist, and competes with opiate-binding sites in the brain. Thus ACTH can counteract morphine analgesia and re- verse catatonic posture (immobility). ACTH administered ICV also appears to affect social behavior in the rat by reducing so- cial interaction and decreasing aggression. Novel or stressful stimuli induce grooming, which has been related to the endoge- nous release of ACTH (69).

The behavioral response to ICV ACTH is characterized by the "stretching yawning syndrome," in which rats display ex- cessive grooming behavior interrupted by bouts of stretching. In contrast to CRH, ACTH given ICV does not appear to increase activity or exploration (58,89). When injected peripherally, ACTH

reduces exploration in a novel environment without influencing locomotion (57). The increased activity produced by ACTH is about 100 times less potent than the changes produced by CRH.

Glucocorticoids

The adrenal gland consists of at least two anatomically and chemically distinct structures: an inner medullary area contain- ing catecholamine-producing chromaffin cells and an outer corti- cal region in which glucocorticoids and other steroids are synthesized. Glucocorticoids are released from the adrenal gland into the general circulation. A high proportion, almost 95%, of circulating cortisol circulates bound to an alpha globulin called "transcortin" or "corticosteroid-binding globulin" (CBG). The bound fraction of cortisol is considered physiologically inactive. The small free-fraction of cortisol in plasma represents the ac- tive fraction of the hormone which exerts negative feedback ef- fects on CRH and ACTH release (190).

Receptors. Glucocorticoid receptors are present throughout the brain, including in the CRH neurons of the hypothalamus (62). The actions of glucocorticoids on the central nervous sys- tem (CNS) are mediated by two separate receptor systems: glu- cocorticoid receptors type I and type II (155,173). Type I ("corticosterone receptors") receptors are found mainly in the neurons of the limbic structures, such as the hippocampus and septum (120). These receptors play a role in modulating the re- sponse to environmental and emotional stimuli, with consequent changes in behavior and HPA axis activity. Type I receptors have a high affinity for the primary glucocorticoid (cortisol/cor- ticosterone), and are similar to "mineralocorticoid receptors" of the kidney (155,173). In the limbic system, these receptors have a high specificity for corticosterone as an agonist, whereas the mineralocorticoid aldosterone appears to be a competitive antag- onist. The Type I receptors that are found in the circumventricu- lar organs function as mineralocorticoid receptors that respond to aldosterone and act to regulate sodium homeostasis, cardio- vascular control and salt appetite (173). With age, the hippo- campus loses approximately 50 percent of its glucocorticoid type I binding sites (172). ACTH appears to have atrophic effect on the CNS and reverses age-related decreases in type I receptor (156).

Type II glucocorticoid receptors are present at high concen- trations in the hypothalamus, particularly in the CRH neurons. Type II receptors also are found in the brain areas that contain POMC, such as the hippocampus, lateral septum, amygdala, and nucleus tractus solitarius (120). At these sites, it is likely that the receptors participate in the behavioral, neuroendocrine and autonomic responses to stress (173). During stress, the occu- pancy of type I receptor changes only minimally, whereas that of type II receptor changes considerably (155). Glucocorticoids exert negative feedback to terminate ACTH release in response to stress and have long-term effects on adaptive behaviors, pre- sumably via the type II receptors. Type II receptors diminish with age, but are not affected by ACTH per se (156, 171-173). Reduction of type II receptors is associated with decreases of the negative feedback action of glucocorticoids, which may re- sult in a more persistent elevation of circulating plasma cortico- steroid levels following stress (167, 169, 171-173). In addition, persistent elevations of circulating glucocorticoids render the neurons of the hippocampus vulnerable to toxic influences with consequent degeneration and death of the cells (i 10, 169-173).

Behavior. Glucocorticoids have two types of action in the CNS. The first is associated with perception and the coordina- tion of the circadian patterns of food intake and sleep. The sec- ond is a negative feedback effect on stress-activated neural

Page 6: Mechanisms of Stress

120 J O H N S O N , KAMILARIS , C H R O U S O S A N D G O L D

circuits and metabolic processes. Interestingly, the acute effects of glucocorticoids are euphorogenic, whereas chronically ele- vated glucocorticoids produce depression in a large number of subjects (27).

Autonomic System

As previously mentioned, the two principal components of the general adaptational response are the CRH and the locus ceruleus-norepinephrine, (LC-NE)/autonomic (sympathetic) ner- vous systems (71,72) (Fig. 3). The LC-NE/sympathetic systems are located in the brainstem (136). Activation of the LC-NE system leads to release of norepinephrine from a dense network of neurons throughout the brain that results in arousal, vigilance and increased anxiety. It has been generally accepted that the sympathetic division of the autonomic system is primarily asso- ciated with conferring an adaptive advantage during stressful sit- uations via its effectors, the sympathetic nerves and the adrenal medulla, located in the periphery. On the other hand, the para- sympathetic division of the autonomic nervous system, which is functionally linked to the sympathetic system, appears to pro- duce effects antithetical to those of the sympathetic nervous sys- tem. Thus its inhibition can produce effects analogous to those of sympathetic activation.

Cannon (37) was the first to note that a variety of stressors resulted in an increase in sympathetic nervous system activity and adrenal medulla output. During stress, E and NE are re- leased into the general circulation and the activity of enzymes that regulate catecholamines biosynthesis is stimulated. Today, it is generally accepted that the general sympathetic and sym- pathomedullary systems are critical elements in the integrated physiological response of an organism to a variety of stressors (8, 111, 118, 124). Central regulation of this response involves components of the central stress system in the cerebral cortex, limbic system, hypothalamus, and brainstem (60).

Acute stress results in secretion of E and NE from the adre- nal medulla and release of NE from the sympathetic nerve ter- minals (205). In contrast, chronic intermittent stress is associated with changes in the adrenal medulla, including increased activ- ity of enzymes involved in catecholamine biosynthesis, increased rates of catecholamine synthesis and elevated tissue concentra- tions of catecholamines (108). Chronic intermittent stressors ini- tially appear to affect catecholamine release during subsequent exposures, as a function of the familiarity of the stressor (101). Thus exposure to homotypic (familiar) stressors in a chronic in- termittent fashion results in a reduced sympathetic, sympath- omedullary response with time. In contrast, following chronic intermittent stress, exposure to an acute heterotypic (novel) stressor results in an enhanced sympathetic, sympathomedullary response (119,197).

There is growing body of studies that demonstrate that ani- mals subjected to inescapable, uncontrollable electric shocks show subsequent deficits in learning to terminate the noxious stimulus even when it is escapable. This phenomenon has been termed "learned helplessness," suggesting that the inability of the individual to control or terminate the stressor results from the initial learning of inescapability (181, 182, 213, 214). Ex- perimentally induced behavioral deficits of this type have been seen as a model of depression. Such a "learning deficit" was considered analogous to some of the mood/cognitive distur- bances observed in patients with depression, who frequently re- port feelings of helplessness or powerlessness to cope with stress. The weight of the available evidence suggests one mech- anism underlying the behavioral deficits observed in animals ex- posed to inescapable stress is a depletion of norepinephrine

NE

\\\ ii ss

\ I (~ituitary~ < \ ~ " E/NE

/ ' \\1 A C T H / IW \ G,oc/o iooi,s

Adrenal

FIG. 3. Schematic diagram of the functional interrelations between cor- ticotropin-releasing hormone (CRH) and locus ceruleus-norepinephrine (LC-NE) systems. These systems are the principal central biologic effec- tors of the generalized stress response. In the periphery, both systems act through glucocorticoids secreted from the adrenal cortex and the cat- echolamines, epinephrine and norepinephrine (E, NE) from the sympa- thetic and sympathomedullary systems (SS). Glucocorticoids are thought to restrain both systems of the stress response, in order to prevent the consequences of prolonged or excessive activation. (Solid lines represent stimulatory effects; broken lines represent inhibitory effects.)

and/or a decrease in tyrosine hydroxylase activity (213,214). The stress system also interacts with other CNS elements that

play a role in information processing, action initiation, as well as setting the emotional tone. The mesocortical dopamine sys- tem which innervates the prefrontal cortex, a brain region be- lieved to be involved in anticipatory phenomena and cognitive function, is activated by the LC-NE/sympathetic systems during stress. In addition, the mesolimbic dopamine system, a region closely linked to the nucleus accumbens, which has been impli- cated in motivational/reinforcement/reward phenomena, is also stimulated by the LC-NE/sympathetic systems during stress (48, 49, 136, 164). Noradrenergic neurons which originate in the brainstem LC-NE/sympathetic system activate the amygdala/hip- pocampus complex during stress (75).

Finally, the LC-NE/sympathetic systems seem to respond similarly to the CRH system to many of the same neurochemi- cal modulators. Serotonin and acetylcholine appear to be excita- tory to the sympathetic system (5, 9, 12, 36, 61, 80), while the gabaergic (9,61) and the opioid peptidergic (9, 23, 34, 80) neu- rotransmissions act to inhibit the LC-NE/sympathetic systems. In addition, the LC-NE/sympathetic systems seem to respond to autoregulation by an alpha-2 adrenergic-mediated inhibition.

CONSEQUENCES OF STRESS ON "'WELL-BEING": THE INTERACTION WITH OTHER PHYSIOLOGIC SYSTEMS

The systems responsible for reproduction, growth and immu- nity are directly linked to the stress system, and each is pro-

Page 7: Mechanisms of Stress

MECHANISMS OF STRESS 121

foundly influenced by the effectors of the stress response. Chronic stress can have physiological and behavioral consequences which affect the well-being of an individual. These include accelerated aging, reproductive suppression, retardation of growth, and im- munosuppression. In addition, some common psychiatric disor- ders, such as depression, panic anxiety and anorexia nervosa, may represent dysregulation of the systems responding to stress.

Stress and Aging

There is general support for the notion that chronic stress can accelerate aging (145) and that an aged animal has impaired ability to terminate the stress response (171). It has been sug- gested that cortisol hypersecretion in aged animals is due to de- generative changes within the aging brain and loss of sensitivity to glucocorticoid-mediated feedback inhibition. These effects are seen specifically in the hippocampal region of the limbic sys- tem, which has been associated with inhibition of the HPA axis (173). With age, the hippocampus loses approximately 50 per- cent of type I ("mineralocorticoid receptors") binding sites, as well as some of the type II ("glucocorticoid receptors") sites (172,173). In addition, the aged hippocampus demonstrates loss of neurons (174). It appears that cumulative exposure to in- creased glucocorticoid concentrations over the lifespan might mediate hippocampal neuron death (110). Chronic stress or pharmacologic doses of glucocorticoid treatment accelerate this process (170, 173, 174). This effect appears to be specific for the hippocampus, since other areas of the brain are spared (170). Some of the damaging effects of glucocorticoids appear to be mediated by type II glucocorticoid receptors. Hence, RU 486, a type II receptor antagonist, attenuates these toxic effects of glu- cocorticoids on the hippocampus (173).

