Vous êtes sur la page 1sur 10

Dermatologic Therapy, Vol. 21, 2008, 22–31 Copyright © Blackwell Publishing, Inc.

, 2008
Printed in the United States · All rights reserved
DERMATOLOGIC THERAPY
ISSN 1396-0296

Psychoneuroimmunology
Blackwell Publishing Inc

FRANCISCO TAUSK*, ILIA ELENKOV† & JAN MOYNIHAN‡


*Department of Dermatology, University of Rochester, Rochester, New York,
†Institute of Neurobiology and Molecular Medicine, Rome, and ‡Department
of Psychiatry, University of Rochester, New York

ABSTRACT: Psychoneuroimmunology (PNI) is a discipline that has evolved in the last 40 years to
study the relationship between immunity, the endocrine system, and the central and peripheral
nervous systems. In this manner, neurotransmitters, hormones, and neuropeptides have been found
to regulate immune cells, and these in turn are capable of communicating with nervous tissue
through the secretion of a wide variety of cytokines. Of critical importance is the effect of products of
the CNS and nerves on the maintenance of the delicate balance between cell-mediated (Th1) and
humoral (Th2) immune responses. A good example of how this concept operates in vivo becomes
evident when analyzing the effects of stressors. Chronic stress affects significantly the function of the
immune system as well as modifies the evolution of a variety of skin diseases, as psychosocial inter-
ventions have proved to be effective in their therapy.

KEYWORDS: dermatology, neuropeptides, psychoneuroimmunology, stress

Introduction: to warn the innate immune system of an impend-


Psychoneuroimmunology ing wound to be inflicted by a predator, and for
lymphocytes to become mobilized to the skin in
The study of mind–body interactions can be traced anticipation. As the brain functions as the control
back to the time of Hippocrates (c. 460 BC–360 BC), center for the entire body, it is entrusted with
although this early work appeared to have been maintaining constant communication with all other
focused more on body-to-brain regulation rather bodily systems, resulting in integrated function of
than on brain regulation of physical health (1). all systems. However, as our understanding of
The discipline of psychoneuroimmunology (PNI) immunology largely unfolded in the Petri dish,
can be argued to have formally begun its emer- immunologists had become quite convinced that
gence in the mid 1970s, with the seminal work of the cells of the immune system function often
Ader and Cohen (2) and the finding that changes autonomously, and are regulated only by each
in immune function could be behaviorally condi- other. Although it is indeed the case that immune
tioned, in a manner analogous to the classical responses can be elicited in vitro, and such stud-
conditioning studies of Pavlov and his canine ies have been critical for our understanding of
subjects. From a practical perspective, that the mechanisms of immune function, immunologists
central nervous system (CNS) might regulate were resistant to the consideration that immune
immune function, and further, that the immune responses in vivo take place in an environment
system might communicate with the CNS, seems that contains nerve fibers, neurotransmitters, and
quite logical and to have clear adaptive value. hormones. Thus, the early days of PNI were
For example, it would makes sense for the brain fraught with tension, as the hard evidence for
brain-behavior-immune system interactions was,
Address correspondence and reprint requests to: Francisco
Tausk, MD, Department of Dermatology, University of
initially, scant.
Rochester, 601 Elmwood Ave., BOX 697, Rochester, NY 14642, or At the present time, there is no longer much
email: Francisco_Tausk@URMC.Rochester.edu. question that the nervous and immune systems
“Adapted from Tausk et al., with permission from Gaspari A, are tightly connected. In addition to growing
Tyring S, Clinical Immunodermatology London: Springer; 2007 support for behavioral conditioning of a variety of
(In press)” types of immune responses, four additional important

