17
Journal of Clinical Laboratory Analysis 4:22-38 (1 990) Human Psychoneuroimmunology Today Massimo Biondi and Giorgio D. Kotzalidis Third Psychiatric Clinic, University of Rome, “La Sapienza, Rome, Italy Studies in human psychoneuroimmunology began around 1919, but a systematic ap- proach wasn’t used until the work of Solomon in the 1960s. Recently, the new specialty has achieved relative independence due to con- siderable data acquisition. Stress research has revealed relation- ships between neuroendocrine and immune changes. In parallel, increasing evidence of immunological alterations in psychiatric dis- eases has expanded the field; presently, immunological correlates of psychosomatic diseases and personality are sought. On the other hand, while immunological disease has been psychologically assessed for many years, a clear-cut link between psyche and immunological changes has yet to be shown. This fact, along with the therapeutic implica- tions of advancing knowledge, will influence strongly the future trends of psychoneuro- immunology. Concepts emerging from the study of this field will be of heuristic value to both psychiatry and immunology and will help define new and expanded limits for both disciplines. Key words: Psychoneuroimmunology, mind-endocrine-immune interactions, schizophrenia, depression, stress, psychosomatic disease, personality, cancer, immunity INTRODUCTION Psychoneuroimmunology examines the analogies and recip- rocal influences, direct or indirect, between the nervous sys- tem and the immune system. The observation that psychic activity can influence immune phenomena dates back to 19 18 (1) and was a consequence of the discovery of conditioned reflex. During the late 1920s, immunological alterations were demonstrated in mental diseases (2), and during the mid- 1950s, a clear relationship was described among brain activ- ity, endocrine organs, and immune function (3). The term psychoimmunology did not appear in Solomon and Moos’ 1964 article (4), as is often reported; however, this article contained many of the ideas that subsequently contributed to some of the basic tenets of psychoneuroimmunology , a term coined in the early 1980s (5,6). Since there was no one journal spe- cifically devoted to the new field, relevant papers ended up in more than 200 journals. At present, the journals Mind, Brain, and Immunity and Progress in Neuroendocrinim- munology and the increasing number of textbooks on the topic bear witness to its great fascination for scientists. Although it has long been evident that immune activity influences neuronal function, a molecular basis for a com- plete neuro-endocrine-immune loop was not suggested until it was demonstrated that interleukin-1 is able to influence the pituitary (7) and the hypothalamus (8-10). The direct influence of neural tissue on immune cells has been substantiated by anatomical studies ( 1 1 - 16); the endo- crine nature of immunocyte subpopulations became appar- ent when it was shown that lymphocytes are able to synthesize 0 1990 Wiley-Liss, Inc. and secrete adrenocorticotropic hormone (ACTH) and beta- endorphin (17,18) and other opioids (1 9). Furthermore, lym- phocytes possess receptors for neurotransmitters and hormones (20) and are particularly susceptible to the effects of stress (21). Soon after its discovery, epinephrine became one of the first hormones to be thought of as a mediator of psychological influences on immune reactivity (l), and the immunodepressant effect of cortisol has been known since the pioneering work of Selye on stress (3). The impact of stress research on the development of psychoneuroimmunological conceptual frame- works can hardly be overemphasized, as will become appar- ent in the next section. IMPACT OF STRESS RESEARCH Selye’s studies were the first series of systematic observa- tions on the effects of stressors on immunity. After exposing animals to stress, he observed lymphatic organ involution, lymphocytopenia, and reduced resistance to infectious agents (22). Subsequent animal studies showed stress to induce delays and decreases in immunoglobulin (Ig) production; reduced B lymphocyte reactivity associated with booster effect sup- pression and secondary response impairment; reductions in T cell responses to mitogens; reductions in helper function, cytotoxic activity, and natural killer (NK) function; altered delayed hypersensitivity and graft-versus-host reactions; and Received March 29, 1989. Accepted June 22, 1989. Address reprint requests to Massimo Biondi, M.D., I11 Clinica Psichiatrica, Universita di Roma, “La Sapienza,” 00185 Roma, Italy.

Human psychoneuroimmunology today

Embed Size (px)

Citation preview

Journal of Clinical Laboratory Analysis 4:22-38 (1 990)

Human Psychoneuroimmunology Today Massimo Biondi and Giorgio D. Kotzalidis

Third Psychiatric Clinic, University of Rome, “La Sapienza, ” Rome, Italy

Studies in human psychoneuroimmunology began around 1919, but a systematic ap- proach wasn’t used until the work of Solomon in the 1960s. Recently, the new specialty has achieved relative independence due to con- siderable data acquisition.

Stress research has revealed relation- ships between neuroendocrine and immune changes. In parallel, increasing evidence of immunological alterations in psychiatric dis- eases has expanded the field; presently, immunological correlates of psychosomatic diseases and personality are sought. On the

other hand, while immunological disease has been psychologically assessed for many years, a clear-cut link between psyche and immunological changes has yet to be shown. This fact, along with the therapeutic implica- tions of advancing knowledge, will influence strongly the future trends of psychoneuro- immunology. Concepts emerging from the study of this field will be of heuristic value to both psychiatry and immunology and will help define new and expanded limits for both disciplines.

Key words: Psychoneuroimmunology, mind-endocrine-immune interactions, schizophrenia, depression, stress, psychosomatic disease, personality, cancer, immunity

INTRODUCTION

Psychoneuroimmunology examines the analogies and recip- rocal influences, direct or indirect, between the nervous sys- tem and the immune system. The observation that psychic activity can influence immune phenomena dates back to 19 18 (1) and was a consequence of the discovery of conditioned reflex. During the late 1920s, immunological alterations were demonstrated in mental diseases (2), and during the mid- 1950s, a clear relationship was described among brain activ- ity, endocrine organs, and immune function (3). The term psychoimmunology did not appear in Solomon and Moos’ 1964 article (4), as is often reported; however, this article contained many of the ideas that subsequently contributed to some of the basic tenets of psychoneuroimmunology , a term coined in the early 1980s (5,6). Since there was no one journal spe- cifically devoted to the new field, relevant papers ended up in more than 200 journals. At present, the journals Mind, Brain, and Immunity and Progress in Neuroendocrinim- munology and the increasing number of textbooks on the topic bear witness to its great fascination for scientists.

Although it has long been evident that immune activity influences neuronal function, a molecular basis for a com- plete neuro-endocrine-immune loop was not suggested until it was demonstrated that interleukin-1 is able to influence the pituitary (7) and the hypothalamus (8-10).

The direct influence of neural tissue on immune cells has been substantiated by anatomical studies ( 1 1 - 16); the endo- crine nature of immunocyte subpopulations became appar- ent when it was shown that lymphocytes are able to synthesize

0 1990 Wiley-Liss, Inc.

and secrete adrenocorticotropic hormone (ACTH) and beta- endorphin (17,18) and other opioids (1 9). Furthermore, lym- phocytes possess receptors for neurotransmitters and hormones (20) and are particularly susceptible to the effects of stress (21). Soon after its discovery, epinephrine became one of the first hormones to be thought of as a mediator of psychological influences on immune reactivity (l), and the immunodepressant effect of cortisol has been known since the pioneering work of Selye on stress (3). The impact of stress research on the development of psychoneuroimmunological conceptual frame- works can hardly be overemphasized, as will become appar- ent in the next section.

IMPACT OF STRESS RESEARCH

Selye’s studies were the first series of systematic observa- tions on the effects of stressors on immunity. After exposing animals to stress, he observed lymphatic organ involution, lymphocytopenia, and reduced resistance to infectious agents (22). Subsequent animal studies showed stress to induce delays and decreases in immunoglobulin (Ig) production; reduced B lymphocyte reactivity associated with booster effect sup- pression and secondary response impairment; reductions in T cell responses to mitogens; reductions in helper function, cytotoxic activity, and natural killer (NK) function; altered delayed hypersensitivity and graft-versus-host reactions; and

Received March 29, 1989. Accepted June 22, 1989.

Address reprint requests to Massimo Biondi, M.D. , I11 Clinica Psichiatrica, Universita di Roma, “La Sapienza,” 00185 Roma, Italy.

Human Psychoneuroimmunology 23

to be lower during between-test periods (32); the elevation in salivary IgA tended to correlate with salivary norepinephrine and to be higher (31) and more persistent in high-power inhib- ited individuals (32). Medical students showed decreased plasma levels but not salivary IgA levels (33), lower NK cell activity (33-33, lower OKT4 + cell percentages, and a lower OKT4/OKT8 ratio (34) on the day of a test compared with 1 mo earlier. They also showed decreased interferon produc- tion on the day of the test compared with 6 wk earlier (35). Increased monocytes and decreased OKT4, OKT8, and interleukin-2 (IL-2)-receptor-positive cells were observed in students of psychology on the day of examinations compared with 6 wk earlier and compared with controls not taking exami- nations (36). T cell response to antigens, mitogens, and allo- geneic cells in the same subjects were decreased from 6 wk before the examinations to 2 wk after.

Preoperative stress was shown to affect the response to phy- tohemagglutinin (PHA) and pokeweed mitogen (PKW) unfa- vorably in individuals particularly sensitive to stress, thus influencing postoperative outcome (37). Nine months of unem- ployment negatively affected the response of T cells to PHA in women (38), and airplane flight induced nonspecific, stress- related alterations in T and B cell responses to mitogens and in T and B cell cooperation (39). Novelty induced a parallel increase in anxiety, plasma cortisol, OKT4 + cell percent- age, and OKT4/OKT8 ration in medical students (40). Decreased mitogen proliferation responses were found in an oral mental arithmetic stress setting (41). In this study, it was shown that type A subjects (those with coronary disease-prone behavior patterns) had significantly higher proliferative responses to PHA than did type B subjects (those with low- coronary-risk behavior patterns). This result is surprising since the former tend to have chronic sympathetic activation, lead- ing to decreased immune responsivity (42). However, sim- ilar studies should be done before conclusions are made.