Early neonatal experience may play a role in shaping indi- vidual patterns of stress susceptibility in later life. Meany et al. (125,126) have demonstrated that neonatal handling of rats in- creases hippocampal glucocordcoid receptor levels. This is con- cordant with their ability to shut off the pituitary-adrenal response to stress. One could postulate that this enhanced capability to terminate the secretion of adrenocortical stress hormones may attenuate the glucocorticoid-dependent degenerative changes in regions of the brain that participate in the restraint of the stress response, particularly the hippocampus (174).

Suppression of Reproductive Function

The state of threatened homeostasis produced by physical or emotional stress has long been recognized as a profound disrup- tive factor in reproductive function. Females under stress may demonstrate delayed puberty, lack of behavioral receptivity, failure of ovulation or embryo implantation, spontaneous abor- tion, or increased infant mortality (4, 10, 30, 117). Males may exhibit suppression of testosterone secretion, spermatogenesis and libido (2, 44, 152, 173). The severe suppression of repro- duction during stress appears to be caused by several hormones secreted during stress (such as CRH, ACTH, beta-endorphin and glucocorticoids) on hypothalamic-pituitary-gonadal (HPG) axis function (54, 81, 158, 161, 191, 211). Although the mecha- nism(s) of these effects on reproductive function are not fully elucidated, possible sites involved include: 1) a centrally medi- ated inhibition of gonadotropin-releasing hormone (GnRH) re- lease by CRH, opioids and glucocorticoids (158,161); 2) a glucocorticoid-mediated decrease in pituitary responsiveness to GnRH, resulting in decreased luteinizing hormone (LH) secre- tion (81); 3) direct gonadal effects of glucocorticoids with sub- sequent alterations in sex steroid output (13,39); and 4)

/ /

/ / /;G}

ACTH / LH,

nal ~ ~ ' G o n a d

Target Tissues

FSH

FIG. 4. Schematic representation of the functional interrelations between the hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-go- nadal (HPG) axes. CRH activates the pituitary-adrenal axis and GnRH the pituitary-gonadal axis, respectively. The activity of the GnRH- secreting neurons during stress is inhibited by CRH directly or via hypo- thalamic (arcuate nucleus) [3-endorphin (~-EP). Glucocorticoids suppress the activity of the reproductive axis at all levels, including the hypothal- amus, pituitary, gonads and target tissues of sex steroids. (Solid lines represent stimulatory effects; broken lines represent inhibitory effects.)

glucocorticoid-induced sex steroid target tissue resistance to go- nadal sex steroid (153) (Fig. 4).

Stress-induced decreases in circulating LH and sex steroid levels have been observed in both males and female rats and monkeys (24, 55, 139, 140). Abbott (1) demonstrated that, in marmosets, socially mediated suppression of reproduction is as- sociated with a significant reduction in LH release, resulting in decreased sexual behaviors in males and lack of behavioral re- ceptiveness and complete ovarian inactivity in females. Simi- larly, O'Byme (139) showed that a summation of the stressful effects of aggressive encounters and physical restraint produced a suppression of LH secretion during estradiol-induced LH surges in female marmosets. Although the site of action of socially in- duced inhibition of LH release was not addressed in this study, the fact that exogenous GnRH administration reversed the LH- decrease in these animals supports the hypothesis that a centrally mediated inhibition of GnRH secretion by hormones secreted during stress could be related with the decrease of LH secretion (1, 139, 140).

Previous studies have shown that disruptive effects of stress on reproductive function in female animals and women may be dependent on decreased gonadotropin secretion induced by ele- vated endogenous CRH and opioid levels (14, 66, 68, 138, 146, 161). From these studies, it appears that the most probable mechanism for reproductive inhibition involves CRH released during stress and acting within the brain to inhibit gonadotropin

Page 8: Mechanisms of Stress

122 J O H N S O N , KAMILARIS , C H R O U S O S A N D G O L D

secretion, directly and/or indirectly through opioids (13-endor- phin) (Fig. 3). This hypothesis is supported by several animal studies in which ICV administration of either CRH neutralizing antibodies or a specific CRH antagonist prevented CRH-induced suppression of the HPG axis (146). Recent observations in the rat have shown that the inhibitory effect of CRH on LH secre- tion is blocked by anti-13-endorphin serum and by a 13-endorphin antagonist, Blockage of [3-endorphin m and e receptors and dynorphin k-receptors reverses footshock-induced decreases in plasma LH levels, suggesting that multiple endogenous opiate systems participate in stress-induced inhibition of reproduction (146). The existence of a putative CRH-opioid interaction is supported by the ability of nalaxone to reverse the CRH-induced decrease in plasma LH levels in monkeys (68). Studies in amen- orrheic anorexic patients showed that decreases in gonadotropin secretion were associated with increased CRH and [3-endorphin concentrations in the CSF (71, 86, 97, 98).

Another mechanism by which activation of the HPA axis may influence reproductive function during stress is by direct effect of glucocorticoids at multiple levels of the HPG axis. Glucocor- ticoids suppress GnRH and gonadotropin secretion at both the hypothalamic and pituitary levels (11,96). At the gonadal level, glucocorticoids have a direct inhibitory effect on testicular Ley- dig cell function. Leydig cell sensitivity to LH and hCG is de- creased by glucocorticoids, probably due to glucocorticoid- induced reductions in testicular LH receptors (13,39). In cycling females, glucocorticoid treatment decreased estradiol concentra- tions and caused resistance of the uterus to estradiol. The latter has been associated with decreased estradiol receptors in the uterus (153). Stress cannot only influence the ability of the fe- male to conceive, but also can adversely affect the fecundity of each conception. In rodents, stress during pregnancy has been shown to result in smaller litter sizes (149,150). In sheep, there also is evidence that stress and hypercortisolism are associated with increased embryo loss following implantation and may pre- cipitate premature labor (16, 87, 114).

Growth Retardation

In humans, linear growth and final adult stature depend on multiple factors. These include genetic constitution (154), nutri- tion (I 15), systemic disease (32,78), hormones (90) and psycho- social environment (151). A putative association between stressful psychosocial environment and subsequent physical stature, intel- lectual and behavioral development has been described (I 29,130).

Psychosocial dwarfism, known also as abuse dwarfism, is a human condition thought to result from disruptions of social re- lationships in the neonatal environment; believed to be of partic- ular importance is the withdrawal of normal "ca re" by the caregiver, which may act as an emotional stressor (151). This syndrome is characterized by three primary reversible impair- ments: 1) delayed physical maturation, defined by both lower than normal body weight and height and delayed onset of pu- berty; 2) retardation of intellectual age, indicated by regressive or bizarre behaviors and retarded psychomotor development; and 3) delayed social maturation and slow psychosexual development (7,77).

Studies of the biochemical basis of growth abnormalities in children with psychosocial dwarfism have indicated that a "func- tional" growth hormone (GH) deficiency exists, which normal- izes rapidly with improved psychosocial environment (76,77). Fasting plasma GH levels and GH response to insulin-induced hypoglycemia are abnormally low in these children (77), and IGF-I levels are in the range characteristically found in hypopi- tuitarism (46). Although abnormalities in thyroid and adrenal

/I /\,, \ I I i I \ \ ~ I V \ \ \

I \ i t ,

\

I .<...I I /

Somatomedm i Adrenal ',. / ~ /

Targel Tissues

FIG. 5. Diagrammatic outline of the putative functional interrelations between the HPA axis and hypothalamic-pituitary-growth axis. Growth hormone (GH) secretion is under dual hypothalamic control of growth hormone-releasing hormone (GRH), which is stimulatory, and soma- tostatin, which is inhibitory. The activity of GRH neurons during stress appears to inhibited by CRH directly and/or via hypothalamic [3-endor- phin (13-EP) and somatostatin. GH exerts peripheral growth-promoting actions directly by stimulating peripheral somatomedin-C (IGF-I) pro- duction. Somatomedin-C appears to participate in a negative feedback regulation of GH secretion at both the pituitary and the hypothalamus. During chronic stress or hypercortisolemic states, glucocorticoids appear to suppress spontaneous secretion of GH at the pituitary level and prob- ably at the hypothalamus. In addition, glucocorticoids induce major tis- sue resistance to somatomedin-C and other growth factors. (Solid lines represent stimulatory effects; broken lines represent inhibitory effects.)

axis function have been reported in a few studies, they are not typical (106).

It is a commonly hypothesized view is that glucocorticoids and/or opioids secreted in response to chronic stress inhibit pitu- itary GH release at the pituitary level and decrease target tissue sensitivity to growth hormone, somatomedin-C or other growth factors (Fig. 5). The recent observation that the central adminis- tration of CRH decreases GH secretion in rats suggests a possi- ble role of endogenous CRH in the modulation of GH secretion during stress (142,159). Studies of the possible mechanisms in- volved in this effect suggest a central site of action, since spe- cific CRH antagonist administration abolished the inhibitory effect of a noxious stimuli on GH secretion (160). Additionally, the available data suggests that release of 13-endorphin and/or somatostatin by CRH may represent important mediators of this effect (15,160).

Studies in laboratory animals and humans have shown that disruption of the infant-caregiver relationship, as in maternal deprivation, rejection, or abuse, contributes to marked physio- logical and behavioral abnormalities in the offspring. For in- stance, it may impair overall skeletal growth and the ease with which social bonds are formed later in development (56,179). The mechanism(s) that mediate the linkage between early care

Page 9: Mechanisms of Stress

M E C H A N I S M S OF STRESS 123

or emotional state and the physical and behavioral manifestations during development are not clear.

In many species, physical contact and touch between young animals and their caregivers appears to be necessary for normal somatic growth (56,179). In humans, the etiology of psychoso- cial growth retardation has been related to unsatisfactory moth- er-infant relationships, often measured in terms of physical contact (76,77). In nonhuman primates, when physical contact is re- strained, even if visual, auditory and olfactory cues are main- mined, the behavioral development of the animal remains abnormal (82). Rat pups that are separated from their mother or placed with an anesthetized dam demonstrate decreases in tissue omi- thine decarboxylase activity and GH secretion and reduced tis- sue sensitivity to exogenous GH (179,180). These deficiencies could be reversed with tactile stimulation (179).

Alterations in bnmunocompetence During Stress

The principal effectors of the stress response exert multiple, complex effects on the immunologic apparatus. Conversely, many humeral components of the immune response exert effects on the central and peripheral components of the generalized stress response (25,212l (Fig. 6l. These interactions are suffi- ciently complex, as well as dose- and context-dependent, that no single statement can simply summarize these interactions, We shall try, therefore, to summarize only a few principles that have been generally established regarding alterations in immunocom- petence during stress.