22
Psychoneuroimmunology

lines of evidence have converged over time to in vitro, to alter some aspect of immunity (e.g.,
establish the connections as incontrovertible. First, (11,12)) Furthermore, not only do lymphoid cells
CNS (hypothalamic) interventions (including lesion- respond to neurochemicals via receptor-mediated
ing studies) alter immunologic reactivity, and the interactions, but T cells, in particular, have been
elicitation of an immune response alters CNS demonstrated to express either immunoreactive
activity. Second, there is sympathetic innervation protein or mRNA for hormones with an historical
of spleen and other lymphoid organs, including nervous system function, including CRH, AVP, and
the skin. Third, changes in hormone or transmitter prolactin (13,14).
levels produce changes in immune function, and Lymphoid cells, then, live and function in a
vice versa. Finally, lymphoid cells express receptors fluctuating sea of neuroendocrine substances,
for a variety of hormones and transmitters. Thus, with levels of many of these hormones changing
lymphoid cells are exposed to neurochemical diurnally, and as a function of either acute and/or
ligands, and express the receptors necessary to chronic stimuli. Not surprisingly, then, a review of
bind them. Although it is beyond the scope of this the stress literature clearly shows that stress effects
article, it is important to acknowledge that the on immune function are not unidirectional and
communication between brain and immune can also vary in magnitude. At the very least, three
system is bidirectional, with signaling back to the broad categories of variables interact with neuro-
brain via the vagus nerve (3), and via soluble sensory signals to ultimately affect changes in
cytokines, including interleukin (IL)-6, IL-1, and immune and/or infectious disease outcomes: the
tumor necrosis factor (TNF)-α. To date, the majority experimental subject (e.g., the age, sex, strain, and
of the PNI literature has focused on the role of previous history of the subject), the nature of the
major neural pathways activated by internal or stimulus itself (e.g., the intensity, duration, and
external neurosensory signals or stressors: the the individual’s perception of the stimulus), and
hypothalamo–pituitary adrenal (HPA) axis and the the immune response that is examined.
sympathetic nervous system (SNS). Chrousos (4,5), One of the best, and now classic, illustrations
Dunn (6), and others (7–9) have elegantly reviewed that stress effects on immune function are not
stressor-induced activation of these pathways. unidirectional comes from the work of Lysle and
Briefly, neurosensory signals are ultimately colleagues (15), using a tail-shock paradigm with
processed in the paraventricular nucleus (PVN) of rats. In this paradigm, a single shock session
the hypothalamus and in the locus ceruleus- (composed of 16 tail shocks delivered on a vari-
noradrenergic center. In response to stressors, the able basis over 60 minutes) was associated with a
hypothalamus secretes corticotropin-releasing very robust suppression of both splenic and
hormone (CRH) and arginine vasopressin (AVP). peripheral blood T-cell proliferative responses. Of
From the PVN, CRH-containing neurons have interest is that with three or five daily equivalent
efferent pathways to the median eminence, and sessions, the response of splenic T cells returned
projections to noradrenergic centers in the brain- to that of the control levels, whereas T-cell prolif-
stem and spinal cord. CRH release further activates erative responses in the peripheral blood remained
the HPA axis, leading to release of peptides from dramatically suppressed. Thus, the effects of stress
the pituitary produced by the differential cleavage differed as a function of both chronicity and the
of pro-opiomelanocortin, most notably adrenocorti- lymphoid organ examined.
cotropic hormone, enkephalins, and endorphins. Relevant to this article are studies that have
Adrenocorticotropic hormone induces downstream focused on PNI and the skin. As the principal
release of glucocorticoids from the adrenal cortex. barrier to environmental insults, the skin has spe-
CRH and noradrenergic neurons in the CNS inner- cialized immunological processes, and also func-
vate and stimulate each other. Activation of the tions as an important component “of the diffuse
noradrenergic pathways by CRH results in secre- brain (16).” An array of neuropeptides is localized
tion of norepinephrine (NE) by the peripheral in the skin, including substance P, calcitonin
SNS and release of NE and epinephrine (EPI) from gene-related peptide, vasoactive intestinal peptide,
the adrenal medulla. The activation of these two and nerve growth factor (NGF). These peptides have
neurochemical pathways and release of hormones been implicated in modulating immune function
and transmitters can have profound downstream in the skin, and appear to play roles in development
effects on immune function (reviewed in (10)) of skin diseases, particularly psoriasis (17), which
Arguably, every one of the hormones/transmitters will be discussed later in this article.
secreted by these nervous system regions has Nervous system–immune system interactions