Stressful life events have been shown to affect immuno- competence in a variety of stress paradigms. High life event scorers showed decreased blastogenic responses to PHA and PKW in a presurgical stress context (37) and decreased NK activity in academic stress (33) and bereavement contexts (43). Another study found no significant correlation between life stress scores and NK activity but did find NK activity to be a measure of good coping (44). Anxiety and depression corre- lated inversely with NK activity in that study (44).

reduced graft rejection and interferon production. [For a review, see Monjan (23)].

Commonly assessed immunological parameters in both stress-related and non-stress-related contexts are listed in Table 1. Immune assessment during stress in humans has been done using several paradigms, including academic stress, pre- operative and forced wakefulness stress, existential stress accu- mulation, frustration, bereavement, unemployment, and marital discord and divorce (Table 2). [For a review, see Palmblad (24) or Biondi and Pancheri (25).]

Stressful Life Events

Bereavement (loss of spouse) was shown to be associated with prolonged depression of T cell (26,27) and B cell reac- tivity to mitogens (27) in longitudinal prospective studies and with decreased NK activity (28) in a tangential study. In the latter study (28), the marital loss and anticipated loss groups showed significantly lower NK activity in comparison with the control group, whereas cortisolemia was significantly higher in the bereaved as compared with the other groups. Renormalization of T cell proliferative response to mitogens did not occur prior to 6 mo after the loss event (27).

Divorce affects immunity in a more complex way. Divorced men show an abnormal antibody response to Epstein-Barr virus (EBV) and a lower OKT4/OKT8 ratio compared than con- trols (married men); however, in men who initiated the sepa- ration, the immune impairment was less severe (29). Divorced women showed a decreased antibody response to EBV, decreased OKT4 + cells, decreased Nk activity, and greater psychic depression with than did controls (married women) (30). Immune dysregulation correlated to both depression and attachment to the exspouse (30).

Academic stress induced an increase in the salivary IgA secretion rate, which tended to decrease after a test (31) and

TABLE 1. Stress-Sensitive Immunological Parameters

Number of circulating lymphocytes Number of circulating monocytes B cell/T cell ratio Killer (suppressor)/helper (inducer) ratio Helper (inducer) percentage Antibody response (plasma, saliva, skin) Response to mitogens (PHA, con-A, PKW) E-rosette formation NK cell activity Surface receptors Steroid receptors

TABLE 2. Stressful Life Events Probably Related to Immunodepression

Death of spouse Divorce Test taking Waiting for surgery

Coping and Personality

Reaction patterns to everyday life stress are probably related to the coping style of an individual and appear to be impor- tant determinants of stress-induced immunodepression. Sub- jects scoring high on the repression-denial scale of the Reaction Pattern Test (45) had low proliferative responses to PHA and reduced E-rosette formation and skin test hyporeactivity com-

Lacking or losing job pared with low deniers in a preoperative setting (46). In a

24 Biondi and Kotzalidis

female sample with breast disease, subjects who showed a poor mitogenic response to PHA were high deniers and scored higher on the depression scale of the MMPI (47).

Loneliness has been reported to correlate to self-perceived stress (48). High scorers on the UCLA Loneliness Scale had significantly lower NK activity levels (33,48) and antibody response to viral challenge [EBV, herpes simplex 1, and cyto- megalovirus (49)J than did low scorers.

Whereas high stress agonizes immunodepression in humans, relaxation acts as an inverse agonist. Frequent relaxation prac- tice resulted in an increase in the percentage of OKT4 + cells (34); different types of relaxation resulted in increased levels of salivary IgA not dependent on cortisolemia (50). Transcen- dental meditation reduced lymphocyte high-affinity beta- adrenoceptor binding, thus rendering lymphocytes less sensitive to the actions of stress (5 1).

Stress and Infection

Altered immunity increases susceptibility to various patho- gens. In healthy individuals, the action of infectious agents is specifically blocked by the immune system, which starts countering the disease as soon as it contacts a pathogen. The brain is influenced by even the mildest infection (52) and influ- ences and organizes the immune response (53-56). A heav- ily stressed brain does not contribute to healing responses in an ideal manner (57-59); hence, it might be that stress’s abil- ity to modulate susceptibility to infection (a largely documented fact [60-621 occurs via both brain and endocrine effects.

While brain effects are difficult to assess due to high indi- vidual differences, endocrine effects have been investigated adequately. In particular, it was shown that catecholamine activity (42,63,64) and plasma corticosteroid levels (63) are increased prior to the onset of disease. Plasma catecholamines and cortisol induce alterations in immune competence, so that it may be theorized that increased stress increases adrenal activ- ity with a consequent rise in epinephrine and cortisol; the two hormones subsequently act on lymphocytes to decrease immune competence, enabling an infectious agent to invade the organism relatively undisturbed and cause illness at a clini- cal level. This explanation of the effects of stress is based on fragmentary though sound experimental evidence. It has yet to be proven convincingly in a single prospective experiment involving subjective stress quantification, serial measurement of plasma cortisol and catecholamines, and serial evaluation of immune function. Also, this simplistic explanation does not take into account that there are other stress hormones that also have definite, although poorly defined, effects on immu- nity. [For a review, see Calabrese et al. (65).] Also not accounted for is that cortisol is not always and exactly an immunosuppressive agent, but rather it is an inhibitor of excessive and unspecific immune reactions (66). However, glucocorticoids have been shown to suppress human NK cell activity in vitro (67). A study by Dorian et al. (68), which

unfortunately did not include hormonal evaluations, showed that, among the various immune parameters tested, NK activ- ity impairment predicted susceptibility to viral illness. Highly stressed individuals were likely to present more psychopa- thology and less NK activity in this study.

Among viral infections, the acquired immunodeficiency syndrome (AIDS) poses the most formidable problems. When AIDS first appeared, it was not possible to assess the stress exerted on the affected individual, nor was it possible to assess premorbid perceived stress. Now, however, it is possible to assess perceived stress in human immunodeficiency virus (HIV)-positive individuals (those experiencing a life- threatening condition) and to compare them with patients with full-blown AIDS (who are almost certain of their fate). Anx- iety and distress are higher in the HIV-positive patients, who report the onset of AIDS almost with relief (69). Controlled studies are warranted to examine whether stress is a factor in determining HIV-seropositivity or progression to AIDS. The reader will profit from referring to a recent book on the neu- ropsychiatric aspects of AIDS (70), although conclusions can- not be made at this point.

Stress and Neoplasia

A mechanism similar to that proposed for infections could be responsible for stress effects on tumorigenesis, a process that is presently thought to be associated with decreased immunosurveilance. As with infectious diseases, the loss of N K cell competence has been associated with malignant growth (71). Lymphocytes damaged by exposure to high corticoid levels may fail to control either latent viruses or malignant mutant cells (72); in healthy people, mutations are corrected by DNA-repairing methyltransferase, a stress- sensitive enzyme (73). Corticoids also act at a nuclear level, thus directing DNA-dependent RNA synthesis; an excess of nuclear steroid receptor occupancy could have mutagenic con- sequences. Highly stressed individuals exhibit poor DNA repairing capacity (74), thus being more susceptible to malig- nancy. Recent life change stress was found to be related to lung cancer in the young (75). Low levels of cooperative cop- ing style combined with pessimism, somatic anxiety, and life threat are related to progression of benign tumor of the cervix to squamous-cell carcinoma (76). Psychic depression has been found to correlate with cancer mortality (77) but not morbidity (78); however, orality-dependency was found to be unrelated to cancer risk (79). Cancer heterogeneity and the involvement of nonpsychological factors render the data on psychological influ- ences difficult to interpret (80). At present, the hypotheses on psychic and immune interactions in determining disease susceptibility have to focus on phenomena impinging on the function of the hypothalamic-pituitary-adrenal (HPA) axis, namely stress, anxiety, and depression (65-83).

In conclusion, although there is sufficient evidence that emotional stress affects the immune system (84), there is no

Human Psychoneuroimmunology 25

been debated on scientific grounds. Depression was found to correlate (77,88) or not to correlate (78,89) with cancer. Schizophrenia, on the other hand, has been correlated inversely with cancer (90,91); however, this finding should be inter- preted with caution. Most schizophrenics are first diagnosed during adolescence and early adulthood (92), whereas cancer is usually diagnosed later in life, and schizophrenics have a risk of death more than double of that of the general population (92)-partly because of a higher suicide rate (93). In view of these facts, studies showing a positive or no rela- tionship between the schizophrenia and cancer [reviewed by Baldwin (94)] should be read in terms of positivity.

Cancer was found to occur more often in high deniers (95,96), in Eysenck’s extroverts (97), in Sjobring’s substable women (98), and in persons who excessively either express or suppress their emotions (99). These results have led to the theorization of a cancer-prone behavior pattern (type C); according to this theory, an overcooperative, appeasing, unas- sertive, harmonizing patient who is alexithymic toward neg- ative emotions, compliant with external authority, and anxious and defensive toward stress has more probability to develop cancer than someone without these traits ( 1 00,lO 1). Work on this promising construct is in progress (102).

Although denial has been assumed to be a positive predic- tor of cancer incidence (95), it has been shown to prolong life after cancer onset (103). On the other hand, a negative prognostic behavioral pattern for cancer survival has been reported to be passivity-helplessness (103- 105). However, methodological discrepancies limit the validity of these data.

clear evidence that it promotes disease in unpredisposed indi- viduals. At present, most studies do not distinguish between acute and chronic stress or between effectively and ineffec- tively coped-with stress. They also do not take into account the adaptation potential of organisms (85). As for life stresses, methodological problems prevent us from drawing firm con- clusions (86). Stress events should be examined in relation to the status of the organism at the starting moment of the pathogenetic process. However, the “incubation” time var- ies in different diseases, and this difference is sufficient to create problems of questionnaire standardization.

PSYCHIATRY AND IMMUNOLOGY

Frequently, infectious diseases present with psychiatric symptoms. Also, specific behavioral changes are often pres- ent in medical diseases for which an immunological causa- tion has been supposed or immunological alterations found. The psychic and the immune may interact at any organ of the body, including the brain.