Among the effectors of the stress response, the hypothala- mic-pituitary-adrenal axis seems to be the most influential. The HPA axis can influence immunologic function through a variety of mechanisms, including CRH-mediated actions on the release of somatostatin, with subsequent inhibition of growth hormone, CRH-mediated release of ACTH and beta-endorphin, and CRH- mediated pituitary-adrenal activation (25, 210, 212). Perhaps the best understood and most widely studied immunologic effects of the HPA axis are those mediated by glucocorticoids (17,133). At plasma levels generally achieved during either emotional or physical stress, including the stress of physical injury or inflam- mation, glucocorticoids generally exert immunosuppressive and antiinflammatory effects (17, 33, 133). These include inhibition of leukocyte traffic, interference with cell-mediated immunity, and enhancement of suppressor T-cell function (22, 67, 85, 123, 131, 133, 134). Moreover, there is a systematic decrease in the production of cytokines and interference with their functional ef- fects, as well as the induction of lymphopenia, thymic involu- tion, and loss of splenic and lymph node tissue mass (17, 123, 131 ). It should be noted that, in some instances, glucocorticoids enhance certain components of the immune response, including the function of specific differentiated clones of lymphocytes (194).

The CRH neuron of the HPA axis has recently been shown to participate in a negative feedback loop, producing pituitary- adrenal activation and concomitant glucocorticoid-mediated im- munosuppression in response to peripheral mediators of the inflammatory response, such as IFN, IL-I, IL-2, and PAF. This negative feedback loop is thought to appropriately restrain the immune and inflammatory response so that it will not overshoot in response to immune triggers (25, 133, 212). Interruption of this feedback loop by the administration of glucocorticoid antag- onists or an endogenous deficiency in the responsiveness of the CRH neuron to a variety of immune mediators results in suscep- tibility to inflammatory disease. Hence, Sternberg et al. have shown that the susceptibility of the LEW/N rat to a variety of inflammatory diseases reflects a deficient responsiveness of the

ACTH

CRH

~ ~ _ P~ituitar~y ~ ? CYet~ikitnes]° f "',.k,,.j/ ~ammation

Glucocorticoids J

Adrenal FIG. 6. A putaiive bidirectional regulatory feedback loop exists between the immune system and the CRH system. Cytokines and inflammatory mediators stimulate the HPA axis primarily by causing secretion of CRH. The HPA axis, in turn, inhibits the immune/inflammatory re- sponse primarily via increases in glucocorticoid secretion. This gluco- corticoid-mediated immunosuppression could prevent excessive inflammatory/immune responses during acute stress. (Solid lines repre- sent stimulatory effects; broken lines represent inhibitory effects.)

CRH neuron to a variety of stimuli, including cytokines (195,196). Conversely, histocompatible F/344N rats are resistant to inflam- matory disease because of a hyperresponsiveness of their CRH neurons to inflammatory mediators, but show the susceptibility of LEW/N rats to a variety of immunogenic stimuli if given small doses of the glucocorticoid antagonist RU 486 (195,196).

CRH has been shown not only to be essential for the activa- tion of the pituitary-adrenal axis, but also to possess a variety of behavioral effects when given ICV. Specifically, the central ad- ministration of CRH not only activates the pituitary-adrenal axis, but also sets into motion a variety of other physiological and behavioral responses that are adaptive during stressful situations. These include activation of the sympathetic nervous system, en- hancement of pathways mediating cautious avoidance and anxi- ety, and inhibition of pathways subserving vegetative functions such as feeding and reproduction. Although not yet definitively demonstrated, the response of the CRH neuron to the humeral mediators of immunity such as IL-I and PAF could not only serve to restrain the immune response to prevent it from over- shooting, but could also promote behavioral adaption of value during the stress of injury (17, 19, 25, 133, 194, 212, 216). As an example, IL-l-mediated CRH release, leading to cautious re- straint and increased anxiety, could serve to protect an animal from exposing itself to further injury during the stress of illness of prior injury. Such a dual role of a central element such as the CRH neuron would bring the discipline of neuroimmunology full circle by showing that the CNS could respond to peripherally mediated inflammatory signals by modulating both the immuno- logic and behavioral response of the organ to increase the likeli- hood of survival.

Page 10: Mechanisms of Stress

124 J O H N S O N , KAMILARIS , C H R O U S O S A N D G O L D

It has been suggested that patients with major depression show immunosuppression as a consequence of the hypercortiso- lism frequently associated with this disorder (33). However. the data have been conflicting. Although we cannot definitively ac- count for the discrepancies, they could reflect the fact that glu- cocorticoids are not always immunosuppressive, but may enhance certain components of the immune response (17,25). A second possibility is that not all depressive syndromes are associated with hypercortisolism (73,74). Indeed, the weight of available data suggests that, while the hyperaroused state of melancholia is frequently associated with hypercortisolism, the hyperphagia and hypersomnia of certain "atypical" depressions may be as- sociated with a subtle central adrenal insufficiency, and hence enhancement of certain immunologic responses. In this regard, our group has recently noted that the atypical depression often associated with the chronic fatigue syndrome, hypothyroidism, and seasonal affective disorder may be associated with a subtle deficiency in the responsiveness of the CRH neuron in associa- tion with subtle adrenal insufficiency.

In addition to the CRH system, the LC-NE system is thought to be the other major effector of the generalized stress response. CRH is itself thought to be a potent stimulus to the LC-NE sys- tem, although many neurotransmitters participate in the regula- tion and counterregulation of this important stress responsive system. NE is thought to have a variety of effects on the immu- nologic response, acting both as a blood-borne humeral media- tor and locally (194). As an example, the spleen and lymph nodes are replete with noradrenergic terminals and adrenergic receptors that modulate the functional activity of these lymphoid organs.

Psychiatric Disorders

It has been proposed that a critical factor in the pathophysi- ology of several psychiatric syndromes, such as major depres- sion, anorexia nervosa and panic anxiety, stems from an abnormality in the counterregulation of the generalized stress re- sponse, resulting in CRH and/or central catecholamine hyperse- cretion (192). In particular, it has been hypothesized that abnormalities in the positive regulation of or defects in counter- regulation of the central components of the adrenocortical and adrenergic system are responsible for these disorders (73,74).

The association between stress and depression stem from several observations: 1) individuals who are depression-prone have a higher than expected incidence of early noxious stress or maternal deprivation; 2) depressive episodes are often associated with major life changes (73,74); 3) acute stress-induced hor- monal and behavioral changes closely resemble the symptom complex of depression (71); and 4) hypercortisolism is a consis- tent feature of the classic form of major depression, melancholia (165).

The symptom complex of melancholia indicates that depres- sion need not be a state of pathological inactivation or suppres- sion, as the term depression implies, but rather a state of pathological hyperarousal (73,74). Indeed, from a clinical per- spective, one can construe melancholia as an organized state of anxiety attached principally to the self, resulting in a profound sense of worthlessness and hopelessness about the future, pros- pects of the worthless self. This anxiety about self and the fu- ture are associated with other signs of hyperarousal or activation of the generalized stress response that include enhanced vigi- lance, as well as inhibition of vegetative functions such as feed- ing, growth, reproduction, and sleep (73,74). Our group has advanced several lines of evidence suggesting that CRH may play a role in the clinical and biochemical manifestations of melancholia (40, 41, 70-74, 101L As an example, we have

shown that the plasma ACTH responses to exogenous CRH are blunted in melancholia and correlate negatively with the basal glucocorticoid levels. These data indicate that the pituitary corti- cotroph cell in major depression is appropriately restrained by high circulating glucocorticoids, and that hypercortisolism in major depression reflects a defect at or above the hypothalamus resulting in the hypersecretion of endogenous CRH (73,74). We also showed that a continuous infusion of CRH to volunteers re- produces the pattern and magnitude of hypercortisolism seen in major depression, while postdexamethasone cortisol levels in patients with major depression correlate positively with CSF CRH levels (73,74). Recently, we have advanced preliminary data that, in CSF sampled continuously for 30 hours, the levels of CRH measured hourly are consistently higher in depressed patients compared to controls (101).

This evidence of activation of one of the principal effectors of the stress response in major depression is also associated with evidence that the other major effector of the stress response, the LC-NE system, is also activated in melancholia. Hence, patients with melancholia show elevated levels of NE in CSF and plasma. while successful responses to antidepressant treatment, regard- less of the class, is associated with a significant fall in the plasma and CSF levels of the principal NE metabolite, MHPG (70-74, 101L In addition, tricyclic antidepressants which are the most effective agents in the treatment of melancholia reduce the LC firing rate, while we have recently shown that chronic but not acute imipramine treatment causes a significant decrease in the expression of TH mRNA in the LC. Taken together, these data suggest that the clinical and biochemical manifestations of major depression represent an activation of the major effectors of the generalized stress response that have escaped their usual counterregulatory elements to become pathologically established as a syndrome of sustained hyperarousal and organized anxiety.

The principal animal models of major depression support such a conclusion. These include the model of inescapable shock or learned helplessness, analogous to the clinical context in which many major depressions develop, namely sustained help- lessness or a burden of internal conflict or external stress that is inescapable (181, 182, 213, 214). Investigators such as Weiss have shown that inescapable shock produces a syndrome in the rat that is very analogous to that of melancholia, consisting of early moming awakening, anorexia, decreased libido and hypo- thalamic hypogonadism, shortened REM latency, and a behav- ioral phenotype compatible with the organism's being overwhelmed by stress (213,214). This syndrome responds to the classic treat- ments for melancholia, including tricyclic antidepressants and electroconvulsant treatment. It appears that the severity of the behavioral disturbances following inescapable shock correlate positively with the LC firing rate.

Although there has been a general emphasis on the role of the aminergic systems in stress and depression, there is a grow- ing body of evidence that suggests that supersensitivity of cen- tral muscarinic mechanisms may be involved in the path- ophysiology of depressive disorders (52, 53, 91-93). Stress re- sults in the rapid activation of the septohippocampal cholinergic system characterized by an increase in high-affinity uptake of choline and the release of acetylcholine (ACh). The latter has been shown to simultaneously induce alterations in behavioral, cardiovascular and neuroendocrine function characteristic of those observed during stress (52, 53, 143, 144). Hence, it has been hypothesized that stress-induced changes in affective, neuroen- docrine, sleep and heart rate profiles may reflect a central mus- carinic cholinergic component. In this regard, in vivo and in vitro data suggest that the muscarinic cholinergic agonist arec- oline stimulates the HPA axis and that this effect is mediated mainly by the release of endogenous CRH (36). In addition, it

Page 11: Mechanisms of Stress

M E C H A N I S M S O F S T R E S S 125

appears that the functional activity of ACh and the secretion of hypothalamic CRH are increased in affective disorders. The physiological relevance of a CRH-mediated cholinergic stimula- tion of the HPA axis is underlined by data showing the involve- ment of cholinergic neurotransmission in both the stress response and the pathophysiology of affective disorders (52, 53, I00, 116).