been shown to have the potential, either in vivo or (as in blood and other secondary lymphoid organs)

23
Tausk et al.

are also not unidirectional in the skin. As an Stress and immunity


example, a decade ago, Dhabhar and colleagues
observed that the effects of acute versus chronic Stress hormones influence numerous physiologic
restraint stress of rats on delayed-type hypersen- processes; however, it is becoming clear that the
sitivity (DTH) responses appear to be in opposite way they regulate inflammatory diseases is through
directions (18). Following a single session of their effects on the balance between cell-mediated
restraint prior to challenge with 2,4-dinitro-1- and humoral immunity and on neurogenic inflam-
fluorobenzene (DNFB)(2–5 hours of restraint), DTH mation in peripheral tissues such as the skin (26)
responses in skin (as measured by ear swelling) (FIG. 1).
were enhanced, but following chronic (daily for ≥ 3 The hypothalamic–pituitary–adrenal (HPA) axis
weeks) restraint sessions, DTH responses were and the SNS represent the peripheral limbs of the
suppressed compared to control mice (19). The stress system (27,28), whose activation occurs within
investigators further showed that adrenally derived the CNS in response to distinct blood-borne, neu-
corticosterone and EPI may mediate these effects; rosensory, and limbic signals. The central compo-
that is, the enhanced DTH response was abrogated nents of the stress system are the CRH and locus
by adrenalectomy (20). Dhabhar proposed that acute ceruleus sympathetic neurons of the hypothala-
stress may induce “redeployment” of peripheral mus and brain stem, which regulate the peri-
blood lymphocytes to the skin and that this would pheral activities of the HPA axis and the SNS,
be an adaptive response during the “fight-flight” respectively (28,29). The stress-induced release of
response. hypothalamic CRH leads ultimately to systemic
Another noteworthy example of the interplay secretion of glucocorticoids (GCs) and catechola-
among psychologic factors, physiologic factors, mines (CAs) (mainly EPI and NE), which in turn
and processes within the skin comes from the influence immune responses. Immune challenges
work of Kiecolt-Glaser et al. Following the group’s such as infections with bacteria release bacterial
earlier studies indicating that examination stress lipopolysaccharides (LPS), which induce the nuclear
was associated with increased antibody produc- factor (NF)kB mediated secretion of IL-1 and IL-6,
tion to presumably reactivation of latent Epstein- which stimulate the hypothalamic stress response
Barr virus (21), and to a slower time to seroconvert (27,28,30).
following hepatitis B vaccinations (22) in normal, Immune responses are regulated by antigen-
healthy medical students, the investigators also presenting cells (APC), such as monocytes/mac-
found that time to heal a mucosal wound was rophages, dendritic cells, and other phagocytic cells
approximately 3 days slower in dental students at that are components of innate immunity, and by
the time of examination (23). the helper T-lymphocyte subclasses Th1, Th2, and
More recently, Kiecolt-Glaser and colleagues Treg that are components of adaptive immunity.
investigated the relationship between the quality Homeostasis within the immune system is largely
of marital relationships and the partners’ time to dependent on cytokines, the chemical messengers
heal a dermal blister wound. For those couples who between immune cells, which play crucial roles in
engage in hostile interactions, the median time to mediating inflammatory and immune responses.
wound healing was 2 days longer than for those Th1 cells primarily secrete interferon (IFN)-γ, IL-2,
couples who have non-hostile interactions (7 vs. 5 and TNF-α, which promote cell-mediated immu-
days) (24), 2005 2705/id}. A related study was nity; whereas Th2 cells secrete a different set of
published in older adults (mean age of 65), who, cytokines, primarily IL-4, IL-10, and IL-13, which
in general, heal wounds slower than young adults. enhance humoral immunity (31–33) (FIG. 1). Naive
In this study, participants who were randomized T cells (Th0) are precursors of Th1 and Th2 cells,
to an exercise intervention (3 days/weeks, 30 minutes and IL-12 (produced by APCs) is the major inducer
cardio work) healed a 3.5 mm dermal wound sig- of Th1 differentiation and, hence, cellular immu-
nificantly faster than participants in a nonexercise nity. Thus, IL-12 and IFN-γ inhibit Th2, whereas
group (29.2 vs. 40 days, on average) (25). Thus, the IL-4 and IL-10 inhibit Th1 cell activities. IL-4 and
process of wound healing is affected by stressful IL-10 promote humoral immunity by stimulating
life conditions, and can be improved through the growth and activation of mast cells and eosi-
intervention and/or changes in lifestyle. It remains nophils, the differentiation of B cells into antibody-
to be determined, however, through what specific secreting B cells, and immunoglobulin switching
mechanisms the nervous system, both central and to IgE. Importantly, these cytokines also inhibit
peripheral, can alter innate and cellular immunity macrophage activation, T-cell proliferation, and the
in the skin in response to a wound. production of proinflammatory cytokines (31–33).