A leading American textbook of psychiatry (87) includes in its chapter on psychosomatic medicine gastrointestinal dis- orders, obesity, anorexia nervosa, cardiovascular disorders, respiratory disorders, endocrine disorders, skin disorders, rheu- matoid arthritis, accident proneness, headache, and immune disorders; cancer is briefly treated in the latter section, whereas sexual disorders are surprisingly absent (they are dealt with in another section of the book). Every section in this chapter, except those on accident proneness and cardiovascular and eating disorders, discusses immunological mechanisms. How- ever, only the sections on immune disorders tried to present an integrated view of mind-brain-immune interactions. In ulcer- Acquired immunodeficiency Syndrome ative colitis, Crohn’s disease, bulimia, coronary-prone behavior pattern (not actually a disease-probably a myocardial infarc- tion-promoting factor), asthma, APUDomas, autoimmune thy- roiditis, Grave’s disease, diabetes mellitus, Cushing’s disease, alopecia areata, rheumatoid arthritis, systemic lupus erythe- matosus (SLE), progressive systemic sclerosis, cluster head- ache, and allergy, both immunological and psychiatric abnormalities occur; whether these abnormalities are interde- pendent or bear a causal relationship remains to be established.

In our opinion, connective tissue diseases, APUDomas, and alopecia areata deserve special attention, as they have been poorly assessed from a psychological viewpoint. Our preliminary results on alopecia areata suggest a possible link with affective disorders, although caution is advised since links of the latter with panic disorders and chronic pain, advanced on purely pharmacotherapeutic grounds (ex adiu- vantibus criterion), proved to have a tenuous biological sub- strate (Teodori, Paga, and Biondi, unpublished results).

Neoplasia

Patients with AIDS quite often present with psychiatric symptoms. High levels of anxiety are usually present, and onset is frequently characterized by marked depression. Symp- toms suggesting schizophrenia may also be observed. Delu- sions may be a feature of presentation. The premorbid personality of the AIDS patient has not yet been fully assessed, so the possibility of psychic disease pre- or coex- istence or facilitation has not been ruled out. Dementia is the only psychiatric symptom associated with AIDS that is known to be a sequela.

The external envelope protein of HIV- 1, gp 120, bears struc- tural homologies to neuroleukin (106,107) and vasoactive intestinal polypeptide (VIP) (108-1 1 1). Neuroleukin is a growth factor that was first isolated in the nervous system and that also has a lymphokine role. In particular, neuroleukin can stimulate T-dependent monocyte proliferation, probably as an autocrine effect (1 12). The stimulated monocytes can then induce a polyclonal B cell activation, thus causing hyperimmunoglobulinemia, a frequently observed finding in people with AIDS (PWAs). A neuroleukin receptor has yet

Galen observed cancer to occur more frequently in melan- cholic women; after about 18 centuries this observation has

to be isolated and localized in the nervous and immune tis- sue; furthermore, the role of neuroleukin in adult brain has

26 Biondi and Kotzalidis

not been assessed and there is no evidence that central ner- vous system (CNS) symptomatology is induced through gp 120 agonism or antagonism of neuroleukin function (I 13).

VIP is an amine precursor uptake and decarboxylation (APUD) family peptide, synthesized and secreted by neurons and, probably, immunocytes. Within the brain, VIP is widely distributed, but it is most prominent in the hypothalamus, the cerebral cortex, and the hippocampus ( 1 14,115), sites involved in neuroendocrine regulation, biological intelligence, information processing, and emotions. VIP receptors are found in the CNS (1 16- 1 18), on nonadherent peripheral lympho- cytes (119,120), and in monocytes (120,121) and can be of both type 1 and type 2 subtype (122). VIP binding to its lym- phocyte receptors induces decreased concanavalin-A (con-A) (1 23,124) and PHA ( 123) responses; besides these actions, VIP inhibits NK cell activity (123 , increases con-A-driven Ig synthesis (122,123, and enhances lymphocyte release in the lymphatic fluid ( 126).

HIV- 1 envelope protein gp 120 binds to CD4, a differenti- ation antigen common to lymphocytes, macrophages, and, among others, neurons with a cortical and subcortical distri- bution (1 27). CD4 has been attributed the role of HIV- 1 recep- tor (128); hence, that peptides binding CD4 may inhibit HIV activity (108,129) has been interpreted in terms of receptor specificity. This interpretation has prompted a search for an endogenous substance with a structural homology with a gp 120 amino acid sequence. VIP 1-12 was found to have both structural homology with gp 120 and CD4 binding ability (108-1 10,129). It was thus hypothesized thatHIV-1 interferes with the normal actions of VIP to produce AIDS symptom- atology (1 11).

The cortical and subcortical distribution of VIP binding sites fits the existence of dementia, affective symptoms, and psychosis. There have also been suggestions that VIP mechanisms may be implicated in depression (130) and in schizophrenia (1 3 1,132), although mechanisms of either hyper- activity or hypoactivity can be implicated. In the rat brain, VIP stimulates postsynaptic dopaminergic mechanisms ( 133); should this also be found to be true in humans, it would asso- ciate VIP hyperactivity with psychosis and hypoactivity with depression. The effects of VIP on the immune system are consistent with an hyperVIPergic effect of HIV rather than with inhibition. It is interesting that the Verner-Morrison syn- drome, a pancreatic or gastrointestinal APUDoma character- ized by watery diarrhea, hypokalemia, and hypochlorhydria, involves VIP hypersecretion and that AIDS most commonly

to be answered. However, the answer to Pert and Dienstfrey’s question-how can a virus produce such great damage by inhibiting the peptides’ good work in few sites? (1 1 I)--could be that this is not the only thing the virus does.

Psychiatric Aspects of Immunological Activity

It recently became apparent that immune products influ- ence behavior. Immune complexes are capable of inducing eating ( I 35) in the rat, an effect that is complement-dependent and mediated through a hypothalamic action (136). SLE may present dementia, lethargy, and psychotic features, either schizophrenia-like or depressive (1 37), and such symptoms have been partly attributed to immune complex deposits (1 38) in the cerebrospinal fluid of patients with SLE (139). In order to relate immune complex deposition to SLE psychiatric man- ifestations, such deposits should be found in cortical or lim- bic regions.

The hypothesis of a relationship between Igs and schizo- phrenic symptomatology has not been confirmed (140). Gen- erally, autoimmune hypotheses of psychiatric diseases are based on findings of circulating autoimmune complexes, which might well be iatrogenic. Also, they are inconsistent, except in bipolar illness, where antinuclear antibodies are pres- ent before pharmacological treatment (141).

Interferons (IFs) have behavioral effects consistent with central, partly morphinomimetic, antidopaminergic, antisero- tonergic, and steroid mechanisms (142-147). IF-alpha is neurotoxic, inducing behavioral depression, retardation, asthe- nia, and irritability (148), symptoms usually associated with fever. These symptoms tend to occur in a milder form with IF-gamma (149), which has been shown to activate the HPA axis (142,143). In turn, memory-related hormones released by acute stress tend to switch off IF-gamma release from lym- phocytes (150,151). Memory disturbance associated with slow wave electroencephalographic (EEG) activity has been ob- served with IF-alpha (152) but not gamma (147). Farkkila et al. (147) found that cerebrospinal fluid serotonin turnover was reduced by IF-gamma, an effect that could result in reduced coricotropin-releasing factor secretion. IF-alpha increased psy- chiatric symptomatology in patients with pre-existing psychi- atric problems such as phobia and obsessive thought (153). The greatest psychic unbalance was shown by HIV-positive patients (153). It might be speculated that, in patients with impaired tolerance to stress, psychiatric symptoms tend to reemerge more easily after IF-alpha treatment.

presents with diarrhea. Although the T peptide sequence from gp 120 does not bind enterohepatic VIP receptors (134), lmmunopsychopharmacology

patients with Verner-Morrison syndrome should receive psy- chiatric and psychologic assessment to check for eventual sim- ilarities between the clinical picture of this histologically benign but prognostically unfavorable tumor and that of AIDS. Whether T peptide interacts with brain and lymphocytic VIP receptors and which is the subtype involved are questions yet

Drugs used in common psychiatric practice have immuno- logical effects. Contrary to the common belief that benzo- diazepines cause immunosuppression, there is indirect evidence that they can actually enhance immune function (154,155). Antipsychotics have a biphasic effect on T helper and total T cell numbers; they increase both at low doses and

Human Psychoneuroimmunology 27

not to be consistent with time (176). These cells might in part represent NK cells, a cell type that tends to be reduced in a particular subpopulation of schizophrenics (179). A decrease in macrophage function has also been described (179). Whether these abnormalities represent a result of neu- roleptic medication is still controversial (176).

The T suppressorlhelper ratio has been described as increased (180) or decreased ( 1 8 I ), and this discrepancy has been attributed to schizophrenic population heterogeneity (176). Theophylline-sensitive T lymphocytes, representing OKT8 + , Fc-receptor positive T suppressors, were found to be reduced in a recent study (182). B cell counts were found to be increased in one study (183). NK, B cell, and T cell reduced responsiveness has been reported in several studies (179,183,184), butwhether it isacause, aneffect, o radrug treatment epiphenomenon has not been settled (176).

A finding of hyper-Igemia would suggest active infection, exaggerated B cell reactivity (of which there exists no con- sistent evidence), or autoimmunity. Igs have been found to be increased (1 85- 187), decreased ( 188), or unchanged ( 189) in schizophrenics; moreover, Ig levels did not differ between schizophrenic and nonschizophrenic family members (190).

Self-directed antibrain antibodies were found in schizophre- nics (180,186), but similar findings may be seen in normals and are thought to be nonspecific. The subtype of schizo- phrenia could be important in trying to establish differences in autoantibodies between schizophrenics and controls; antimyelin basic protein antibodies were found increased in catatonics compared with other schizophrenics compared with normals (191), but the sample was small so that inferences cannot be made. An eventual antibody raised against dopa- minergic receptors would be consistent with the dopamine hypothesis of schizophrenia, but the search for such a mole- cule has yielded no positive results (192).