Our group has recently advanced data that some forms of major depression may not be associated with activation of stress- responsive neurotransmitter systems, but rather with their inacti- vation. This subtype, termed atypical depression and characterized by evidence o f pathological hypoarousal such as profound leth- argy, hypersomnia, and hyperphagia, occurs across the bound- aries o f medical illnesses such as Cushing 's disease, chronic fatigue syndrome, hypothyroidism, and seasonal affective disor- der (73,74). We have advanced several lines of evidence that these disorders are associated with a functional decrease in the responsiveness of the CRH neuron in these disorders s temming from a variety of pathophysiological mechanisms. In Cushing 's disease, we postulate that the long-standing peripherally medi-

ated (pituitary) hypercortisolism of this disorder causes a sus- tained suppression of the hypothalamic CRH neuron contributing to an atypical depression-like syndrome that is a frequent con- comitant of Cushing 's disease. In experimental models of hy- po thyro id i sm, we have shown that there is a decrease in hypothalamic CRH mRNA expression and content in association with a subtle central adrenal insufficiency (96). In chronic fa- tigue syndrome, we have shown that a significant decrease in 24-hour urinary free cortisol excretion is associated with several lines of indirect evidence indicative of a subtle central adrenal insufficiency, including robust cortisol responses to low-dose ACTH but blunted cortisol responses to high-dose ACTH, and ACTH responses to CRH analogous to those seen in postopera- tive Cushing 's disease patients thought to manifest a centrally mediated adrenal insufficiency (101). Taken together, these data suggest that a second major subtype of primary affective disor- der, namely atypical depression, reflects a pathological inactiva- tion rather than a pathological activation of at least one arousal- producing neurotransmitter system that is thought to be a principal effector o f the generalized stress response.

REFERENCES

1. Abbott, D. H. Behavioral and physiological suppression of fertility in subordinate marmoset monkeys. Am. J. Primatol. 6:169-186; 1984.

2. Abbott, D. H. Behaviorally mediated suppression of reproduction in female primates. J. Zool. (Lond.) 213:455-470; 1987.

3. Adams, F. The genuine works of Hippocrates. Baltimore: Williams and Wilkins Company; 1939.

4. Adams, M. R,; Kaplan, J. R.; Koritnik, D. R. Psychosocial influ- ences on ovarian endocrine and ovulatory function in Macaca fac- icularis. Physiol. Behav. 35:935-940; 1985.

5. Aghajanian, G. K. Mescaline and LSD facilitate the activation of the locus coeruleus neurons by peripheral stimuli. Brain Res. 186: 492-498; 1980.

6. Aguilera, G. Corticotropin releasing factor receptors: Characteriza- tion and actions in the anterior pituitary gland. In: Chrousos, G, P.; Loriaux, D. L.; Gold, P. W., eds. Mechanisms of physical and emotional stress. Advances in experimental medicine and biology, volume 245. New York: Plenum Press; 1988:83-105.

7. Alexlrod, J.; Reisine, T. D. Stress hormones: Their interaction and regulation. Science 224:45--459; 1984.

8. Annecillo, C.; Money, J.; Lobatto. C.; Intelligence (IQ) lost and regained: The psychoneuroendocrinology of failure to thrive, catch-up growth, the syndrome of abuse dwarfism, and Munchausen's syn- drome in proxy. In: Holmes, C. S., ed. Psychoneuroendocrinol- ogy: Brain, behavior and hormonal interactions. New York: Springer- Verlag; 1990:113-126.

9. Aston-Jones, G.; Foote, S, L.; Bloom, F. E. Anatomy and physi- ology of locus coeruleus neurons; functional implications. In: Zie- gler, M. G.; Lake. C. R., eds. Norepinephrine. Baltimore: Williams and Wilkins; 1984:92-116.

10. Bachman, G. A.; Kemmann, E. Prevalence of oligomenorrhea and amenorrhea in a college population. Am. J. Obstet. Gynecol. 1'44: 98-102; 1982.

11. Baldwin, D. M.; Sawyer, C. H. Effects of dexamethasone on LH release and ovulation in the cyclic rat. Endocrinology 94:1397- 1403; 1974.

12. Bagdy, G,; Calogero, A. E.; Murphy, D.; Szemeredy, K. Seroto- nin agonists cause parallel activation of the sympathoadrenomedul- lary system and the hypothalamo-pituitary-adrenocortical axis in conscious rats. Endocrinology 165:2664-2669; 1989.

13. Bambino, T. H.; Hsueh, A. J. W. Direct inhibitory effect of glu- cocorticoids upon testicular luteinizing hormone receptor and ste- roidogenesis in vivo and in vitro. Endocrinology 108:2142-2148; 1981.

14. Barbarino, A.; De Marinis, L.; Tofani, A.; Casa, S.; D'Amico, C.; Mancini, A.; Corsello, S. M.; Sciuto, R.; Barini, A, Corti- cotropin-releasing hormone inhibition of gonadotropin release and

the effect of opioid blockade. J. Clin. Endocrinol. Metab. 68:523- 528; 1989.

15. Bartolome, J. V.; Barotome, M. B.; Daltner, L. A.; Evans, C. J.; Barchas. J. D.; Kuhn, C. M.; Schanberg, S. M. Effects of [3-en- dorphin on ornithine decarboxylase in tissues of developing rats: A potential role for this endogenous neuropeptide in the modulation of tissue growth. Life Sci. 38:2355-2362; 1986.

16. Bassen, J. J,; Thorburn, G. D. Foetal plasma corticosteroids and the initiation of parturition in the sheep. J. Endocrinol. '44:285- 286; 1969.

17. Bateman. A.; Singh, A.; Kral, T.; Solomon, S. The immune-hy- pothalamic-pituitary-adrenal axis. Endocr. Rev. 10:92-112; 1989.

18. Bernard, C. Les phenomenes de la vie. Vol. 1. Paris: Librairie J-B Bailliere et Fils; 1878: 879.

19. Bernardini, R,; Calogero, A. E.; Ehrlich, Y. H.; Brucke, T.; Chrousos, G. P.; Gold. P. W. The alkyl-ether phospholipid plate- let-activating factor is a stimulator of the hypothalamic-pituitary- adrenal axis in the rat. Endocrinology 1:1067-1073; 1989.

20. Bernardini, R,; Chiarenza, A.; Calogero, A. E.; Gold, P. W.; Chrousos, G. P. Arachidonic acid metabolites modulate rat hypo- thalamic corticotropin releasing hormone secretion in vitro. Neu- roendocrinology 50:708-715; 1989.

21. Bernardini, R.; Kamilaris, T. C.; Calogero, A. E.; Johnson, E. O.; Gold, P. W,; Chrousos. G. P. Interactions between tumor ne- crosis factor-a, hypothalamic corticotropin-releasing hormone and adrenocorticotropin secretion in the rat. Endocrinology 126:2876-- 2881 ; 1990.

22. Besedovsky, J. O.; del Rey, A. E,; Sorkin, E. Neuroendocrine immunoregulation. In: Fabris, W.; Garaci, E.; Hadden, J., eds. Immunoregulation. London: Plenum Press; 1983.

23. Bird, S. J.; Kuhar, M. J. Iontophoretic application of opiates to the locus ceruleus. Brain Res. 122:523-33; 1977.

24. Blake, C. A. Effects of "stress" on pulsatile luteinizing hormone release in ovariectomized rats. Exp. Biol. Med. 148:813-815; 1975.

25, Blalock, J. E. A molecular basis for bidirectional communication between the immune and neuroendocrine systems. Physiol. Rev. 69:1-32: 1989.

26. Bloom, R. E.; Battenberg, E. L. F.; Rivier. J.; Vale, W. Corti- cotropin releasing factor (CRFI: Immunoreactive neurones and fi- bers in rat hypothalamus, Regul. Pept. 4:43-48; 1982.

27. Bohus, B.; De Kloet, E.; Veldhuis, H. Adrenal steroids and be- havioral adaptation: Relationships to brain corticoid receptors. In: Ganten, R.; Pfaff, D., eds. Progress in Neuroendocrinology. vol 2. Berlin: Springer-Verlag; 1983:1.

28. Britton, D. R,; Koob, G. F.; Rivier, J.; Vale, W. Intraventricular corticotropin-releasing factor enhances behavioral effects of nov-

Page 12: Mechanisms of Stress

126 J O H N S O N , K A M I L A R I S , C H R O U S O S A N D G O L D

elty. Life Sci. 31:363-367; 1982. 29. Britton, D. R.; Lee, G.; Dana, R.; Risch, S. C.; Koob, G. F. Ac-

tiviating and "anxiogenic" effects of corticotropin-releasing factor are not inhibited by blockade of the pituitary-adrenal system with dexamethasone. Life Sci. 39:1281-1286; 1986.

30. Broadhurst, P. L. Experiments in psychogenetics applications of biomedical genetics to the inheritance of behavior. In: Eysenck, H. J., ed. Experiments in personality psychogenetics and psychophar- macology, vol. 1. London: Routledge and Keegan Paul; 1960:1- 102.

31. Brown, M. R.; Fisher, L. A.; Spiess, J.; Rivier, J.; Rivier, C.; Vale, W. Corticotropin releasing factor: Actions on the sympa- thetic nervous system and metabolism. Endocrinology 111:928- 931: 1982.

32. Cacciari, E.; Salardi, S.; Lazzari, R., et al. Short stature and ce- liac disease: A relationship to consider even in patients with no gastrointestinal symptoms. J. Pediatr. 103:708-711; 1983.

33. Callabrese, J. R.; Kling, M. A.; Gold, P. W. Alterations in im- munocompetence during stress, bereavement, and depression: Fo- cus on neuroendocrine regulation. Am. J. Psychiatry 144:1123- 1134; 1987.

34. Calogero, A. E.; Gallucci, W. T.; Gold, P. W.; Chrousos, G. P. Multiple feedback regulatory loops upon rat hypothalamic corti- cotropin-releasing hormone secretion. J. Clin. Invest. 82:767-774; 1988.

35. Calogero, A. E.; Bernardini, R.; Gold, P. W.; Chrousos, G. P. Regulation of rat hypothalamic corticotropin-releasing hormone se- cretion in vitro: Potential clinical implications. In: Chrousos, G. P.; Loriaux, D. L.; Gold, P. W., eds. Mechanisms of physical and emotional stress, vol. 245. Advances in experimental medicine and biology. New York: Plenum Press; 1988:167-181.