24
Psychoneuroimmunology

FIG. 1. Effects of Stress on Immunity and Inflammation.


The information of the presence of a stressor is processed through the Hypothalamus resulting in the release of CRH,
ACTH, NE and eventually cortisol. The latter hormones mediate the differentiation of Th0 (naïve T Helper cells) towards the
Th2 humoral immune response to the detriment of the Th1 cell-mediated response. APC’s secrete cytokines that mediate Th1
differentiation, however the presence of bacterial products such as LPS that bind to Toll-like Receptors induce the production
of IL-1 and IL-6, which cross the blood-brain barrier and trigger the hypothalamic CRH-stress response. In this manner, a
blood borne stressor of infectious nature can activate the HPA axis. Th1 effects are mediated by the cytokines IL-12,18,2
and γInterferon and T cells and Macrophages. Th2 responses are mediated by IL-4,6,13 and B Cells, Eosinophils and Mast Cells.
CRH: Corticotropin releasing Hormone; NE: Norepinephrin; Th0: Naïve Helper cells; APC: Antigen Presenting Cell; LPS:
Lipopolysaccharide; HPA: Hypothalamic-Pituitary-Adrenal Axis.

25
Tausk et al.

Each subset of T cells (Th1 and Th2) is mutually duction of cortisol, elevated blood pressure, heart
exclusive. rate, and EPI levels (59,60).
Both GCs and CAs systemically mediate a Th2 Numerous psychosocial interventions aimed at
shift by up-regulating Th2-cytokine production the reduction of stress have proved to be success-
and also by suppressing APCs and Th1, cytokine ful in the treatment of psoriasis (61,62). For example,
synthesis (34). Thus, GCs and CAs suppress the a brief exposure to meditation tapes during pho-
production by APCs of IL-12, the main inducer totherapy sessions decreased the length of photo-
of Th1 responses (35–38). GCs also have a direct therapy; the subjects in the intervention group
effect on Th2 cells by up-regulating their IL-4, cleared their psoriasis significantly faster (by 50%)
IL-10, and IL-13 production (36,39). Because when compared to those that received phototherapy
catecholamine-binding β2-adrenoceptors are only only (63,64). Psoriasis patients improved significantly
present in Th1 cells (40), CAs do not directly affect during hypnosis sessions, where they received
Th2 cells. suggestions that they were being exposed to “what-
ever they believed ... would ameliorate their con-
dition.” This observation, in addition to the high
Stress and skin diseases rates of placebo responses in some clinical trials
for psoriasis drugs, may suggest that the placebo
Diseases of the skin more than any other organ effect could be harnessed and used in the phar-
appear to be influenced by emotional factors, and macotherapy of these patients (65,66).
most dermatologists encounter patients who report
a temporal relationship between disease flares and
Atopic dermatitis
stressful life events. Emotional stressors have been
linked to the development or evolution of a variety Atopic dermatitis (AD) exemplifies the relationship of
of cutaneous diseases including acne (41), vitiligo the delicate balance between genetic, environmental,
(42,43), alopecia areata (44), lichen planus (45), and psychosocial factors, and evolution of a
seborrheic dermatitis, herpes simplex infections skin disease. Patients with AD have a severe
(46), pemphigus (47), urticaria (48), psoriasis, and impairment in their quality of life resulting in
atopic eczema (26). significant emotional distress (67). This relationship
is bidirectional, as stressors are important contribu-
tors to the flares and exacerbation of the disease
Psoriasis
(68,69). Experimental stressors have been found to
Emotional stressors have been reported to precede impair the recovery of transepidermal water loss
the onset of psoriasis (49), as well as precipitate (70), thus playing a detrimental role in the skin
flares (50,51). Farber attempted to explain this barrier function. This in turn, creates increased
phenomenon when he encountered patients who susceptibility to cutaneous inoculation with envi-
suffered traumatic severance of sensory innerva- ronmental agents (e.g., allergens as dust mites,
tion; he observed that the plaques of psoriasis dander, bacteria, and viruses), which are potential
present in the areas innervated by the sectioned precipitants of atopic flares (71).
nerves resolved, and only reappeared when nerve Patients with AD have an inherited deficiency
fibers regenerated and the sensitivity returned. in the function of the hypothalamus, resulting in a
This observation highlighted the role played by blunted response of the HPA axis. When exposed
sensory cutaneous nerves, leading to the proposal to experimental stressors or to the injection of CRH,
that locally secreted neuropeptides contributed to they respond with a blunted production of cortisol,
the maintenance of psoriatic disease. Subsequently, which could help explain flares during taxing stres-
it was found that psoriatic plaques display increased sors (72–74). Additionally, these subjects display an
nerve fiber density and altered content of neu- up-regulation of glucocorticoid receptors on periph-
ropeptides such as calcitonin gene-related peptide, eral leukocytes (74), making them hyperreactive
SP, vasoactive intestinal peptide, and NGF (52–55). to steroid stimulation. Thus, in spite of a blunted
High expression of NGF mediates T-cell and kera- HPA axis response to stress, effector cells that are
tinocyte proliferation, mast cell migration, degranu- exquisitely sensitive to systemic glucocorticoid
lation, and memory T-cell chemotaxis, which are release may respond in a hyperreactive fashion to
all hallmarks of psoriasis (52,56–58). Psoriasis patients stress-induced cortisol elevations, accentuating the
who reported stress-related exacerbations had an cytokine shift from Th1 to the Th2 immune response.
inherited dysfunction in the HPA axis, responding Stress-induced local and systemic secretion of
to experimental stressors with a decreased pro- EPI or its metabolites may also play a role in the