Viral hypotheses involve the herpes class of viruses (193), cytomegalovirus (194), and retroviruses (195), but negative results have been reported (1 96,197). That a viral infection or autoimmunity is present in schizophrenia is suggested by the presence of higher interferon levels in schizophrenics than in normals (198), but alternative explanations are possible. The observation that populations fed with gluten-free diets show a low incidence of schizophrenia led to the theory that persons with a genetic deficit in gluten-metabolizing capac- ity could develop antibodies against gluten; therefore, either gluten (199) or immune complexes could induce psychotic behavior (200). This hypothesis has found no experimental support (200).

Other approaches seeking a link between brain and immune correlates of schizophrenia are the immunogenetic and the lymphocyte surface receptor investigations. HLA studies showed a great variety of associations (201). Recent studies have showed either negative results (202) or a weak associa- tion with A1 and B37 and a somewhat stronger association with a combination of the two (203). Tritiated spiroperidol

diminish them at high doses (156). This biphasic effect and methodological inaccuracies partly account for discrepancies observed in the past and reviewed by Kaufmann and cowork- ers (1 57). Response to viruses can be affected bidirectionally by antipsychotics, depending on the virus (158). Mechanisms involving phenothiazine effects on lymphocytic RNA (158, 159), independent of dopaminergic blockade (159), are thought to mediate these effects. However, chlorpromazine dose-dependently inhibits DNA synthesis in human lympho- cytes in vitro (1 60).

Lithium induces an abnormal thyroid-stimulating hormone (TSH) response to protirelin (TRH) (161) and increased antithyroid Ig formation (162). Another drug used in bipolar illness, carbamazepine, is myeloplegic and induces autoim- munity (141). Tricyclic antidepressants have not been inves- tigated extensively with regard to their influence on the immune system. However, since they are likely to bind to a lymphocytic muscarinic acetylcholine receptor ( 163), prob- ably of the M2 subtype (164), they may improve immune function. It is remarkable that levamisol, an immunomodu- lating drug, induced a reversal of tricyclic resistance in a small sample of depressed patients, along with improvement in immune parameters and mood (165). For readers at ease with Italian, the topic is dealt with more comprehensively in Cerbo et al. (166).

Immunocyte receptors are not exactly the same as CNS drug receptors ( 1 67- 169), and different types of interaction are possible between the same molecules in the immune versus nervous circuitry. An example of this is that lympho- cytes process the ACTH-beta-endorphin precursor differently than do the pituitary and the hypothalamus (170). Whereas CNS activity of pro-opiomelanocortin fragments can be uni- directional, immunomodulating effects can be bidirectional ( 17 1). To account for bidirectional effects of opioids on immu- nity, a receptor-specific model has been proposed (172). Fur- thermore, local autocrine phenomena are likely to play as important a role as do long and short feedback loops, in the sense that double servomechanisms are available at a hypo- thalamic and pituitary level. For example, IL-1 effects on ACTH secretion can be mediated either tonically directly at the pituitary (173) or phasically, and more rapidly, at the hypo- thalamus (174), probably through a neuronal IL-lergic path- way (174,175).

Psychoneuroimmunology and Psychiatric Disease

Schizophrenia

Immunological abnormalities in schizophrenia were believed to exist as early as the late 19th century, but the matter was approached more systematically in the 1950s and 1960s [see DeLisi (1 76) for a review]. Technological progress has con- tributed to the understanding of immune function disturbance in schizophrenia. Initial studies revealed an increase in atyp- ical circulating lymphocyte numbers (177,178), found later

28 Biondi and Kotzalidis

binding studies showed an increase in the lymphocyte bind- ing sites in schizophrenic patients (204,205), but whether this represents upregulation of true dopamine receptors is a mat- ter of speculation.

The putative immunological alterations of schizophrenia are summarized in Table 3. Taken together, these data sug- gest that something is wrong with immunity in schizophre- nics, but individual variability and disease heterogeneity prevent the picture from being clear. This result is similar to responses to various neuroendocrine challenges, which can be abnor- mal in many ways in the same population, thus yielding no consistent pattern. It is possible that immunity and responses to neuroendocrine challenges are related somehow. Evidence for immune ego disintegration is lacking or incomplete.

Depression

Depression is the psychiatric condition that has been most thoroughly investigated from the immunological standpoint. Depressed patients show decreased circulating lymphocyte numbers (206-210) but no B/T cell (206) or helper/suppressor ratio differences (210,211) compared with controls. Neutro- phi1 counts tend to be elevated in depressives (207) and man- ics (212).

Depressed patients show decreased lymphocyte prolifera- tion responses to all mitogens (206,2 1 1,2 13-2 17). Major depressives had significantly lower mitogenic responses than minor depressives (2 17). However, caution is needed in inter- preting these results unidirectionally (208,218,219); mitogenic responses are not always different from those of controls (2 18), and inpatients behave differently from outpatients (206,208). Manic patients showed decreased responses to PHA and con-A but not to PKW, a result consistent with a T cell impairment (220).

The somewhat increased incidence of autoantibodies in depressives is nonspecific (221,222). It is interesting that an IgG directed against beta-endorphin has been detected in the plasma of depressive patients more frequently than in healthy controls (222), but the significance of this finding is pres- ently unclear.

Given the high frequency of altered dexamethasone-induced cortisol suppression and blunted TSH response to TRH in depressed patients, whether the immunological abnormali-

TABLE 3. Putative Immunological Abnormalities in SchizoDhrenia

Probable Decreased NK and phagic activity Abnormal helper (inducer)/killer (suppressor) ratio

Decreased suppressor (OKT8 + ) number Increased B cell count Upregulation of dopamine high-affinity binding sites Aberrant immunocytic cooperation

Possible

ties found in depression correlated with neuroendocrine impairment has been investigated. Results concerning the HPA axis were negative. Plasma cortisol (215,223) and the dexamethasone suppression test (214,217) did not correlate with immune phenomena. Interestingly, the TRH-stimulated TSH output of monocytes from depressed patients was decreased compared with that from normal controls in a recent study (224).

Acute depressive illness induces lymphocyte glucocorticoid receptor downregulation (225); however, this effect is subject to tolerance (226). Recovered patients are found to have normal lymphocyte glucocorticoid receptors (227). Beta-adrenergic receptors were found to be decreased on lymphocytes from unipolar and bipolar patients (228,229), unchanged on lym- phoblasts from bipolars (230), and with decreased affinity in monocytes from endogenous depressives (23 1). Mimicry or symmetrical behavior with respect to the brain and adapta- tion to stress have been invoked to explain these findings.

Immune abnormalities reported to occur in depression are summarized in Table 4. Globally, depressives tend to have a dull immune responsivity more often than do normals, as in their tendon reflexes or in dealing with life. Since depressed patients are demonstrably more stressed than normals, the lack of correlation between stress and immune indices might mean that there are other pathways of brain activity control- ling the immune response which bypass the HPA and hide the effect of the latter.

The protective role of the pineal in carcinogenesis has been recently demonstrated (232,233). Among pineal products, the peptide epithalamin and the indole melatonin have shown anti- tumor activity (232,234). Melatonin is the hormone that plasma levels are most potently influenced by photoperiod. The normal periodicity of melatonin secretion appears to be protective against tumor growth (235). The role of chrono- biological factors and melatonin in immune modulation is being increasingly recognized (236-239). Recently, the pathophysiology of seasonal affective disorder has been related to melatonin, and a photoperiodically based thera- peutic method has been developed to treat it (240). Since melatonin rhythm affects the sensitivity of cells to stress hor- mones (241), it would be possible to view the relationships among depression, cancer, immunity, stress, pineal, and bio- rhythms in a more integrative way. Chronobiological consid- erations should therefore always be made when dealing with neuroimmunomodulation.

TABLE 4. Putative Immunological Abnormalities in Depression

Probable Decreased circulating lymphocytes Neutrophilia Decreased response to mitogens Decreased lymphocyte beta-adrenoceptors

Decreased TRH-induced lymphocytic TSH release Possible

Human Psychoneuroimmunology 29

tion time is extremely difficult; considerable shifts between the endocrine and immune consequences of psychoneural events are possible. Such difficulties are increased in human stress research.

Other Psychiatric Conditions

There have been few studies dealing with the psychoneuro- immune aspects of other psychiatric conditions. One study found decreased polymorphonuclear and monocytic phago- cytosis in phobic patients (242), while another showed autis- tic children not to have more antibrain antibodies than depressed subjects or normal controls (243). Lymphocytic beta-adrenoceptors were found to be decreased in patients with panic disorders (244) and of increased responsiveness, unre- lated to high plasma norepinephrine levels, in bulimic women (245). These results suggest that noradrenergic regulation is altered in these disorders (244,245). At least in panic, this alteration bears no relationship to depression (244). That addic- tion to opiates is mediated partly through an immune response (145,246,247) is a suggestive proposal that needs to be experi- mentally tested in humans.

Psychoneuroimmunology and Personality

Studies involving the evaluation of personality parameters in relation to immune parameters have been scanty and meth- odologically imperfect. Two studies investigated only sali- vary IgA levels, finding them to be decreased in chronically frustrated power-motivated males (64) and stressed high- internal-locus-of-control individuals (248). Two other stud- ies attempted to correlate MMPI parameters to immunological parameters; one used an MMPI special allergy scale but no laboratory parameter and found a positive correlation with both hypomania and schizophrenia scales (249). The other (250) investigated only NK cell function in healthy young adults and found it to correlate inversely with psychopathol- ogy; all MMPI scales except hysteria and social isolation cor- related with NK function.