36. Calogero, A. E.; Kamilaris. T. C.; Gomez, M. T.; Johnson, E. O.; Tartaglia, M. E.; Gold, P. W.; Chrousos, G. P. The muscar- inic cholinergic agonist arecoline stimulates the rat hypothalamic- pituitary-adrenal axis through a centrally-mediated corticotropin- releasing hormone-dependent mechanism. Endocrinology 125:2"44- 2453; 1989.

37. Cannon, W. B. The wisdom of the body. Physiol. Rev. 9:399- 431; 1929.

38. Carrol, B.; Eeinberg, M.; Greden, J.; Tarika, J.; Albala, A.; Has- kett, R.; James, N.; Kronfol, Z.; Lohr, N.; Steiner, M.; de Vigne, J.; Young, E. A specific laboratory test for the diagnosis of mel- ancholia. Arch. Gen. Psychiatry 38:15-22; 1981.

39. Charpenet, G.; Tache, Y.; Forest, M. G.: Haour, F.; Saez, J. M.; Bemier, M.; Ducharme, J. R.; Collu, R. Effects of chronic inter- mittent immobilization stress on rat testicular androgenic function. Endocrinology 109(4): 1254-1258; 198 I.

40. Chrousos, G. P.; Schulte, H. M.; Oldfield, E. H.; Loriaux, D. L.; Cutler, G. B.; Kellner, C. H.; Gold, P. W. Hypothalmic hormones in neuropsychiatric disease. Psychopharmacol. Bull. 19:416--421; 1983.

41. Chrousos, G. P.; Schuermeyer, T. H.; Doppman, J.: Oldfield, E. H.; Schulte, H. M.; Gold, P. W.; Loriaux, D. L. Clinical applica- tions of corticotropin-releasing factor. Ann, Intern. Med. 102:344- 358; 1985.

42. Chrousos, G. P.; Loriaux, L. D.; Gold, P. W. The concept of stress and its historical development. In: Chrousos, G. P.; Loriaux, L. D.; Gold, P. W., eds. Mechanisms of physical and emotional stress, vol. 245. Advances in experimental medicine and biology. New York: Plenum Press; 1988:3-7.

43. Chrousos, G. P.; Laue, L.; Nieman, L. K.; Kawai, S.; Udelsman, R. U.; Brandon, D. D.; Loriaux, D. L. Glucocorticoids and glu- cocorticoid antagonists: Lessons from RU 486. Kidney Int. 34(Suppl. 26):18-23; 1988.

44. Collu, R.; Gibb, W.; Ducharme, J. R. Effects of stress on the go- nadal function. J. Endocrinol. Invest. 7:529-537; 1984.

45. Cummings, S.; Elde, R.; Ell, J.; Lendall, A. Corticotropin-releas- ing factor immunoreactivity is widely distributed within the central nervous system of the rat: An immunohistochemical study. J. Neu- rosci. 3:1355-1368; 1983.

46. D'Ercole, A. J.; Underwood, L. E.; Van Wyk, J. J. Serum so- matomedin-C in hypopituitarism and in other disorders of growth. J. Pediatr. 90:375-381; 1977.

47. De Souza, E. B.; lnsel, T. R.; Perrin, M. H.; Rivier, J.; Vale, W. W.; Kuhar, M. J. Corticotropin-releasing factor receptors are widely distributed within the rat central nervous system: An autoradio- graphic study. J. Neurosci. 5:3189-3203; 1985.

48. Deutch, A. Y.; Clark, W. A.; Roth, R. H. Prefrontal cortical do- pamine depletion enhances the responsiveness of the mesolimbic dopamine neurons to stress. Brain Res. 521:311-315; 1990.

49. Deutch, A. Y.; Goldstein, M.; Roth, R. H. Activation of the lo- cus ceruleus by selective stimulation of the ventral tegmental area. Brain Res. 363:307-314: 1986.

50. De Wied, D. Pituitary-adrenal system hormones and behavior. In: Schmitt, F. G.; Worden, F. G., eds. The neurosciences. Third study program. Cambridge: MIT Press; 1974:653-666.

51. De Wied, D. Pituitary-adrenal system hormones and behavior. In: Selye, H., ed. Selye's guide to stress research, vol 1. New York: Van Nostrand Reinhold; 1980:252-279.

52. Dilsaver, S. C. Effects of stress on muscarinic mechanisms. Neu- rosci. Biobehav. Rev. 12:23-28; 1988.

53. Dilsaver S. C.; Alessi, N. E. Chronic inescapable footshock pro- duces cholinergic system supersensitvity. Biol. Psychiatry 22:914- 918; 1987.

54. Doerr, P.; Pirke, K. M. Cortisol-induced suppression of plasma testosterone in normal adult males. J. Clin. Endocrinol. Metab. 43:622-629; 1976.

55. Euker, J. S.; Meites, J.; Riegte, G. D. Effects of acute stress on serum LH and prolactin in intact, castrate and desamethasone- treated male rats. Endocrinology 96:85-92; 1975.

56. Field, T. Supplemental stimulation of preterm neonates. Early Hum. Dev. 4:301-314; 1980.

57. File, S. E. ACTH but not corticosterone impairs habituation and reduces exploration. Pharmacol. Biochem. Behav. 9:161-166; 1978.

58. File, S. E.; Clarke, A. Exploration motor activity after intraven- tricular ACTH, morphine and naloxone. Behav. Brain Res. 2:223- 227; 1980,

59. Fisher, L. A.; Rivier, J.; Rivier, C.; Speiss, J.; Vale, W.; Brown, M. R. Corticotropin releasing factor (CRF): Central effects on mean arterial pressure and heart rate in rats. Endocrinology 110: 2222-2224; 1982.

60. Foote, S. L.; Aston-Jones, G.; Bloom, F. E. Inpulse activity of locus coeruleus neurons in awake rats and monkeys is a function of sensory stimulation and arousal. Proc. Natl. Acad. Sci. USA 77:3033-3037; 1980.

61. Foote. S. L; Bloom, F. E.; Bloom, F. E. Nucleus locus ceruleus: New evidence for anatomical and physiological specificity. Phys- iol. Rev. 63:8'44-914; 1983.

62. Fuxe, K.; Wikstrom, A.; Okret, S.; Agnati, L.; Harfstrand, A.; Yu, Z.; Granholm, L.; Zoli, M.; Vale, W.; Gustafsson, J., et al. Mapping of glucocorticoid receptor immunoreactive neurons in the rat teland diencephalon using a monoclonal antibody against rat liver glucocorticoid receptor. Endocrinology 117:1803-1812; 1985.

63. Ganong, W. E. The stress response--a dynamic overview. Hosp. Pract. 93:155-171; 1988.

64. Gardiner, S. M.; Compton, A. M.; Bennett, T. Cardiovascular in- stability induced by intracerebroventricular administration of vaso- pressin or corticotrophin-releasing factor in conscious Long Evans and Brattleboro rats. J. Neuroendocrinol. 2:39--43; 1990.

65. Garrison, F. H. An introduction to the history of medicine. Phila- delphia: Saunders; 1929.

66. Gilbeau, P. M.; Smith, C. G. Naloxone reversal of stress induced reproductive effects in the male rhesus monkey. Neuropeptides 5:335-338; 1985.

67. Gillis, S.; Crabtree, G. R.; Smith, K. A. Glucocorticoid induced inhibition of T cell growth factor production I. The effect of mito- gen induced lymphocyte proliferation. J. lmmunol. 123:1624-1631 ; 1979.

68. Gindoff, P. R.; Ferin, M. Endogenous opioid peptides modulate the effect of corticotropin-releasing factor on gonadotropin release in the primate. Endocrinology 121:837-842; 1987.

69. Gipsen, W. H.; van der Poel, A.; van Wimersma Greidanus, T. B. Pituitary adrenal influences on behavior. Responses to test situ- ations with or without electric footshock. Physiol. Behav. 10:345- 350; 1973.

70. Gold, P. W.; Chrousos, G. P. Clinical studies with corticotropin

Page 13: Mechanisms of Stress

M E C H A N I S M S O F S T R E S S 127

releasing factor: Implications for the diagnosis and pathophysiol- ogy of depression, Cushing's disease, and adrenal insuffiency. Psychoneuroendocrinology 10:401-419; 1985.

71. Gold, P. W.; Loriaux, D. L.; Roy, A.; Kling, M. A.; Calabrese, J. R.; Kellner, C. H.; Nieman, L. K.; Post, R. M.; Pickar, D.; Gallucci, W.; Avgerinos, P.; Paul, S.; Oldfield, E. H.; Cutler, G. B.; Chrousos, G. P. Responses to corticotropin-releasing hormone in the hypercortisolism of depression and Cushing's disease. N. Engl. J. Med. 314:1329-1335; 1986.

72. Gold, P. W.; Gwirtsman, H.; Avgerinos, P. C.; Nieman, L. K.; Gallucci, W. T.; Kaye, W.; Jimerson, D.; Ebert, M.; Rittmaster, R.; Loriaux, D. L.; Chrousos, G. P. Abnormal hypothalamic-pitu- itary-adrenal function in anorexia nervosa: Pathophysiologic mech- anisms in underweight and weight-corrected patients. N. Engl. J. Med. 314:1335-1342; 1986.

73. Gold, P. W.; Goodwin, F. K.; Chrousos, G. P. Clinical and bio- chemical manifestations of depression: Relation to the neurobiol- ogy of stress. (first of two parts). N. Engl. J. Med. 319:348-353; 1988.

74. Gold, P. W.; Goodwin, F. K.; Chrousos, G. P. Clinical and bio- chemical manifestations of depression: Relation to the neurobiol- ogy of stress. (second of two parts). N. Engl. J. Med. 319:413- 420; 1988.

75. Gray, T. S. Amygdala: Role in autonomic and neuroendocrine re- sponses to stress. In: McCubbin, J. A.; Kaufman, P. G.; Nemer- off, C. B., eds. Stress, neuropeptides and systemic disease. New York: Academic Press; 1991:37-53.

76. Green, W. H.; Campbell, M.; David, R. Psychosocial dwarfism: A critical review of the evidence. J. Am. Acad. Clin. Child Ado- lesc. Psychiatry 23:39--48; 1984.

77. Green, W. H. A theoretical model for classical psychosocial dwarf- ism (psychosocially determined short stature). In: Holmes, C. S., ed. Psychoneuroendocrinology: Brain, behavior and hormonal in- teractions. New York: Springer-Verlag; 1990:92-112.

78. Groll, A.; Preece, M. A.; Candy, D. C. A., et al. Short stature as the primary manifestation of celiac disease. Lancet 2:1097-1105; 1980.

79. Guillemin, R.; Vargo, T.; Rossier, J.; Minick, S.; Ling, N.; Riv- ier, C.; Vale, W.; Bloom, F. Beta-endorphin and adrenocorticotro- pin are secreted concomitantly by the pituitary gland. Science 197: 1367-1368; 1977.