26
Psychoneuroimmunology

worsening of AD, as these patients have increased suppressing lymphocyte proliferation (107) and
mononuclear cell activation of intracellular type 4 natural killer cell activity (108,109).
phosphodiesterases (75). This may result from We recently reported a model of stress-induced
elevated levels of NE (76) found in these individuals, carcinogenicity (110), where UV-treated mice exposed
leading to the secretion of IL-13 and IL-4 (77), and repeatedly to the presence of a predator scent
Th2 differentiation. Stressors shift the immune developed squamous cell carcinomas significantly
response, impairing TH1 and favoring the Th2 earlier (week 8) compared to nonstressed controls
humoral immune response (78–80) and a redistri- (week 21), an observation that was subsequently
bution of lymphocytes and eosinophils (81,82). confirmed by others (111).
Pharmacologic doses of corticosteroids are also Stress reduction interventions have been shown
able to suppress the production of IL-4, which is to significantly prolong the survival of individuals
an agent of differentiation toward Th2 cells (79,83– with metastatic neoplasms (112). For example,
86), explaining the beneficial effect of systemic or patients with metastatic melanoma were found
topical application of these agents. to benefit from limited sessions of psychosocial
interventions, thereby significantly increasing their
6 years survival rate (113). One possible explana-
Urticaria
tion for the benefit of these interventions could
The relationship between stress and some forms be the modulatory effect of stress and stress
of urticaria is supported by numerous anecdotal reduction on NK cell function, presumed to represent
observations. Patients suffering from adrenergic one of the first innate lines of immune defense
urticaria report that their symptoms invariably against foreign (including cancerous) cells (114).
follow acute stressful events. The finding that stress Another example is relaxation training, which
mediates the degranulation of mast cells via CRH significantly increased older adults’ cytotoxic function
and neuropeptides (87), and the up-regulation of (115), even though chronic stress has been shown
mast cell CRH receptors (88) supports its putative to suppress NK activity (116,117).
role in the pathogenesis of urticaria.

Infections Conclusions
Numerous studies have demonstrated the delete- Psychoneuroimmunology is an evolving area of
rious effects of stressors on the evolution of science that will help us understand the relation-
infections (89), including experimental bacterial ship between the mind and the body. The past 30
infections of the skin (90,91) and recurrent herpetic years of research in the field of PNI have validated
infection precipitated by psychiatric illness, life the close relationship between the CNS and the
events, and disgust (92). Stress appears to signifi- immune system. There is a growing body of evi-
cantly modulate the evolution of human infection dence to support the effect of the psyche – stress,
with herpes simplex virus (92). Experimental in particular – on immune responses and a multi-
restraint stress correlates with the reactivation of tude of skin conditions. The effects of stress on
latent herpes simplex virus infection in the dorsal shifting the immune response are not completely
root ganglion neuron of rats. Human studies show understood; however, research has shown that
that persistent, but not single or acute, stressors stress modifies the delicate balance between health
are associated with the frequency of recurrences, and disease. Just as interesting are the numerous
which is in agreement with findings that chronic, studies demonstrating that a nonpharmacologic
but not acute stress, correlates with the develop- approach can ameliorate certain dermatologic dis-
ment of experimental viral infections in normal eases. Seeking alternative interventions can only
volunteers (46,93,94). Furthermore, psychosocial enhance our ability to treat patients.
interventions have been shown to decrease the
frequency of recurrences of herpes simplex infec-
tions (95). References
1. Medicine NLo. Emotions and Disease: an exhibition at
Cancer the National Library of Medicine. Friends of the National
Library of Medicine 1997.
Studies have suggested that natural or experimental 2. Ader R, Cohen N. Behaviorally conditioned immunosup-
stressors can modulate the evolution of malignan- pression. Psychosom Med 1975: 37: 333–340.
cies in humans as well as other animals (96–106), 3. Watkins LR, Maier SF, Goehler LE. Cytokine-to-brain