The validity of current psychoneuroimmunologic data is restricted by methodological limitations. Psychologists, psy- chiatrists, and endocrinologists employ primitive immuno- logical assessment methods, and immunologists use simple and inadequate neuroendocrine tests (25 1). Gross methods are apt to detect bold links, but if such links were present in the neural, the immune, and the endocrine tissues, they would have been found by the less sophisticated technology of the past. It is conceivable that the relationships between the ner- vous and the immune system are subtle, and the effects of each system on the other might not be apparent. It is evident that the two systems are relatively independent, and each can respond to environmental stimuli with no aid from the other. However, these systems exchange information, which can be quantitatively unimportant with regard to a given effect but can have a weight in the presence of a given environmental setting. Alternatively, subliminal information might sensitize a target if contact is permitted with sufficient frequency, kin- dling the effect after some time. The corollaries of these con- siderations are that a large number of variables should be taken into account in designing a study and that deciding evalua-

THE FUTURE

The data now available through clinical and basic research have established psychoneuroimmunology as a separate domain. Data are now being gathered in lymphocyte subset alterations, in NK cell activity, in lymphocyte response, and in neurotransmitter and peptide binding. These data are from disease groups such as the anxiety disorders, the affective disorders, and the psychoses. It is possible that such disease groups will eventually be subclassed on immunological grounds or that distinct diseases within a group will present distinctive and characteristic immunological features.

It is hoped that studies on the effects of psychopharmaco- logical agents on immune function will be performed more intensely and systematically in the future to cover all drug classes used in psychiatric practice. The evidence for the existence of a benzodiazepine receptor on the monocyte plasma membrane (252) should spark research for benzo- diazepine agonist and antagonist effects on immune respon- sivity, but the other drug classes should by no means be overlooked, since receptors known to be affected by their action are found on lymphocytes. It is to be expected that studies investigating psychoactive drug effects on immunity will have a major impact on drug selection and on preclinical screening of future drugs.

The involvement of other than pro-opiomelanocortin- derived peptides in immune regulation has been recently rec- ognized (122,253,254). This should prompt the assessment of immunocyte binding of all peptides of the brain-gut axis, especially the most recently discovered (for example, galanin- and diazepam-binding inhibitor). Conversely, lymphocytes should be assessed for neuroendocrine peptide production; the new DNA cloning techniques may help this assessment. Should these assessments give positive results, than some new lymphocyte subpopulations could be seen to be part of the integrated diffuse neuroendocrine system. The endocrine nature of some thymic cells has been known for some time (255) , but only recently has it been understood that neuro- hormone-secreting cells of the diffuse neuroendocrine sys- tem reside within the thymus (256). In parallel, the map of brain distribution of immune system-related molecules will eventually be completed and their physiology at that site elu- cidated. Furthermore, in view of recent evidence (257), the relationship between the immune system and platelets should be investigated, so that the mass of data on platelet markers of psychiatric disease can be integrated within a psycho- neuroimmunologic frame.

The relationship of personality to patterns of immune responsiveness will be another issue to settle. Methods of

30 Biondi and Kotzalidis

assessing personality other than the MMPI should be tested in future studies, and the immune parameters to correlate to personality parameters should take into account the progress in immune biology. This is a field in which immunogenetics and behavior genetics may establish some changes.

Also in the future, the psycho-neuroendocrine-immune ring will be closing. It will only be a matter of time, as research is already entrained in this direction. The only differences will be in the extent of the explored domain, in the sense that some more neglected areas will receive due consideration. Psychosomatic disease treatment will undoubtedly benefit from immune function monitoring, and psychotherapeutic techniques will influence the course of diseases associated with immunological impairment in a more specific way than has been possible up to now. It is worth mentioning that new psychotherapeutic techniques are already being tried in the treatment of cancer; these techniques take into account an immunological basis for the pathogenesis of cancer (258). Furthermore, interesting parallels will be made re- garding immune impairment and schizophrenia and psycho- somatic disorders.

It has been hypothesized that schizophrenia could be asso- ciated with left-handedness (259), but conclusive evidence is lacking (260), although an increased frequency of left- handedness was found in schizophrenia with a particular clin- ical course (261). Likewise, left-handedness was found more frequently in immune disorders (53,262,263), and differen- tial interhemispheric regulation of immune parameters has been shown in the mouse cerebral cortex (264-269). Abnor- mal lateralization in schizophrenic patients involves increased oxygen consumption in the left brain (270) and is consistent with a relative increase in dopaminergic function within right rostra1 cortical or subcortical structures (271). It is important for neurons to be present and functioning in adequate num- bers and at an adequate firing rate, but what is more impor- tant is that they establish the right contacts; otherwise, the processing of information leads to faulty and multiple meanings-and this is one of the things that happen in schiz- ophrenics. In the mouse, left frontal and prefrontal cortex lesions decrease and right symmetric lesions increase prolif- eration responses to mitogens and NK cell activity (264-268). Right parietal and occipital lobe lesions induce decreased mito- gen proliferation responses, whereas bilateral lesions are nec- essary to decrease suppressor function (269).

Increased prefrontal and limbic dopaminergic activity has been postulated to exist in acute schizophrenia (272) and to be localized in the right brain (271). Comparing the effects of cortical lesions in the mouse on immunity with immune dysfunction in schizophrenia (Table 3), we noted that decreased NK and B cell function could be consistent with a relative hypofunction of the left frontal cortex, whereas decreased (1 82) or increased (180) suppressor function may be due to impaired left-to-right hemisphere cooperation, should cortex in humans modulate immunity the same way it does in the mouse. Dis-

crepancy in suppressor cell evaluation might be due either to differences in patient populations (176) or to differences in laboratory methods (182,273). It would be interesting to assess the role of dopamine in the modulation of immunity in mouse paradigms. An eventual confirmation that pathological alter- ations in the CNS responsible for schizophrenic symptom- atology also lead to immune system impairment would leave open the question of how such lesions are produced. It is pos- sible that cortex information processing and immune map dis- tribution (similar to that of somatotopic sensory and motor maps) are both altered in schizophrenia due to a basic com- mon defect more caudad in the brain.

What seems to be most exciting in future developments in psychoneuroimmunology is not the hope to cure the depressed with immune enhancers or the cancer patient with psycho- therapy. Such utopistic trends are not considered by even the most optimistic workers in the field of psychoimmunology. Instead, excitement focuses on the conceptual impact of three apparently unrelated medical disciplines. Endocrinology, psy- chiatry, and immunology focus on systems dealing with com- munication, with translation of externally derived stimuli into information bits for internal system use. The similarities of functioning, apart from molecular homologies, are not many, but the analogies are striking. Such analogies have long been recognized [for a review, see Jankovic (274)], and their inter- disciplinary and cybernetic implications are now being increas- ingly recognized.

The fact that neural cell adhesion molecules (N-CAMS) contributing to neuron morphology are homologous to Igs does not mean that neurons and lymphocytes are closer rela- tives than other bodily cells (275), but rather that they used a common molecule with morphogenetic properties to achieve their features and, subsequently, to respond to environmen- tal challenge. Both use an early narcissistic pattern to ego development, to use psychological terms, and use parts of it in a subsequent mature interaction with the outside world. Both make images of the external world by using their own counterimages, demonstrating themselves to be strongly autopoietic. These two useful metaphors, which are based on one biological observation, could be of heuristic value when trying to extrapolate on system development and orga- nization. Recently, an immunological metaphor has been used to better understand combat stress reactions (276).

Other lessons we may be taught by studying closely the neuroendocrine and immune reactions to external challenge are relevant to the process of learning. Learning capacity is the property of a system to respond to a stimulus and thus to reduce the entropic impact of the stimulus or to enhance its organizing properties; learning is demonstrated when the system changes its behavior when faced with the same stim- ulus at different times in such a way that either its response is more effective or quicker. To demonstrate learning, a system should have memory of the stimulus, that is, a trace of it stored within the system. Learning and memory are proper-

Human Psychoneuroimmunology 31

4. Solomon GF, MOOS RH: Emotions, immunity, and disease. A specu- lative theoretical integration. Arch Gen Psychiatr I 1:657-674, 1964.

5. Ader R (ed.): Psychoneuroimmunology. Academic Press, New York, 1981.

6. Solomon GF, Amkraut AA: Psychoneuroendocrinological effects of the immune response. AnnRev Microbiol35:155-184, 1981.

7. Besedovsky H, del Rey A, Sorkin E, Dinarello CA: lmmunoregulatory feedback between interleukin- 1 and glucocorticoid hormones. Science 233:652-654, 1986.

8. Sapolsky R, Rivier C, Yamamoto G, Plotsky P, Vale W: Interleukin- I stimulates the secretion of hypothalamic corticotropin-releasing fac- tor. Science 238522-524, 1987.

9. Berkenbosch F, van Oers J, del Rey A, Tilders F, Besedovsky H: Corticotropin-releasing factor-producing neurons in the rat activated by interleukin-1. Science 238524-528, 1987.

10. Rettori V, Jurkovicova J, McCann SM: Central action of interleukin-1 in altering the release of TSH, growth hormone, and prolactin in the male rat. JNeurosciRes 18:179-183, 1987.

1 1 . Bulloch K: Neuroanatomy of lymphoid tissue: A review. In Neural Modulation of Immunity, R Guillemin, M Cohn, T Melnechuk, eds. Raven Press, New York, 1985, pp 1 1 1-141.

12. Felten DL, Felten SY, Carlson SL, Olschowka JA, Livnat S: Noradrenergic and peptidergic innervation of lymphoid tissue. J Immunol I3S(suppl):755S-765S, 1985.

13. Bulloch K, Cullen MR, Schwartz RH, Longo DL: Development of innervation within syngeneic thymus tissue transplanted under the kid- ney capsule of the nude mouse: A light and ultrastructural microscope study. JNeurosciRes 18:16-27, 1987.

14. Felten DL, Ackerman KD, Wiegand SJ, Felten SY: Noradrenergic sym- pathetic innervation of the spleen: I . Nerve fibers associate with lym- phocytes and macrophages in specific compartments of the splenic white pulp. JNeurosciRes 18:28-36, 1987.