80. Guynet, P. G.; Aghajanian, G. K. Acetylcholine, substance P and met-enkephalin in the locus coeruleus: Pharmacological evidence for independent sites of action. Eur. J. Pharmacol. 53:319-328; 1979.

81. Hagino, N.; Watanabe, M.; Goldzieher, J. W. Inhibition by adrenocorticotrophin of gonadotrophin-induced ovulation in imma- ture female rats. Endocrinology 84:308-314; 1969.

82. Harlow, H. F.; Zimmerman, R. R. Affectational responses in the infant monkey. Science 130:421-432; 1959.

83. Harris, G. W. Neural control of the pituitary gland. Physiol. Rev. 28:134-179; 1948.

84. Hatfield, J. M.; Daikh, D. I.; Adelman, J. P.; Douglass, J.; Bond, C. T.; Allen, R. G. In situ hybridization detection of marked dif- ferences in pre-proopiomelanocortin messenger ribonucleic acid content of individual corticotropes and melanotropes. Endocrinol- ogy 124:1359-1364; 1989.

85. Haynes, B. F.; Fanci, A. S. The differential effect of in vivo hy- drocortisone on the kinetics of subpopulations of human peripheral blood thymus-derived lymphocytes. J. Clin. Invest. 61:703-707; 1978.

86. Hotta, M.; Shibasaki, T.; Masuda, A.; Imaki, T.; Demura, H.; Ling, N.; Shizume, K. The response of plasma adrenocorticotro- pin and cortisol to corticotropin releasing hormone (CRH) and cerebrospinal fluid immunoreactive CRH in anorexia nervosa pa- tients. J. Clin. Endocrinol. Metab. 62:319-324; 1986.

87. Howarth, B., Jr.; Hawk, H. W. Effect of hydrocortisone on em- bryonic survival in sheep. J. Anita. Sci. 27:117-121; 1968.

88. Ingle, D. J. Permissibility of hormone action. A review. Acta En- docrinol. 17:172-186; 1954.

89. Isaacson, R. L.; Green, E. T. The effects of ACThI_24 on loco- motion, exploration, rearing and grooming. Behav. Biol. 24:118- 122; 1978.

90. Isaksson, O. G. P.; Jansson, J. 0. ; Gansel, A. M. Growth hor- mone stimulates longitudinal bone growth directly. Science 216: 1237-1241; 1982.

91. Janowsky, D. S.; El-Yousef, M. K.; Davis, J. H.; Sereke, H. S. A cholinergic adrenergic hypothesis of mania and depression. Lan- cet 19:675-681; 1972.

92. Janowsky, D. S.; Risch, S. C. Acetylcholine hypothesis of stress modulation. Integ. Psychiatry 3:3-9; 1985.

93. Janowsky, D. S.; Risch, S. C. Cholinomimetic and anticholinergic drugs used to investigate a cholinergic hypothesis of affective dis- orders and stress. Drug Dev. Rev. 4:125-142; 1984.

94. Johnson. H. M.; Smith, E. M.; Torres. B. A.; Blalock, J. E. Reg- ulation of the in vitro antibody response by neuroendocrine hor- mones. Proc. Natl. Acad. Sci. USA 79:4171--4174; 1982.

95. Johnson, H. M.; Toress. B. A.; Smith, E. M.; Dion, L. D.; Bla- lock. J. E. Regulation of lymphokine (-,/-interferon) production by corticotropin. J. Immunol. 132:246-250; 1984.

96. Kamilaris, T. C.; DeBold, C. R.; Johnson, E. O.; Mamalaki, E.; Listwak, S. J.; Calogero, A. E.; Kalogeras, K. T.; Gold, P. W.; Orth, D. N. Effects of short and long duration hypothyroidism and hyperthyroidism on the plasma adrenocorticotropin and corticoste- rone responses to ovine corticotropin-releasing hormone in rats. Endocrinology 128:2567-2576; 1991.

97. Kaye, H.; Pickar, D.; Naber, D.; Ebert, M. Cerebrospinal fluid opioid activity in anorexia nervosa. Am. J. Psychiatry 139:643- 645; 1982.

98. Kaye, W.; Gwirtsman, H.; George, D.; Ebert, M.; Jimerson, D.; Tomai, T.; Chrousos, G.; Gold, P. Elevated cerebrospinal fluid levels of immunoreactive corticotropin releasing hormone in an- orexia nervosa: Relation to state of nutrition, adrenal function, and intensity of depression. J. Clin. Endocrinol. Metab. 64:203-208; 1987.

99. Keller-Wood, M.; Dallman, M. Corticosteroid inhibition of ACTH secretion. Endocr. Rev. 5:1-24; 1984.

100. Kennedy, S.; Thompson, R.; Stancer, H. C.; Roy, A.; Persad, E. Life events precipitating mania. Br. J. Psychiatry. 142:398-403; 1983.

101. Kling, M. A.; Roy, A.; Doran, A. R.; Calabrese, J. R.; Rubinow, D. R.; Whitfield, H. J.; Conrad, M.; Post, R. M.; Chrousos, G. P.; Gold, P. W. Cerebrospinal fluid immunoreactive corticotropin- releasing hormone and adrenocorticotropin secretion in Cushing's disease and major depresssion: Potential clinical implications. J Clin. Endocr. Metab. 72:260-271; 1991.

102. Koob, G. F.; Bloom, F. E. Corticotropin-releasing factor and be- havior. Fed. Proc. 44:259-263; 1985.

103. Kopin, I. J.; Eisenhofer, G.; Goldstein, D. Sympathoadrenal med- ullary system and stress. In: Chrousos, G. P.; Loriaux, D. L.; Gold, P. W., eds. Mechanisms of physical and emotional stress. vol. 245. Advances in experimental medicine and biology. New York: Plenum Press; 1986:11-23.

104. Kopin, I. J.; Eisenhofer, G.; Goldstein, D. Adrenergic response following recognition of stress. In: Breznits, S.; Zinder, O., eds. Molecular biology of stress. New York: Alan R. Liss; 1989:123- 132.

105. Krieger, D. Cushing's Syndrome. vol 22. Monographs in endocri- nology. Berlin: Springer-Verlag; 1982.

106. Krieger. D.; Good, M. H. Adrenocortical and thyroid function in the deprivation syndrome. Am. J. Dis. Child. 120:95-102; 1970.

107. Krieger, D. T.; Liotta, A. S.; Brownstein, M. J. Presence of cor- ticotropi in brain of normal and hypophysectomized rats. Proc. Natl. Acad. Sci. USA 74:648-652; 1977.

108. Kvetnansky, R.; Nemeth, S.; Vegas, M.; Oprsalova, Z.; Jurcovi- cova, J. Plasma catecholamines in rats during adaptation to in- termittent exposure to different stress. In: Usdin, E.; Kvetnansky, R.; Alexlrod, J., eds. Stress: The role of catecholamines and other neurotransmitters. New York: Gordon and Breach; 1984:537- 562.

109. Lamberts, S. W.; Zwiderwizk, J.; Uitterlinder, P.; Blizd, J.; Bruning, H.; De Jong, F. Characterization of adrenal autonomy in Cushing's syndrome: A comparison between in vivo and in vitro responsiveness of the adrenal gland. J. Clin. Endocrinol. Metab. 70:192-199; 1990.

110. Landfield, P.; Baskin, R.; Pitier, T. Brain-aging correlates: Retar-

Page 14: Mechanisms of Stress

128 J O H N S O N , K A M I L A R I S , C H R O U S O S A N D G O L D

dation by hormonal-pharmacological treatments. Science 214:581- 584; 1981.

111. Landsberg, L. Catecholamines and the sympathoadrenal system. In: Ingbar, S., ed. The year in endocrinology 1975-1976. New York: Plenum; 1976:177.

112. Levine, S. Psychobiologic consequences of disruption in mother- infant relationships. In: Krasnegor, N. A.; Blass, E. M.; Hofer, M. A.; Smotherman, W. P., ed. Perinatal development: A psycho- biological perspective. New York: Academic Press; 1987:359-376.

113. Levine, S.; Goldman. L.; Coover, G. D. Expectancy and the pitu- itary-adrenal system. In: Physiology, emotion and psychosomatic illness. CIBA Foundation symposium 8. Amsterdam: Elsevier/Ex- cerpta Medica; 1972:281-291.

114. Liggens, G. C.; Fairclough, R. J.; Grieves, S. A.; Kendall, J. W., et al. The mechanism of parturition in the ewe. Recent Progr. Horm. Res. 29:111-150; 1973.

115. Linder, B.; Cassorla, F. Short stature: Etiology, diagnosis, and treatment. JAMA 260:3171-3175; 1988.

116. Lloyd, C. Life events and depressive disorder: II. Events as pre- cipitating factors. Arch. Gen. Psychiatry. 37:541-548; 1980.

117. Loucks, A. B.; Mortola, J. F.; Girton, L.; Yen, S. C. Alterations in the hypothalamic-pituitary-ovarian and the hypothalamic-pitu- itary-adrenal axes in athletic women. J. Clin. Endocrinol. Metab. 68:402-411 ; 1989.

118. McCarty, R. Stress, behavior and the sympathetic-adrenal medul- lary system. In: Pohorecky, L. A.; Brick, J., ed. Stress and alco- hol use. New York: Elsevier Science Publishers; 1983:7.

119. McCarty, R.; Stone, E. A. Chronic stress and regulation of the sympathetic nervous system. In: Usdin, E.; Kvetnansky, R.; Alex- lrod, J., eds. Stress: The role of catecholamines and other neuro- transmitters. New York: Gordon and Breach; 1984:563-576.

120. McEwen, B. S.; Weiss, J.; Schwartz, L. Selective retention of corticosterone by limbic structures in rat brain. Nature 220:911- 912; 1968.

121. Makara, G. B. Mechanisms by which stressful stimuli activate the pituitary-adrenal system. Fed. Proc. 44:149-153; 1985.

122. Mains, R.; Eipper, E.; Ling, N. Common precursor to corticotro- pin and endorphins. Proc. Natl. Acad. Sci. USA 74:3014-3018; 1977.

123. Marsh, J. T.; Rasmussen, A. F. Response of adrenals, thymus, spleen and leukicytes to shuttle box and confinement stress. Proc. Soc. Exp. Biol. Med. 104:180-183; 1960.

124. Mason, J. W. A review of psychoendocrine research on the sym- patho-adrenal medullary system. Psychosom. Med. 30:576-607; 1968.

125. Meany, M.; Aitken, D. The effects of early, postnatal handling on hippocampal glucocorticoid receptor concentrations: Temporal pa- rameters. Brain Res. 22:30-304; 1985.