27
Tausk et al.

communication: a review and analysis of alternative mecha- 24. Kiecolt-Glaser JK, Loving TJ, Stowell JR, et al. Hostile mari-
nisms. Life Sci 1995: 57: 1011–1026. tal interactions, proinflammatory cytokine production,
4. Chrousos GP. Stressors, stress, and neuroendocrine inte- and wound healing. Arch Gen Psychiatry 2005: 62: 1377–
gration of the adaptive response. The 1997 Hans Selye 1384.
Memorial Lecture. Ann N Y Acad Sci 1998: 851: 311–335. 25. Emery CF, Kiecolt-Glaser JK, Glaser R, Malarkey WB, Frid DJ.
5. Charmandari E, Tsigos C, Chrousos G. Endocrinology Exercise accelerates wound healing among healthy older
of the stress response. Annu Rev Physiol 2005: 67: 259– adults: a preliminary investigation. J Gerontol 2005: 60:
284. 1432–1436.
6. Dunn A. Psychoneuroimmunology, stress and infection. 26. Sandoz A, Kusnir D, Koenig T, Tausk F Psychocutaneous
In: Friedman K, ed. Psychoneuroimmunology, stress, infec- medicine. In: Fitzpatrick’s dermatology in general medi-
tion. Boca Raton, FL: CRC, 1995: 25–45. cine. New York: Mc Graw-Hill, 2007.
7. Sternberg EM. Overview of the conference and the field. 27. Chrousos GP. The hypothalamic-pituitary-adrenal axis
Ann N Y Acad Sci 1998: 840: 1– 8. and immune-mediated inflammation. N Engl J Med 1995:
8. McEwen BS. Protection and damage from acute and 332: 1351–1362.
chronic stress: allostasis and allostatic overload and rele- 28. Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES. The sympa-
vance to the pathophysiology of psychiatric disorders. thetic nerve – an integrative interface between two super-
Ann N Y Acad Sci 2004: 1032: 1–7. systems: the brain and the immune system. Pharmacol
9. Korte SM, Koolhaas JM, Wingfield JC, McEwen BS. The Rev 2000: 52: 595–638.
Darwinian concept of stress: benefits of allostasis and 29. Besedovsky HO, del Rey AE, Sorkin E. What do the
costs of allostatic load and the trade-offs in health and immune system and the brain know about each other.
disease. Neurosci Biobehav Rev 2005: 29: 3–38. Immunol Today 1983: 4: 342–346.
10. Moynihan JA, Stevens SY. Mechanisms of stress-induced 30. Besedovsky H, del Rey A, Sorkin E, Dinarello CA. Immu-
modulation of immunity in animals. In: Ader R, Felten noregulatory feedback between interleukin-1 and gluco-
DL, Cohen N, eds. Psychoneuroimmunology. New York: corticoid hormones. Science 1986: 233: 652–654.
Academic Press, 2001: 227–249. 31. Fearon DT, Locksley RM. The instructive role of innate
11. Levite M. Neuropeptides, by direct interaction with T immunity in the acquired immune response. Science
cells, induce cytokine secretion and break the commit- 1996: 272: 50–53.
ment to a distinct T helper phenotype. Proc Natl Acad Sci 32. Mosmann TR, Sad S. The expanding universe of T-cell
USA 1998: 95: 12544–12549. subsets: Th1, Th2 and more. Immunol Today 1996: 17:
12. Khansari DN, Murgo AJ, Faith RE. Effects of stress on the 138–146.
immune system. Immunol Today 1990: 11: 170–175. 33. Trinchieri G. Interleukin-12 and the regulation of innate
13. Baker C, Richards LJ, Dayan CM, Jessop DS. Corticotro- resistance and adaptive immunity. Nature Rev 2003: 3:
pin-releasing hormone immunoreactivity in human T 133–146.
and B cells and macrophages: colocalization with arginine 34. Elenkov IJ, Chrousos GP. Stress Hormones, Th1/Th2
vasopressin. J Neuroendocrinol 2003: 15: 1070–1074. patterns, pro/anti-inflammatory cytokines and suscep-
14. Gerlo S, Verdood P, Hooghe-Peters EL, Kooijman R. Mod- tibility to disease. Trends Endocrinol Metab 1999: 10:
ulation of prolactin expression in human T lymphocytes 359–368.
by cytokines. J Neuroimmunol 2005: 162: 190–193. 35. Elenkov IJ, Papanicolaou DA, Wilder RL, Chrousos GP.
15. Lysle DT, Lyte M, Fowler H, Rabin BS. Shock-induced Modulatory effects of glucocorticoids and catechola-
modulation of lymphocyte reactivity: suppression, habit- mines on human interleukin-12 and interleukin-10 pro-
uation, and recovery. Life Sci 1987: 41: 1805–1814. duction: clinical implications. Proc Assoc Am Physicians
16. Urpe M, Buggiani G, Lotti T. Stress and psychoneuroim- 1996: 108: 374–381.
munologic factors in dermatology. Dermatol Clin 2005: 23: 36. Blotta MH, DeKruyff RH, Umetsu DT. Corticosteroids
609 –617. inhibit IL-12 production in human monocytes and
17. Saraceno R, Kleyn CE, Terenghi G, Griffiths CE. The role enhance their capacity to induce IL-4 synthesis in CD4+
of neuropeptides in psoriasis. Br J Dermatol 2006: 155: lymphocytes. J Immunol 1997: 158: 5589–5595.
876 –882. 37. Hasko G, Szabo C, Nemeth ZH, Salzman AL, Vizi ES. Stim-
18. Dhabhar FS, McEwen BS. Acute stress enhances while ulation of beta-adrenoceptors inhibits endotoxin-induced
chronic stress suppresses cell-mediated immunity in IL-12 production in normal and IL-10 deficient mice. J
vivo: a potential role for leukocyte trafficking. Brain Behav Neuroimmunol 1998: 88: 57–61.
Immun 1997: 11: 286–306. 38. Panina-Bordignon P, Mazzeo D, Lucia PD, et al. Beta2-
19. Dhabhar FS. Stress-induced enhancement of cell-mediated agonists prevent Th1 development by selective inhibition
immunity. Ann N Y Acad Sci 1998: 840: 359–372. of interleukin 12. J Clin Invest 1997: 100: 1513–1519.
20. Dhabhar FS, McEwen BS. Enhancing versus suppressive 39. Ramirez F, Fowell DJ, Puklavec M, Simmonds S,
effects of stress hormones on skin immune function. Proc Mason D. Glucocorticoids promote a TH2 cytokine
Natl Acad Sci USA 1999: 96: 1059–1064. response by CD4+ T cells in vitro. J Immunol 1996: 156:
21. Glaser R, Kiecolt-Glaser JK, Speicher CE, Holliday JE. 2406–2412.
Stress, loneliness, and changes in herpesvirus latency. J 40. Sanders VM, Baker RA, Ramer-Quinn DS, Kasprowicz DJ,
Behav Med 1985: 8: 249–260. Fuchs BA, Street NE. Differential expression of the beta2-
22. Glaser R, Kiecolt-Glaser JK, Bonneau RH, Malarkey W, adrenergic receptor by Th1 and Th2 clones: implications
Kennedy S, Hughes J. Stress-induced modulation of the for cytokine production and B cell help. J Immunol 1997:
immune response to recombinant hepatitis B vaccine. 158: 4200–4210.
Psychosom Med 1992: 54: 22–29. 41. Chiu A, Chon SY, Kimball AB. The response of skin dis-
23. Marucha PT, Kiecolt-Glaser JK, Favagehi M. Mucosal ease to stress: changes in the severity of acne vulgaris as
wound healing is impaired by examination stress. Psy- affected by examination stress. Arch Dermatol 2003: 139:
chosom Med 1998: 60: 362–365. 897–900.