15. Felten SY, Olschowka J: Noradrenergic sympathetic innervation of the spleen: 11. Tyrosine hydroxylase (TH)-positive nerve terminals form synapticlike contacts on lymphocytes in the splenic white pulp. J Neurosci Res 18:37-48, 1987.

16. Ackerman KD, Felten SY, Bellinger DL, Felten DL: Noradrenergic sympathetic innervation of the spleen: 111. Development of innerva- tion in the rat spleen. JNeurosci Res 18:49-54, 1987.

17. Smith EM, Blalock JE: Human lymphocyte production of corticotro- pin and endorphin-like substances: Association with leukocyte inter- feron. Proc NutlAcadSci USA 18:7530-7535, 1981.

18. Lolait SJ, Clements JA, Marwick AJ, Cheng C, McNally M, Smith AI, Funder JW: Pro-opiomelanocortin messenger ribonucleic acid and posttranslational processing of beta endorphin in spleen macrophages. JClinlnvest 77:1716-1779, 1986.

19. Martin J, Prystowsky MB. Angeletti RH: Preproenkephalin mRNA in T-cells, macrophages, andmastcells. J NeurosciRes 18:82-87, 1987.

20. Wybran J: Enkephalins, endorphins. substance P, and the immune sys- tem. In Neural Modulation of Immunity, R Guillemin, M Cohn, T Melnechuk, eds. Raven Press, New York, 1985, pp 157-161.

21. Stein M: Bereavement, depression, stress, and immunity. In Neural Modularion oflmmuniy, R Guillemin, M Cohn, T Melnechuk, eds. Raven Press, New York, 1985, pp 29-44.

22. Selye H.: Stress. Acta, Montreal, 1950. 23. Monjan AA: Stress and immunologic competence: Studies in animals.

In Ps?choneuroimmunologv, R Ader, ed. Academic Press, New York,

24. Palmblad J: Stress and immunologic competence: Studies in man. In: Psychoneuroimmunology, R Ader, ed. Academic Press, New York.

25. Biondi M, Pancheri P: Mind and immunity. A review of methodology in human research. In Research Paradigms in Psychosomaric Medi- cine, GA Fava, TN Wise, eds. Karger, Basel, 1987, pp 234-25 I.

1981, pp 185-228.

1981, pp 229-256.

ties of living and nonliving matter as well, but in the nervous and immune system they show particular characteristics because of the potential novelty of the stimulation these sys- tems are apt to receive and the dynamic nature of the within- system interactions.

Neural learning as related to the immune response has been studied systematically in various conditioning paradigms; behaviorally conditioned immunosuppression (277-282) and immunoenhancement (283,284) have been demonstrated. The mechanism remains obscure (282), and there is still consid- erable debate on the nature of the phenomenon (251). How- ever, conditioning paradigms have been used to conceptualize defensive phenomena, both neural and immune, as system- specific expressions of the same adaptive process (285).

Immunocytes seek each other when they have something to communicate, and they establish strict relationships until the message is received; neurons send their axons to estab- lish synapses with appropriate neurons, and they maintain the synapse as long as it is useful. Once the message passes to the receiver of information, it gives rise to substance buildup that promotes responne to the environment (antibody forma- tion, action); this substance is part of the memory and pro- motes a quicker response upon restimulation (secondary immune response, better performance). The molecules in- volved in intracellular message formation and storage in the two systems can be identical, thus pointing to an interesting parallel between nervous and immune system functions (286).

The learning phenomenon associated with sensitization can lead in the nervous system to the phenomenon of kindling (287). This phenomenon has been used as a model of epi- lepsy (288,289), alcoholic withdrawal syndrome (290), depres- sion (291), psychoses (292), and recovery from long-term illness (293) and involves alterations in plasticity (294). Kin- dling is a mechanism whereby biological organisms adapt in the long run and in a stepwise manner, and it is present in nature more than it is actually realized. Autoimmune disease and cancer could be good candidates to test a kindling model, and the influence of psychosocial factors, particularly stress- ful events, should be investigated.

The above are but a few of the possible paradigms that can be transferred from the immune domain to the neural one and vice versa. We believe that as data from animal and human studies accumulate, conceptual frameworks of psychoneuro- immunology and the contributing disciplines will continue to change gradually. Sudden transformations will take place when the data are be sufficient to catalyze progress in scien- tific thinking.

REFERENCES

1. lshigami T: The influence of psychic acts on the progress of pulmo-

2. Dameshek W: The white blood cells in dementia precox and demen-

3 . Selye H: The Stress ofLife. Longmans, Green, London, 1956, p 1 15.

nary tuberculosis. Am Rev Tuberculosis 2:470-484, 1918-1919.

tia paralytica. Arch Neurol Psychiarr 2:363, 1930.

32 Biondi and Kotzalidis

26. Bartrop RW, Lazarus L, Luckhurst E, Kiloh LG, Penny R: Depressed lymphocyte function after bereavement. Lancet 1:834-836, 1977.

27. Schleifer SJ, Keller SE, Camerino M, Thornton JC, Stein M: Sup- pression of lymphocyte stimulation following bereavement. JAMA 250:374-377, 1983.

28. Irwin MR, Risch SC, Daniels M, Bloom E, Weiner H: Plasma corti- sol and immune function in bereavement (abstr). Psychosom Med 49:210-211, 1987.

29. Kiecolt-Glaser JK, Kennedy S , Malkoff S , Fisher L, Speicher CE, Glaser R: Marital discord and immunity in males. Psychosom Med

30. Kiecolt-Glaser JK, Fisher L, Ogrocki P, Stout JC, Speicher CE, Gla- ser R: Marital quality, marital disruption, and immune function. P.cychosom Med 9: 13-34, 1987.

31, McClelland DC, Ross G, Patel V: The effect of an academic exami- nation on salivary norepinephrine and immunoglobulin levels. J Human Stress 11:42-59, 1985.

32. Jemmott JB 111, Borysenko JZ, Borysenko M, McClelland DC, Chap- man R , Meyer D, Benson H: Academic stress, power motivation, and decrease in secretion rate of salivary secretory immunoglobulin A. Lan- cet 1:1400-1402, 1983.

33. Kiecolt-Glaser JK, Gamer W, Speicher C, Penn GM, Holliday J , Gla- ser R: Psychosocial modifiers of imrnunocompetence in medical stu- dents. Psychosom Med 46:7-14, 1984.

34. Kiecolt-Glaser JK, Glaser R, Strain EC, Stout JC, Tarr KL, Holliday JE, Speicher CE: Modulation of cellular immunity in medical students. J Behav Med 95-21. 1986.

35. Glaser R, Rice J , Speicher CE, Stout JC, Kiecolt-Glaser JK: Stress depresses interferon production by leukocytes concomitant with a decrease in natural killer cell activity. Behav Neurosci 100:675-678, 1986.

36. Halvorsen R, Vassend 0: Effects of examination stress on some cellu- lar immunity functions. J Psychmom Res 3 I :693-701, 1987.

37. Linn BS, Linn MW: The effects of psychophysical stress on surgical outcome (abstr). Psychosom Med 49:210, 1987.

38. Arnetz BB, Wasserman J , Petrini B, Brenner S-0, Levi L, Eneroth P, Salovaara H, Hjelm R, Salovaara L, Theorell T, Petterson I-L: Immune function in unemployed women. Psychom Med 49:3- 12, 1987.

39. Taylor GR, Neale LS, Dardano JR: Immunological analyses of U.S. space shuttle crewmembers. Aviat Space Environ Med 57:213-217, 1986.

40. Baker GHB, Byrom NA, lrani MS, Brewerton DA, Hobbs JR, Wood RJ, Nagvekar NM: Stress, cortisol, and lymphocyte subpopulations. Loncet 1574, 1984.

41. Blasdell K, Mills PJ, Wallace RK, Schneider H: lmmune response in type A and type B behavior after stress and rest (abstr). Psychosom Med49:21 I, 1987.

42. Gruchow HW: Catecholamine activity and infectious disease episodes. J Human Stress 5( 3): 1 1 - 1 7, 1 979.

43. Irwin M, Daniels M, Bloom ET, Weiner H: Life events, depression, and natural killer cell activity. Psychopharmacol Bull 22: 1093- 1096, 1986.

44. Locke S. Kraus L, Leserman J , Hunt MW, Heisel JS, Williams RM: Life change stress, psychiatric symptoms, and natural killer cell activ- ity. Psychosom Med 46441-453, 1984.

45. Pancheri P, Falaschi P, Maione-Marchini A, Rocco A, Zibellini M, Di Cesare G: Emotional and hormonal response to sexually arousing stim- uli. In Biorhythms and Stress in the Physiopathology of Reproduction, P Pancheri, L Zichella, eds. Hemisphere Publishing, New York, 1988,

46. Biondi M, Conti C, Pancheri P, Sega FM, Sega E Reattiviti emozionale e reattivita immunitaria: studio preliminare in pazienti in situazione di stress pre-chirurgico. Rivisfa Psichiarr 16:378-384, 1981.

47. Biondi M, Pancheri P, Cotugno A: Personalita e coping styles nella resposta immunitaria dei linfociti T in condizioni di stress emozionale.

50~213-229, 1988.

~ ~ 4 0 1 - 4 3 4 .

In Stress, Emozioni e Cancro, P Pancheri, M Biondi, eds. II Pensiero Scientific0 Editore, Rome, 1987, pp 95-102.

48. Kiecolt-Glaser JK, Ricker D, George J , Messick G, Speicher CE, Gar- ner W, Glaser R: Urinary cortisol levels, cellular immunocompetency, and loneliness in psychiatric inpatients. Psychosom Med 46: 15-23, 1984.

49. Glaser R. Kiecolt-Glaser JK, Speicher CE, Holliday JE: Stress, loneliness, and changes in herpesvirus latency. J Behav Med 8: 249-260.

SO. Green RG, Green ML: Immunoenhancement: A comparison of four relaxation methods (abstr). Psychosom Med 48:304, 1986.