126. Meany, M.; Aitken, D.; Bodnoff, S.; Iny, L.; Tatarevicz, J.; Sa- polsky, R. Early postnatal handling alters glucocorticoid receptor concentrations in selected brain regions. Behav. Neurosci. 99:765- 770; 1985.

127. Meyer, J. Biochemical effects of corticosteroids on neural tissues. Physiol. Rev. 65:946-1020; 1985.

128. Millan, M. A.; Abous Samra, A. B.; Wynn, P. C.; Catt, K. J.; Aquilera, G. Receptors and actions of corticotropin releasing hor- mone in the primate pituitary gland. J. Clin. Endocrinol. Metab. 64:1036-1041; 1987.

129. Money, J. The syndrome of abuse dwarfism (psychosocial dwarf- ism or reversible hyposomatotropinism): Behavioral data and case report. Am. J. Dis. Child. 131:508-513; 1977.

130. Money, J.; Annecillo, C.; Kelley, J. F. Abuse-dwarfism syndrome: After rescue, statural and intellectual catchup growth correlate. J. Clin. Child Psychol. 12:279-283; 1983.

131. Monjan, A. A. Stress and immunologic competence: Studies in animals. In: Ader, R., ed. Psychoneuroendocrinology. New York: Academic Press; 1985:185.

132. Morely, J. E.; Levine, A. S. Corticotropin-releasing factor, groom- ing and ingestive behavior. Life Sci. 31:1459-1464; 1982.

133. Munck, A.; Guyre, P.; Holbrook, N. Physiological functions of glucocorticoids in stress and their relation to pharmacological ac- tions. Endocrin. Rev. 5:25-44; 1984.

134. Munck, A.; Guyre, P. M. Glucocorticoid physiology, pharmacol-

ogy and stress. In: Chrousos, G. P.; Loriaux, D. L.; Lipsett, M. B., eds. Steroid hormone resistance. New York: Plenum Press; 1986:81-96.

135. Naito, Y.; Fukata, J.; Tominaga, T.; Nakai, Y.; Tamai, S.; Mori, K.; Imura, H. Interleukin 6 stimulates the secretion of adrenocorti- cotropic hormone in conscious, freely moving rats. Biochem. Bio- phys. Res. Commun. 155:1459-1463; 1988.

136. Nauta, W. J. H.; Feirtag, M. Fundamental neuroanatomy. New York: W. H. Freeman; 1986.

137. Nikolarakis, K. E.; Almeida, O. S. X.; Sirinathsinghji, D. J. S.; Herz, A. Stimulation of hypothalamic beta-endorphin and dynor- phin release by corticotropin-releasing factor (in vitro). Brain Res. 399:152-155; 1986.

138. Nikolarakis, K. E.; Almeida, O. S. X.; Sirinathsinghji, D. J. S.; Herz, A. Concomitant changes in the in vitro and in vivo release of opioid peptides and luteinizing hormone-releasing hormone from the hypothalamus following blockade of receptors for corticotro- pin-releasing factor. Neuroendocrinology 47:545-550; 1988.

139. O'Byrne, K. T.; Lunn, S. F.; Dixson, A. F. Effects of acute stress and the patterns of LH secretion in the common marmoset (Cal- lithrixjacchus). J. Endocrinol. 118:259-264; 1988.

140. O'Byrne, K. T.; Lunn, S. F.; Dixson, A. F. Naloxone reversal of stress-induced suppression of LH release in the common marmo- set. Physiol. Behav. 45:1077-1080; 1989.

141. Olschowka, J. A.; O'Donohue, T. L.; Mueller, G. P.; Jacobowitz, D. M. The distribution of corticotropin releasing factor-like immu- noreactive neurons in rat brain. Peptides 3:995-1015; 1982.

142. Ono, N.; Lumpkin, M. D.; Samson, W. K.; McDonald, J. K.; McCann, S. M. Intrahypothalamic action of corticotrophin-releas- ing factor (CRF) to inhibit growth hormone and LH release in the rat. Life Sci. 35:1117-1123; 1984.

143. Overstreet, D. H.; Janowsky, D. S.; Gillin, J. C.; Shiromani, P. J.; Sutin, E. L. Stress induced immobility in rats with cholinergic supersensitivity. Biol. Psychiatry 21:657-664; 1986.

144. Overstreet, D. H.; Russel, R. W.; Crocker, A. D.; Janowsky, D. S.; Gillin, J. C. Genetic and pharmacological models of cholin- ergic supersensitivity. Experientia 44:465-472; 1988.

145. Pare, W. The effect of chronic environmental stress on premature aging in the rat. J. Gerontol. 20:78-84; 1965.

146. Petraglia, F.; Vale, W.; Rivier, C. Opioids act centrally to modu- late stress-induced decrease in luteinizing hormone in the rat. En- docrinology 119:2445-2450; 1986.

147. Petraglia, F.; Sutton, S.; Vale, W.; Plotsky, P. Corticotropin-re- leasing factor decreases plasma LH levels in female rats by inhib- iting gonadotropin-releasing hormone release into hypo-physial- portal circulation. Endocrinology 20:1083-1088; 1987.

148. Plotsky, P.; Bruhn, T.; Otto, S. Central modulation of immunore- active arginine vasopressin and oxytocin secretion into the hy- pophysial-portal circulation by CRF. Endocrinology 116:1639- 1641; 1985.

149. Pollard, I.; Dyer, S. L. Effect of stress administered during preg- nancy on the development of fetal testes and their subsequent function in the adult rat. J. Endocrinol. 107:241-245; 1985.

150. Pollard, I. Prenatal stress effects over two generations in rats. J. Endocrinol. 109:239-244; 1986.

151. Powell, G. F.; Brasel, J. A.; Blizzard, R. M. Emotional depriva- tion and growth retardation simulating idiopathic hypopituitarism. Clinical evaluation of the syndrome. N. Engl. J. Med. 276:1271- 1278; 1967.

152. Rabin, D.; Gold, P. W.; Margioris, A. N.; Chrousos, G. P. Stress and reproduction: Physiologic interactions between the stress and reproductive axes. In: Chrousos, G. P.; Loriaux, D. L.; Gold, P. W., eds. Mechanisms of physical and emotional stress, vol 245. Advances in experimental medicine and biology. New York: Pren- rice-Hall; 1988:377-387.

153. Rabin, D. S.; Johnson, E. O.; Brandon, D. D.; Liapi, C.; Chrou- sos, G. P. Glucocorticoids inhibit estradiol-mediated uterine growth: Possible role of the uterine estradiol receptor. Biol. Reprod. 42: 74-80; 1990.

154. Ranke, M. B.; Pfluger, H.; Rosendahl, W., et al. Turner syn- drome: Spontaneous growth in 150 cases and review of the litera- ture. Eur. J. Pediatr. 141:81-88; 1983.

155. Reul, J.; DeKloet, E. Two receptor systems for corticosterone in

Page 15: Mechanisms of Stress

M E C H A N I S M S O F S T R E S S 129

rat brain: Microdistribution and differential occupation. Endocrinol- ogy 177:2505-2511; 1985.

156. Rigter, J.; Veldhuis, H.; De Kloet, E. Spatial orientation and the hippocampal corticosterone recptor systems of old rats; Effect of ACTI-I4_ 9 analogue ORC2766. Brain Res. 309:393-398; 1984.

157. Rivier, C.; Vale, W. Modulation of stress-induced ACTH release by CRF, catecholamines, and vasopressin. Nature 305:325-327; 1983.

158. Rivier, C.; Vale, W. Influence of corticotropin-releasing factor on reproductive functions in the rat. Endocrinology 114:914-921; 1984.

159. Rivier, C.; Vale, W. Corticotropin-releasing factor (CRF) acts centrally to inhibit growth hormone secretion in the rat. Endocri- nology 114:2409-2411; 1984.

160. Rivier, C.; Vale, W. Involvement of corticotropin-releasing factor and somatostatin in stress-induced inhibition of growth hormone secretion in the rat. Endocrinology 117:2478-2482; 1985.

161. Rivier, C.; Rivier, J.; Vale, W. Stress-induced inhibition of repro- ductive functions: Role of endogenous corticotropin-releasing fac- tor. Science 231:607-609; 1986.

162. Rose, R. M. Endocrine response to stressful psychological events. Psychiatr. Clin. North Am. 3:251; 1980.

163. Rosenblum, L. A. Influences of environmental demand on mater- nal behavior and infant development. In: Krasnegor, N. A.; Blass, E. M.; Hofer, M. A.; Smotherman, W. P., eds. Perinatal develop- ment: A psychobiological perspective. New York: Academic Press; 1987:377-395.

164. Roth, R. H.; Tam, S. Y.; Lda, Y.; Yang, J. S.; Deutch, A. Y. Stress and the mesocorticolimbic dopamine systems. Ann. NY Acad. Sci. 537:138-147; 1988.

165. Sachet, R. J.; Hellman, C.; Roffwarg, H. P.; Halpern, F. S.; Fukushima, D. K.; Gallagher, T. F. Disrupted 24-hour patterns of cortisol secretion in psychotic depression. Arch. Gen. Psychiatry 28:19-24; 1973.

166. Saffran, M.; Schally, A. V.; Bentey, B. G. Stimulation of the re- lease of corticotropin from the adenohypophysis by a neurohy- pophyseal factor. Endocrinology 57:439~.44; 1955.

167. Sapolsky, R. Glucocorticoid toxicity in the hippocampus: Tempo- ral aspects of neuronal vulnerability. Brain Res. 359:300-305; 1985.

168. Sapolsky, R. M. Endocrine and behavioral correlates of drought in wild olive baboons (Papio anubis). Am. J. Primatol. 11:217-227; 1986.

169. Sapolsky, R.; McEwen, B. S. Down-regulation of neural cortico- sterone receptors by corticosterone and dexamethasone. Brain Res. 339:161-165; 1985.

170. Sapolsky, R.; Pulsinelli, W. Glucocorticoids potentiate ischemic injury to neurons: Therapeutic implications. Science 229:1397- 1399; 1985.

171. Sapolsky, R.; Krey, L.; McEwen, B. The adrenocortical stress-re- sponse in the aged male rat: Impairment of recovery from stress. Exp. Gerontol. 18:55-63; 1983.

172. Sapolsky, R.; Krey, L.; McEwen, B. Corticosterone receptors de- cline in a site-specific manner in the aged rat brain. Brain Res. 289:235-240; 1983.

173. Sapolsky, R.; Krey, L. C.; McEwen, B. S. The neuroendocrinol- ogy of stress and aging: The glucocorticoid cascade hypothesis. Endocr. Rev. 7:284-301; 1986.

174. Sapolsky, R.; Krey, L. C.; McEwen, B. S. Prolonged glucocorti- cold exposure reduces hippocampal neuron number: Implications for aging. J. Neurosci. 5:1222-1227; 1985.