28
Psychoneuroimmunology

42. Papadopoulos L, Bor R, Legg C, Hawk JL. Impact of life 63. Benhard JD, Kristeller J, Kabat-Zinn J. Effectiveness of
events on the onset of vitiligo in adults: preliminary evi- relaxation and visualization techniques as an adjunct to
dence for a psychological dimension in aetiology. Clin Exp phototherapy and photochemotherapy of psoriasis. J Am
Dermatol 1998: 23: 243–248. Acad Dermatol 1988: 19: 572–574.
43. Barisic-Drusko V, Rucevic I. Trigger factors in childhood 64. Kabat-Zinn J, Wheeler E, Light T, et al. Influence of a
psoriasis and vitiligo. Coll Antropol 2004: 28: 277–285. mindfulness meditation-based stress reduction interven-
44. Gulec AT, Tanriverdi N, Duru C, Saray Y, Akcali C. The tion on rates of skin clearing in patients with moderate
role of psychological factors in alopecia areata and the to severe psoriasis undergoing phototherapy (UVB) and
impact of the disease on the quality of life. Int J Dermatol photochemotherapy (PUVA). Psychosom Med 1998: 60:
2004: 43: 352–356. 625–632.
45. Chaudhary S. Psychosocial stressors in oral lichen planus. 65. Ader R. The role of conditioning in pharmacotherapy.
Aust Dent J 2004: 49: 192–195. Cambridge: Harvard University Press, 1997.
46. Cohen F, Kemeny ME, Kearney KA, Zegans LS, Neuhaus 66. Ader R. Classical conditioning in the treatment of psoria-
JM, Conant MA. Persistent stress as a predictor of genital sis. Cutis 2000: 66: 370–372.
herpes recurrence. Arch Intern Med 1999: 159: 2430–2436. 67. King RM, Wilson GV. Use of a diary technique to investi-
47. Goldberg I, Ingher A, Brenner S. Pemphigus vulgaris trig- gate psychosomatic relations in atopic dermatitis. J Psy-
gered by rifampin and emotional stress. Skinmed 2004: 3: chosom Res 1991: 35: 697–706.
294. 68. Langan SM, Bourke JF, Silcocks P, Williams HC. An
48. Berrino AM, Voltolini S, Fiaschi D, et al. Chronic urticaria: exploratory prospective observational study of environ-
importance of a medical-psychological approach. Allerg mental factors exacerbating atopic eczema in children. Br J
Immunol 2006: 38: 149–152. Dermatol 2006: 154: 979–980.
49. Naldi L, Peli L, Parazzini F, Carrel CF. Family history of 69. Faulstich ME, Williamson DA, Duchmann EG, Conerly SL,
psoriasis, stressful life events, and recent infectious dis- Brantley PJ. Psychophysiological analysis of atopic dermati-
ease are risk factors for a first episode of acute guttate tis. J Psychosom Res 1985: 29: 415–417.
psoriasis: results of a case-control study. J Am Acad Der- 70. Altemus M, Rao B, Dhabhar FS, Ding W, Granstein RD.
matol 2001: 44: 433–438. Stress-induced changes in skin barrier function in healthy
50. Fortune DG, Richards HL, Griffiths CEM, Main CJ. Psy- women. J Invest Dermatol 2001: 117: 309–317.
chological stress, distress and disability in patients with 71. Zane L. Psychoneuroendocrinimmunodermatology:
psoriasis: consensus and variation in the contribution pathophysiological mechanisms of stress in cutaneous
of illness perceptions, coping and alexithymia. Br J Der- disease. In: Koo JYM, Lee CS, ed. Psychocutaneous medi-
matol 2002: 41: 157–174. cine. New York: Marcel Dekker Inc., 2003: 65–95.
51. Fortune DG, Richards HL, Griffiths CEM, Main CJ. Psy- 72. Buske-Kirschbaum A, Jobst S, Psych D, et al. Attenuated
chologic factors in psoriasis: consequences, mechanisms, free cortisol response to psychosocial stress in children
and interventions. Dermatol Clin 2005: 4: 681–694. with atopic dermatitis. Psychosom Med 1997: 59: 419–
52. Raychaudhuri SP, Farber FE, Raychaudhuri SK. Role of 426.
nerve growth factor in RANTES expression by kerati- 73. Buske-Kirschbaum A, Geiben A, Hollig H, Morschhauser
nocytes. Acta Derm Venereol 2000: 80: 247–250. E, Hellhammer D. Altered responsiveness of the hypo-
53. Nickoloff BJ, Schroder JM, von den Driesch P, et al. Is Psoria- thalamus-pituitary-adrenal axis and the sympathetic
sis a T-cell disease? Exp Dermatol 2000: 9: 357–375. adrenomedullary system to stress in patients with
54. Farber EM, Nall L. Psoriasis: a stress-related disease. Cutis atopic dermatitis. J Clin Endocrinol Metab 2002: 87:
1993: 51: 322–326. 4245–4251.
55. Farber EM, Nickoloff BJ, Recht B, Fraki JE. Stress, symme- 74. Rupprecht M, Rupprecht R, Kornhuber J, et al. Elevated
try, and psoriasis: possible role of neuropeptides. J Am glucocorticoid receptor concentrations before and after
Acad Dermatol 1986: 14: 305–311. glucocorticoid therapy in peripheral mononuclear leuko-
56. Aloe L, Alleva E, Fiore M. Stress and nerve growth factor: cytes of patients with atopic dermatitis. Dermatologica
findings in animal models and humans. Pharmacol Bio- 1991: 183: 100–105.
chem Behav 2002: 73: 159–166. 75. Delgado M, Fernández-Alfonso MS, Fuentes A. Effect of
57. Raychaudhuri SP, Jiang WY, Farber EM. Psoriatic kerati- adrenaline and glucocorticoids on monocyte cAMP-
nocytes express high levels of nerve growth factor. Acta specific phospodiesterase (PDE4) in a monocyte cell line.
Derm Venereol 1998: 78: 84–86. Arch Dermatol Res 2002, 190–197.
58. Raychaudhuri SP, Jiang WY, Smoller BR, Farber EM. 76. Schallreuter KU, Pittelkow MR, Swanson NN, et al. Altered
Nerve growth factor and its receptor system in psoriasis. catecholamine synthesis and degradation in the epider-
Br J Dermatol 2000: 143: 198–200. mis of patients with atopic eczema. Arch Dermatol Res
59. Richards HL, Ray DW, Kirby B, et al. Response of the 1997: 289: 663–666.
hypothalamic-pituitary-adrenal axis to psychological 77. Chan SC, Brown MA, Willcox TM, et al. Abnormal IL-4
stress in patients with psoriasis. Br J Dermatol 2005: 153: gene expression by atopic dermatitis T lymphocytes is
1114–1120. reflected in altered nuclear protein interactions with IL-4
60. Schmid-Ott G, Jacobs R, Jäger B, et al. Stress induced transcription regulatory element. J Invest Dermatol 1996:
endocrine and immunological changes in psoriasis 106: 1131–1136.
patients and healthy controls. Psychother Psychosom 78. Kagi MK, Wutrich B, Montano E, Barandun J, Blaser K,
1998: 67: 37–42. Walker C. Differential cytokine profiles in peripheral
61. Ginsburg IH. Coping with psoriasis: a guide for counsel- blood lymphocyte supernatants and skin biopsies from
ing patients. Cutis 1996: 57: 323–325. patients with different forms of atopic dermatitis, psoriasis
62. Tausk F, Whitmore SE. A pilot study of hypnosis in the and normal individuals. Int Arch Allergy Immunol 1994:
treatment of patients with psoriasis. Psychother Psycho- 103: 332–340.
som 1999: 68: 221–225. 79. Mori A, Yamamoto K, Dohi M, Suko M, Okudaira H.