51. Mills PJ, Schneider R, Hill D, Walton K, Wallace RK: Lymphocyte beta-adrenergic receptors and cardiovascular responsivity in TM par- ticipants and type A behavior (abstr). Psychosom Med49:211, 1987.

52. Smith AP, Tyrrell DAJ, Al-Nakib W, Coyle KB, Donovan CB, Hig- gins PG, Willman JS: Effects of experimentally induced respiratory virus infections and illness on psychomotor performance. Neuropsy- chobiology 18:144-148, 1987.

53. Behan PO, Geschwind N: Hemispheric laterality and immunity. In Neural Modularion oflmmuniry, R Guillemin, M Cohn, T Melnechuk, eds. Raven Press, New York, 1985, pp 73-80.

54. Biziere K, Guillaumin JM, Degenne D, Bardos P, Renoux M , Renoux G : Lateralized neocortical modulation of the T-cell lineage. In Neural Modulation ofImmunity, R Guillemin, M Cohn, T Melnechuk, eds. Raven Press, New York, 1985, pp 81-94.

5 5 . Roszman TL, Cross RJ , Brooks WH, Markesbery WR: Neuroim- munomodulation: Effects of neural lesions on cellular immunity. In Neural Modulation of Immunify, R Guillemin, M Cohn, T Melnechuk, eds. Raven Press, New York, 1985, pp 95-109.

56. Belluardo N, Mud6 G , Cella S, Santoni A , Forni G, Bindoni M: Hypothalamic control of certain aspects of natural immunity in the mouse. Immunology 62:321-327, 1987.

57. Brown WA, Shrivastava R K , Arato M: Pretreatment pituitary- adrenocortical status and placebo response in depression. Psycho- pharmacol Bull 23:155-159, 1987.

58. Coryell W, Nesse R: Placebo response in panic disorder. Am J Psychiatr 145:1138-1140, 1988.

59. Peselow ED, Lautin A, Wolkin A, Rohrs C, Novatt A, Siekierski J , Rotrosen J: The dexamethasone suppression test (DST) and response to placebo. J Clin Psychopharmacol6:286-29 I , 1986.

60. Plaut SM, Friedman SB: Psychosocial factors in infectious diseases. In PsychoneuroirnmunoloRy, R Ader, ed. Academic Press, New York, 1981,pp3-30.

61. Biondi M: Psicoimmunologia. In Trattato di Medicina Psicosomarica, P Pancheri. ed. USES, Florence, 1984.

62. Biondi M: Psiconeuroimmunologia: Dati sperimentali e impliczioni cliniche. In Progressi in Psichiatria, Vol. 2 , Parte I : Psichiatria e Medicina, P Pancheri, GC Reda, eds. Edizioni Internazionali, Rome, 1989, pp 3-18,

63. Mason JW, Buescher EL, Belfer ML, Artenstein MS, Mougey EH: A prospective study of corticosteroid and catecholamine levels in rela- tion to viral respiratory illness. J Human Stress S(3) : 18-28, 1979.

64. McClelland DC, Floor E, Davidson RJ, Saron C: Stressed power moti- vation, sympathetic activation, immune function, and illness. J Human Stress6(2):11-19, 1980.

65. Calabrese JR, Kling MA, Gold PW: Alterations in immunocompe- tence during stress, bereavement, and depression: Focus on neuroen- docrine regulation. Am J Psychiatr 144:1123-1134, 1987.

66. Allen AD: Creation, evolution and biological psychiatry (lett). Biol Psychiutr 23:209, 1988.

67. Pedersen BK, Beyer JM: Characterization of the in vitro effects of glucocorticosteroids on NK cell activity. Allergy 41 :220-224, 1986.

68. Dorian BJ, Garfinkel PE, Keystone EC, Gorczynski R, Darby P: Stress, immunity and illness (abstr). Psychosom Med 48:304-305, 1986.

Human Psychoneuroimmunology 33

92. Babigian HM: Schizophrenia: Epidemiology. In Comprehensive Text- book of Psychiatry, HI Kaplan, AM Freedman, BJ Sadock, eds. Wil- liams & Wilkins, Baltimore, 1980, pp l 113-1 121.

93. Lehman HE: Schizophrenia: Clinical features. In Comprehensive Text- book of Psychiatry, HI Kaplan, AM Freedman, BJ Sadock, eds. Wil- liams & Wilkins, Baltimore, 1980, pp 1153-1 192.

94. Baldwin JA: Schizophrenia and physical disease. Psycho1 Med 9:611-618, 1979.

95. Bahnson CB, Bahnson MB: Role of ego defenses: Denial and repres- sion in the etiology of malignant neoplasm. Ann NY Acad Sci 164546-577, 1969.

96. Biondi M, Pancheri P: Stress, personality, immunity, and cancer: A challenge for psychosomatic medicine. In Behavioral Epidemiology and Disease Prevention. RM Kaplan, MH Criqui, eds. Plenum Press, New York. 1985, pp 271-298.

97. Coppen A, Metcalfe M: Cancer and extraversion. Br Med J 2: 18-19, 1963.

98. Hagnell 0: The premorbid personality of persons who develop cancer in a total population investigated in 1947 and 1957. Ann NYAcad Sci 125:846-855, 1966.

99. Greer S, Morris T: Psychological attributes of women who develop breast cancer. Acontrolledstudy. JPsvchosom Res 19:147-153, 1975.

100. Ternoshock L: Emotion, adaptation, and disease. In Emotions in Health and Illness. Theoretical Research Foundations, L Temoshock, C Van Dyke, LA Zegans, eds. Gmne & Stratton, New York, 1983, pp 207-234.

101. Greer S . Watson M: Towards a psychobiological model of cancer: Psy- chological considerations. Soc Sci Med 20773-777, 1985.

102. Baltrusch H-JF, Seidel J, Stangel W, Waltz ME: Psychosocial stress, agingandcancer. Ann NYAcadSci521:1-15, 1988.

103. Greer S. Morris T, Pettingale K, Haybittle J: Mental attitude to can- cer. Lancet l:750, 1985.

104. Greer S, Morris T, Pertingale K: Psychological response to breast can- cer: Effect on outcome. Lancet 2:785-787, 1979.

105. Levy SM: Behavior and Cancer. Life-Sfyle and Psychosocial Factors in the Initiation and Progression of Cancer. Jossey-Bass, San Fran- cisco, 1985, pp 188-189.

106. Gurney ME, Heinrich SP, Lee MR, Yin H-s: Molecular cloning and the expression of neuroleukin, a neurotrophic factor for spinal and sen- sory neurons. Science 234566-574, 1986.

107. Lee MR, Ho DD, Gurney ME: Functional interaction and partial homol- ogy between human immunodeficiency virus and neuroleukin. Science 237: 1047- I05 I , 1987.

108. Ruff RM, Martin BM, Ginns El, Farrar WL, Pert CB: CD receptor binding peptides that block HIV infectivity cause human monocyte chemotaxis: Relationship to vasoactive intestinal polypeptide. FEBS Letf211:17-22, 1987.

109. Sacerdote P, Ruff MR, Pert CB: VIP 1-12 is a ligand for the CD4 human immunodeficiency virus receptor. Ann NY Acad Sci 527574-578. 1988.

110. Sacerdote P, Ruff MR, Pert CB: Vasoactive intestinal peptide 1-12: A ligand for the CD4 (T4)ihuman immunodeficiency virus receptor. J NeurosciRes 18:102-107, 1987.

1 11. Pert C, Dienstfrey H: The neuropeptide network. Ann NY Acad Sci 521 : 189-194, 1988.

112. Gurney ME, Apatoff BR, Spear GT, Baumel MJ, Antel JP, Bania MB, Reder AT: Neuroleukin: A lymphokine product of lectin-stimulated T cells. Science 234574-581, 1986.

113. Barnes DM: Brain damage by AIDS under active study. Science 235: 1574- 1577, 1987.

114. Emson PC. Fahrenkrug J, Spokes EGS: Vasoactive intestinal polypep- tide (VIP) distribution in normal human brain and in Huntington’s dis- ease. Brain Res 173:174-178, 1979.

115. Emson PC, Marley PD: Cholecystokinin and vasoactive intestinal poly- peptide. In Handbook of PsvchopharmacoloRy. Vol. 16: Neuropeptides,

69. King MB: Psychosocial status of 192 out-patients with HIV infection and AIDS. Br J Psychiatr 154:237-242, 1987.

70. Bridge TP, Mirsky AF, Goodwin FK, eds. Psychological Neuropsy chiatric. and Substance Abuse Aspects of Aids. Raven Press, New York, 1988.

71. Lewis JW, Shavit Y. Terman GW, Nelson LR, Martin FC. Gale RP, Liebeskind JC: Involvement of opioid peptides in the analgesic, immu- nosuppressive, and tumor-enhancing effects of stress. Psychopharmacol Bull 21:479-464, 1985.

72. Riley V: Psychoneuroendocrine influences on immunocompetence and neoplasia. Science 212:llOO-1109, 1981.

73. Glaser R, Thorn BE, Tarr KL: Effects of stress on methyltransferase synthesis: An important DNA repair enzyme. Health P.yvcho14:403-412, 1985.

74. Kiecolt-Glaser JK, Stephens RE, Lipetz PD, Speicher CE, Glaser R: Distress and DNA repair in human lymphocytes. J Behav Med 8:311-320, 1985.

75. Lehrer S: Life change and lung cancer. J Human Stress 7(4):7- I I , 198 I . 76. Goodkin K , Antoni MH, Blaney PH: Stress and hopelessness in the

promotion of cervical intraepithelial neoplasia to invasive squarnous cell carcinoma of the cervix. J Psychosom Res 30:67-76, 1986.

77. Shekelle RB, Raynor WJ Jr, Ostfeld AM, Garron DC, Bieliauskas LA, Liu SC, Maliza C, Paul 0: Psychological depression and 17-year risk of death from cancer. Psychosom Med 43: I 17- 125, I98 I .