175. Sapolsky, R.; Rivier, C.; Yamamoto, G.; Plotsky, P.; Vale, W. Interleukin 1 stimulates the secretion of hypothalamic corticotropin releasing factor. Science 238:522"--524; 1987.

176. Sawchenko, P. E.; Swanson, L. W.; Joseph, S. A. The distribu- tion and cells of origin of ACTH (1-39) stained varicosities in the paraventricular and supraoptic nuclei. Brain Res. 232:365-374; 1982.

177. Sawchenko, P.; Swanson, L.; Vale, W. Co-expression of CRF and vasopressin immunoreactivity in parvocellular neurosecretory neu- rons of the adrenalectomized rat. Proc. Natl. Acad. Sci. USA 81: 1883-1887; 1984.

178. Schally, A. V.; Chang, R. C.; Arimura, A.; Redding, T. W.; Fishback, J. B.; Vigh, S. High molecular weight peptide with cor-

ticotropin-releasing factor activity from porcine hypothalami. Proc. Natl. Acad. Sci. 78:5197-5201; 1981.

179. Schanberg, S. M.; Field, T. M. Maternal deprivation and supple- mental stimulation. In: Field, T. M.; McCabe, P. M.; Schneider- man, N., eds. Stress and coping across development. Hillsdale, NJ: Erlbaum; 1988:3-25.

180. Schanberg, S. M.; Kuhn, C. M. Maternal deprivation: An animal model of psychosocial dwarfism. In: Usdin, E.; Squrkes, T. L.; Youdim, M. B. H., eds. Enzymes and neurotransmitters in mental disease. New York: John Wiley and Sons; 1980:372-393.

181. Seligman, M. E. P. Reversal of performance deficits and percep- tual deficits in learned helplessness and depression. J. Abnorm. Psychol. 85:11-26; 1976.

182. Seligman, M. E. P.; Maler, S. F. Failure to escape traumatic shock. J. Exp. Psychol. 74:1-9; 1967.

183. Selye, H. A syndrome produced by diverse nocuous agents. Na- ture 138:32; 1936.

184. Selye, H. The general adaptation syndrome and the diseases of ad- aptation. J. Clin. Endocrinol. 6:117-173; 1946.

185. Selye, H. Stress. The physiology and pathophysiology of exposure to stress. Montreal: Acta; 1950.

186. Selye, H. Hormones and resistance. New York: Springer-Verlag; 1971.

187. Selye, H. The evolution of the stress concept. Am. Scient. 61:692- 699; 1973.

188. Seyle, H. Selye's guide to stress research, vol 1. New York: Van Nostrand Reinhold; 1980:v.

189. Shibihara, Y.; Morimoto, Y.; Furutani, Notake, M.; Takahashi, H.; Shimizu, S.; Horikawa, S.; Numa, S. Isolation and sequence analysis of the human corticotropin-releasing factor precursor gene. EMBO J. 2:775-779; 1983.

190. Siiteri, P. K.; Mural, J. T.; Hammond, G. L.; Nisker, J. A.; Ray- moure, W. J.; Kuhn, R. W. The serum transport of steroid hor- mones. Recent Prog. Horm. Res. 38:457-510; 1982.

191. Sirinathsinghji, D. J. S.; Rees, L. H.; Rivier, J.; Vale, W. Corti- cotropin-releasing factor is a potent inhibitor of sexual receptivity in the female rat. Nature 305:232-235; 1983.

192. Smith, M. A.; Kling, M. A.; Whitfield, H. J.; Brandt, H. A.; Demitrack, M. A.; Geracioti, T. D.; Chrousos, G. P; Gold, P. W. Corticotropin-releasing hormone: From endocrinology to psychobi- ology. Horm. Res. 31:66-71; 1989.

193. Snyder, D. S.; Unanue, E. R. Corticosteroids inhibit murine mac- rophage Ia expression and interleukin q production. J. Immunol. 129:1803-1865; 1982.

194. Steinberg, E. M. Monokines, lymphokines, and the brain. In: Cruse, J. M.; Lewis, R. E., eds. The year in immunology 1988: Vol. 5. Immunoregulatory cytokines and cell growth. Basel: Karger; 1988:205-217.

195. Steinberg, E. M.; Hill, J. M.; Chrousos, G. P.; Kamilaris, T.; Listwack, S. J.; Gold, P. W.; Wilder, R. Inflammatory mediator- induced hypothalamic-pituitary-adrenal axis activation is defective in streptococcal cell wall arthritis-susceptible Lewis rats. Proc. Natl. Acad. Sci. USA 86:2374.-2378; 1989.

196. Sternberg, E. M.; Young, W. S.; Bernardini, R.; Calogero, A. E.; Chrousos, G. P.; Gold, P. W.; Wilder, R. L. A central nervous system defect in biosynthesis of corticotropin-releasing hormone is associated with susceptibility to streptococcal cell wall-induced ar- thritis in Lewis rats. Proc. Natl. Acad. Sci. USA 86:4771--4775; 1989.

197. Stone, E. A.; McCarty, R. Tyrosine hydroxylase activity and cate- cholamine release. Neurosci. Biobehav. Rev. 7:29-34; 1983.

198. Stock, G.; Rupprecht, U.; Strumf, H.; Schlor, H. Cardiovascular changes during arousal elicited by stimulation of amygdala, hypo- thalamus and locus cernleus. J. Auton. Nerv. Syst. 3:503-510; 1981.

199. Suda, T.; Tomori, N.; Tozawa, F.; Demura, H.; Shizume, K.; Mouri, T.; Miura, Y.; Sasaro, N. lmmunoreactive corticotropin and corticotropin-releasing factor in human hypothalamus, adrenal, lung cancer and phenechromocytoma. J. Clin. Endocrinol. Metab. 58:919-924; 1984.

200. Suomi, S. Genetic and maternal contributions to individual differ- ences in rhesus monkey biobehavioral development. In: Krasnegor, N.; Blass, E.; Hofer, M.; Smotherman, W., eds. Perinatal devel-

Page 16: Mechanisms of Stress

130 J O H N S O N , K A M I L A R I S , C H R O U S O S A N D G O L D

opment: A psychobiological perspective. New York: Academic Press; 1987: 397--419.

201. Suomi, S. J.; Kraemer, G. W.; Baysinger, C. M.; Delizio, R. D. Inherited and experiential factors in anxious behavior displayed by rhesus monkeys. In: Klein, D. F.; Rabkin, J., eds. Anxiety: New research and changing concepts. New York: Raven; 1981:179-200.

202. Sutton, R. E.; Kooh, G. F.; Le Moal, M.; Rivier, J.; Vale, W. Corticotropin releasing factor (CRF) produces behavioural activa- tion in rats. Nature 297:331-333; 1982.

203. Szemeredi, K.; Bagdy, G.; Stull, R.; Calogero, A. E.; Kopin, I. J.; Goldstein, D. S. Sympathoadrenalmedullary inhibition by chronic glucocorticoid treatment in conscious rats. Endocrinology 123: 2585-2590; 1988.

204. Terman, G.; Shavit, Y.; Lewis, J.; Cannon, J.; Liebeskind, J. In- trinsic mechanisms of pain inhibition: Activation by stress. Science 226:1270-1277; 1984.

205. Tilders, F. J. H.; Berkenbosch, F. CRF and catecholamines; Their place in the central and peripheral regulation of the stress response. Acta Endocrinol. (Suppl. 1 (Copenh.) 276:63-75; 1986.

206. Udelsman, R.; Goldstein, D. S.; Loriaus, D. L.; Chrousos, G. P. Catecholamine-glucocorticoid interactions during surgical stress. J. Surg. Res. 43:539; 1987.

207. Vale, W.; Spiess, J.; Rivier, C.; Rivier, J. Characterization of a 41 residue ovine hypothalamic peptide that stimulates the secretion of corticotropin and 13-endorphin. Science 213:1394-1397; 1981.

208. Valentino, R. J.; Cha, C. I.; Foote, S. L. Anatomic and physio- logic evidence for innervation of noradrenergic locus coeruleus by neuronal corticotropin-releasing factor. Soc. Neurosci. Abstr. 12: 1003; 1986.

209. Valentino, R. J. CRH effects on central noradrenergic neurons: Relationship to stress. In: Chrousos, G. P.; Loriaux, D. L.; Gold, P. W., eds. Mechanisms of physical and emotional stress. Ad- vances in experimental medicine and biology, vol. 245. New York: Plenum Press; 1988:47-64.

210. Van Liessum, P. A.; Swinkels, L. M.; Pieters, G. F.; Ross, A. A.; Smals, A. G.; Benraad, T. J.; Kloppenborg, P. W. Lack of antibody formation during long-term subcutaneous treatment with the somatostatin analogue octreotide in acromegaly. Acta Endo- crinol. (Copenh.) 122:309-312; 1990.

211. Vreeburg, J. T. M.; De Greef, W. J.; Ooms, M. P.; Van Wouw, P.; Weber, R. F. A. Effects of adrenocorticotropin and corticoste- rone on the negative feedback action of testosterone in the adult male rat. Endocrinology 115(3):977-983; 1984.

212. Weigent, D. A.; Blalock, J. E. Interactions between the neuroen- docrine and immune systems: Common hormones and receptors. Immunol. Rev. 100:79-10g; 1987.

213. Weiss, J. M.; Stone, E. A.; Hanell, N. Coping behavior and brain norepinephrine levels in rats. J. Comp. Physiol. Psychol. 72:153- 160; 1970.

214. Weiss, J. M.; Goodman, P. A.; Losito, B. G.; Corrigan, S.; Chatty, J. M.; Baily, W. H. Behavioral depression produced by an uncontrollable stressor: Relationship to norepinephrine, dopa- mine and serotonin levels in various regions of rat brain. Brain Res. Rev. 3:167-205; 1981.

215. Winokur, G.; Pfohl, B.; Sherman, B. The relationships of histori- cally defined subtypes of depression to ACTH and cortisol levels in depression: Preliminary study. Biol. Psychiata-y 20:751-757; 1985.

216. Woloski, B. R. M. N. J.; Smith, E. M.; Mejer, W. J. III; Fuler, G. M.; Blalock, J. E. Corticotropin releasing activity of mono- kines. Science 230:1035-1037; 1985.

217. Wynn, P. C.; Jarqood, J. P.; Cart, K. J.; Aguilera, G. Regulation of corticotropin-releasing factor (CRF) receptors with the rat pitu- itary gland: Effect of adrenaiectomy on CRF receptors and corti- cotroph responses. Endocrinology 116:1653-1659; 1985.

218. Yates, E.; Marsh, D.; Maran, J. The adrenal cortex. In: Mount- castle, V., ed. Medical physiology, 14th ed. St. Louis: C. V. Mosby; 1980:1558-1601.