29
Tausk et al.

Interleukin-4 gene expression in human peripheral severe recurrent genital herpes virus. Biofeedback Self
blood mononuclear cells. Int Arch Allergy Appl Immunol Regul 1985: 10: 181–188.
1991: 95: 282–284. 96. Yang EV, Glaser R. Stress-induced immunomodulation:
80. Tamir A, Ophir J, Brenner S. Pemphigus vulgaris trig- implications for tumorigenesis. Brain Behav Immun 2003:
gered by emotional stress [letter]. Dermatology 1994: 17 (Suppl. 1): S37–S40.
189: 210. 97. Reiche EM, Nunes SO, Morimoto HK. Stress, depression,
81. Schmid-Ott G, Jaeger B, Adamek C, et al. Levels of the immune system, and cancer. Lancet Oncol 2004: 5:
circulating CD8(+) T lymphocytes, natural killer cells, and 617–625.
eosinophils increase upon acute psychosocial stress in 98. Reiche EM, Morimoto HK, Nunes SM. Stress and depres-
patients with atopic dermatitis. J Allergy Clin Immunol sion-induced immune dysfunction: implications for the
2001: 107: 171–177. development and progression of cancer. International
82. Schmid-Ott G, Jaeger B, Meyer S, Stephan E, Kapp A, Werfel review of psychiatry (Abingdon, England). Int Rev Psychiatry
T. Different expression of cytokine and membrane mole- 2005: 17: 515–527.
cules by circulating lymphocytes on acute mental stress 99. Lillberg K, Verkasalo PK, Kaprio J, Teppo L, Helenius H,
in patients with atopic dermatitis in comparison with Koskenvuo M. Stressful life events and risk of breast
healthy controls. J Allergy Clin Immunol 2001: 108: 455– cancer in 10,808 women: a cohort study. Am J Epidemiol
462. 2003: 157: 415–423.
83. Braun CM, Huang SK, Bashian GG, Kagey-Sobotka A, 100. Riley V. Psychoneuroendocrine influences on immuno-
Lichtenstein LM, Essayan DM. Corticosteroid modulation competence and neoplasia. Science 1981: 212: 1100–1109.
of human, antigen-specific Th1 and Th2 responses. J 101. Wu W, Yamaura T, Murakami K, et al. Social isolation
Allergy Clin Immunol 1997: 100: 400–407. stress enhanced liver metastasis of murine colon 26-L5 car-
84. Byron KA, Varigos G, Wootton A. Hydrocortisone inhibi- cinoma cells by suppressing immune responses in mice.
tion of human interleukin-4. Immunology 1992: 77: 624– Life Sci 2000: 66: 1827–1838.
626. 102. Ramirez AJ, Craig TK, Watson JP, Fentiman IS, North WR,
85. Cupps TR, Gerrard TL, Falkoff RJ, Whalen G, Fauci AS. Rubens RD. Stress and relapse of breast cancer [see com-
Effects of in vitro corticosteroids on B cell activation, pro- ments]. Br Med J 1989: 298: 291–293.
liferation, and differentiation. J Clin Invest 1985: 75: 754– 103. Havlik RJ, Vukasin AP, Ariyan S. The impact of stress on
761. the clinical presentation of melanoma. Plast Reconstr Surg
86. Schwiebert LM, Beck LA, Stellato C, et al. Glucocorticos- 1992: 90: 57–61; discussion 2–4.
teroid inhibition of cytokine production: relevance to anti- 104. Reynolds P, Kaplan GA. Social connections and risk for
allergic actions [published erratum appears in J Allergy cancer: prospective evidence from the Alameda County
Clin Immunol 1996 Sep; 98(3):718]. J Allergy Clin Immu- Study. Behav Med 1990: 16: 101–110.
nol 1996: 97: 143–152. 105. Jacobs JR, Bovasso GB. Early and chronic stress and their
87. Theoharides TC, Donelan JM, Papadopoulou N, Cao J, relation to breast cancer [In Process Citation]. Psychol
Kempuraj D, Conti P. Mast cells as targets of corticotropin- Med 2000: 30: 669–678.
releasing factor and related peptides. Trends Pharmacol 106. Grossarth-Maticek R, Eysenck HJ, Boyle GJ, Heeb J, Costa
Sci 2004: 25: 563–568. SD, Diel IJ. Interaction of psychosocial and physical risk
88. Papadopoulou N, Kalogeromitros D, Staurianeas NG, factors in the causation of mammary cancer, and its pre-
Tiblalexi D, Theoharides TC. Corticotropin-releasing vention through psychological methods of treatment.
hormone receptor-1 and histidine decarboxylase expres- J Clin Psychol 2000: 56: 33–50.
sion in chronic urticaria. J Invest Dermatol 2005: 125: 107. Laudenslager ML, Ryan SM, Drugan RC, Hyson RL, Maier
952–955. SF. Coping and immunosuppression: inescapable but not
89. Biondi M, Zannino LG. Psychological stress, neuroimmu- escapable shock suppresses lymphocyte proliferation.
nomodulation, and susceptibility to infectious diseases in Science 1983: 221: 568–570.
animals and man: a review. Psychother Psychosom 1997: 108. Ben-Eliyahu S, Page GG, Yirmiya R, Shakhar G. Evidence
66: 3–26. that stress and surgical interventions promote tumor
90. Bailey MT, Engler H, Sheridan JF. Stress induces the development by suppressing natural killer cell activity. Int
translocation of cutaneous and gastrointestinal micro- J Cancer 1999: 80: 880–888.
flora to secondary lymphoid organs of C57BL/6 mice. 109. Ben-Eliyahu S. The promotion of tumor metastasis by
J Neuroimmunol 2006: 171: 29–37. surgery and stress: immunological basis and implications
91. Rojas IG, Padgett DA, Sheridan JF, Marucha PT. Stress- for psychoneuroimmunology. Brain Behav Immun 2003:
induced susceptibility to bacterial infection during cuta- 17 (Suppl. 1): S27–S36.
neous wound healing. Brain Behav Immun 2002: 16: 74– 110. Parker J, Klein SL, McClintock MK, et al. Chronic stress
84. accelerates ultraviolet-induced cutaneous carcinogene-
92. Buske-Kirschbaum A, Geiben A, Wermke C, Pirke KM, sis. J Am Acad Dermatol 2004: 51: 919–922.
Hellhammer D. Preliminary evidence for Herpes labialis 111. Saul AN, Oberyszyn TM, Daugherty C, et al. Chronic
recurrence following experimentally induced disgust. stress and susceptibility to skin cancer. J Natl Cancer Inst
Psychother Psychosom 2001: 70: 86–91. 2005: 97: 1760–1767.
93. Cohen S, Frank E, Doyle WJ, Skoner DP, Rabin BS, Gwalt- 112. Spiegel D, Bloom JR, Kraemer HC, Gottheil E. Effect of
ney JM Jr. Types of stressors that increase susceptibility to psychosocial treatment on survival of patients with meta-
the common cold in healthy adults [see comments]. static breast cancer [see comments]. Lancet 1989: 2: 888–
Health Psychol 1998: 17: 214–223. 891.
94. Cohen S, Tyrrell DA, Smith AP. Psychological stress and 113. Fawzy FI, Fawzy NW, Hyun CS, et al. Malignant melanoma.
susceptibility to the common cold [see comments]. N Effects of an early structured psychiatric intervention,
Engl J Med 1991: 325: 606– 612. coping, and affective state on recurrence and survival 6
95. VanderPlate C, Kerrick G. Stress reduction treatment of years later. Arch Gen Psychiatry 1993: 50: 681–689.

30
Psychoneuroimmunology

114. Herberman RB, Ortaldo JR. Natural killer cells: their Holliday J, Glaser R. Psychosocial modifiers of immuno-
roles in defenses against disease. Science 1981: 214: 24– competence in medical students. Psychosom Med 1984:
30. 46: 7–14.
115. Esterling BA, Kiecolt-Glaser JK, Glaser R. Psychosocial 117. Glaser R, Rice J, Speicher CE, Stout JC, Kiecolt-Glaser JK.
modulation of cytokine-induced natural killer cell activity Stress depresses interferon production by leukocytes
in older adults. Psychosom Med 1996: 58: 264–272. concomitant with a decrease in natural killer cell activity.
116. Kiecolt-Glaser JK, Garner W, Speicher C, Penn GM, Behav Neurosci 1986: 100: 675–678.

31

Vous aimerez peut-être aussi