78. Niemi T, Jaaskelainen J: Cancer morbidity in depressive persons. J Psychosom Res 22: 117-120, 1978.

79. Greenberg RP, Dattore PJ: The relationship between dependency and the development of cancer. Psychosom Med43:35-43, 1981.

80. Kiecolt-Glaser JK, Glaser R: Psychological influences on immunity. Psychosomatics 27:621-624, 1986.

8 I . Eysenck HJ: Anxiety, learned helplessness, and cancer: A causal the- ory. JAnxDisord 1:87-104, 1987.

82. Schindler BA: Stress, affective disorders. and immune function. Med Clin North Am 69585-597, 1985.

83. Stein M, Schleifer SJ. Keller SE: Stress, depression and immunity. In Neuroregulation of Autonomic, Endocrine and Immune Svsrems, RCA Frederickson, HC Hendrie, J N Hingtgen, MH Aprison, eds. Martinus Nijhoff, Boston, 1986, pp 367-388.

84. Croiset G, Heijnen CJ, Veldhuis HD, de Wied D, Ballieux RE: Mod- ulation of the immune response by emotional stress. Life Sci 40: 775-782, 1987.

85. Sklar LS, Bruto V, Anisman H: Adaptation to the tumor-enhancing effects of stress. Psychosom Med43:331-342, 1981.

86. Creed F: Life events and physical illness. J Psychosom Res 29: 1 13- 123, 1985.

87. Kaplan HI, Knapp PH, Cheren S, Stunkard AJ, Halmi KA, Lipowski z1, Weiner H, Reiser MF, Whisnant L, Engels WD, Wittkower ED, Silverman AJ, Joseph ED, Schwartz AH, Frazier SH, Stein M, Schleifer S, Keller S , Enelow AJ: Psychologic factors affecting physical condi- tions (psychosomatic disorders). In Comprehensive Textbook of P s y chiatry, HI Kaplan, AM Freedman, BJ Sadock, eds. Williams & Wilkins, Baltimore, 1980, 1843-1985.

88. Kerr TA, Schapira K , Roth Sir M: The relationship between pre-mature death and affective disorders. BrJ Psychiatr 115:1277-1282, 1969.

89. Evans NJR, Baldwin JA, Gath D: The incidence of cancer among in-patients with affective disorders. Br J Psychiarr 124:518-525, 1974.

90. Bahnson CB, Bahnson MB: Cancer as an alternative to psychosis: A theoretical model of somatic and psychological regression. In Psycho- somatic Aspects of Neoplastic Disease, DM Kissen, L LeShan, eds. Pitman, London, 1964, pp41-64.

91, Levi RN, Waxman S: Schizophrenia, epilepsy, cancer, methionine, and folate metabolism pathogenesis of schizophrenia. Lancet 2: I 1 - 14, 1975.

34 Biondi and Kotzalidis

LL Iversen, SD Iversen, SH Snyder, eds. Plenum Press, New York, 1983, pp 235-306.

116. Robberecht P, De Neef P, Lammens M, Deschot-Laakmann M, Christoph JP: Specific binding of vasoactive intestinal peptide to brain membranes from the guinea pig. Eur J Biochem 90:147-154, 1978.

117. Robberecht P, Konig W, Deschodt-Laakmann M, de Neef P, Christoph J: Specificity of receptors to vasoactive intestinal peptide in guinea pig brain. Life Sci 25:879-884, 1979.

118. Gammeltoft S , Staun-Olsen P, Fahrenkrug J , Bartels PD, Ottesen B: Vasoactive intetinal polypeptide specific binding to peripheral synap- tic membranes from rat brain. Regul Pep? [Suppl] I :S40, 1980.

119. Danek A, O’Dorisio MS, O’Dorisio TM, George JM: Specific bind- ing sites for vasoactive intestinal polypeptide on nonadherent periph- eral bloodlymphocytes. JImmunol 131:1l73-1177. 1983.

120. Calvo JR, Molinero P, Jimenez J , Goberna R, Guerrero JM: Interaction of vasoactive intestinal peptide (VIP) with rat lymphoid cells. Pep- tides 7 : 177- 18 I . 1986.

121. Ottaway CA, Bernaerts C, Chan B, Greenberg GR: Specific binding of vasoactive intestinal peptide to human circulating mononuclear cells. Can J Physiol Pharmacol61:664-671, 1983.

122. Roche JK: Immune response modulation by intestinal neuropeptides. Prog Neuroendocrinimmunol 1(2):4-7, 1988.

123. Ottaway CA, Greenberg GR: Interaction of vasoactive intestinal pep- tide with mouse lymphocytes: Specific binding and the modulation of mitogen responses. J Immunol132:417-423, 1984.

124. Ottaway CA: Selective effects of vasoactive intestinal peptide on the mitogenic response ofmurineTcells. Immunology62:291-297, 1987.

125. Drew PA, Shearman JC: Vasoactive intestinal peptide: A neurotrans- mitter which reduces human NK cell activity and increases IgG syn- thesis. Aust J E x p Biol MedSci 63:313-318, 1985.

126. Krco CJ, Gores A, Go VLW: Gastrointestinal regulatory peptides mod- ulate in vitro immune reactions of mouse lymphoid cells. Clin Immunol Immunopathol39:308-318, 1986.

127. Hill JM, Farrar WL, Pert CB: Localization of the T4 antigeniAlDS virus receptor in monkey and rat brain: Prominence in cortical region. Psychopharmacol Bull 22:686-689, 1986.

128. Klatzman D, Champagne E, Chamaret S, Gruest J . Guetard D, Hercend T, Gluckman JC, Montagnier L: T lymphocyte T4 molecule behaves as the receptor for human retrovirus LAV. Nature 312:767-768, 1985.

129. Pert CB, Hill JM, Ruff MR, Bennan RM, Robey WG, Arthur LO, Ruscetti FW, Farrar WL: Octapeptides deduced from the neuropeptide- like pattern of antigen T4 in brain potently inhibit human immunode- ficiency virus receptor binding and T cell infectivity. Proc Nut1 Acad Sci USA 83:9254-9258, 1986.

130. Gjerris A, Rafaelsen OJ: Catecholamines and vasoactive intestinal polypeptide in cerebrospinal fluid in depression. In Frontiers in Bio- chemical and Phurmacologicul Research in Depression, E Usdin, M Asberg, L Bertilsson, F Sjoqvist, eds. Raven Press, New York, 1984, pp 159-160.

13 I . Roberts GW, Ferrier NI, Lee Y, Crow TJ, Johnstone EC, Owens DGC, Bacarese-Hamilton AJ, McGregor G, O’Shaughnessey D, Polak JM, Bloom SR: Peptides, the limbic lobe and schizophrenia. Brain Res 288:199-211, 1983.

132. Henn FA: Neurotransmitter abnormalities in schizophrenia. In Biologi- cal Perspectives of Schizophrenia, H Helmchen, FA Henn, eds. Wiley & Sons, New York, 1987, pp 299-309.

133. Katsuura G , Itoh S: Effects of cholecystokinin and vasoactive intesti- nal peptide on locomotion and rearing in neofrontal decorticated rats. In Endocoids, H Lal, F LaBella, J Lane, eds. Alan R. Liss, Inc., New York, 1985, pp 147-150.

134. Nguyen TD: Peptide T from human immunodeficiency virus envelope does not interact with hepatic, intestinal and colonic vasoactive intes- tinal peptide (VIP) receptors. Peptides 9:425-428, 1988.

135. Schupf N, Williams CA: Effect of immune-complex-forming reactions

on catecholamine-modulated behaviors in rat hypothalamus. J Neuro- immunol9:305-316, 1985.

136. Schupf N, Williams CA: Psychopharmacological activity of immune complexes in rat brain is complement dependent. J Neuroimmunol

137. Koranyi EK, Dewar A: Neuropsychiatric features of drug induced and spontaneous systemic lupus erythematosus. Psychiatr J Univ Ottawa

138. Feinglas EJ, Arnett FC, Dorsch CA, Zizac TM, Sterns MB: Neuro- psychiatric manifestations of systemic lupus erythematosus: Diagno- sis, clinical spectrum and relationship to other features of the disease. Medicine 55:323-339, 1976.

139. Keefe EB, Bardana EJ, Hrbeck BRJ, Pirofsky B, Carr RI: Lupus meningitis-Antibody to DNA and DNA:anti-DNA complexes in cere- brospinal fluid. Ann Intern Med 80:58-60, 1974.

140. Solomon GF, Moos RH, Fessel WJ, Morgan EE: Globulins and behav- ior in schizophrenia. In: J Neuropsychiatr 2:20-26, 1966.

141. Calabrese JR, Gulledge AD, Hahn K, Skwerer R, Kotz M, Schumacher OP, Gupta MK, Clough JD, Krupp NE, Gold PW: Antinuclear anti- bodies and antithyroid antibodies in psychiatric patients. In Viruses, Immunity, and Mental Disorders, E Kurstak, ZI Lipowski, PV Morozov, eds. Plenum Medical, New York, 1987, pp 353-362.

142. Krishnan R, Ellinwood EH Jr, Laszlo J , Hood L, Ritchie J: Effect of gamma interferon on the hypothalamic-pituitary-adrenal system. Biol Psychiatr 22:1163-1166, 1987.

143. Holsboer F, Stalla GK, von Bardeleben U, Hammann K, Mueller H, Mueller OA: Acute adrenocortical stimulation by recombinant gamma interferon in human controls. Life Sci 42: 1-5, 1988.

144. Weinberger SB, Schulteis G, Fernando AG, Bakhit C, Martinez JL Jr: Decreased locomotor activity produced by repeated, but not single, administration of murine-recombinant interferon-gamma in mice. Life Sci42:1085-1090, 1988.

145. Dafny N: Interferon: A candidate as the endogenous substance pre- venting tolerance and dependence to brain opioids. Prog Neuropsy- chopharmacol Biol Psychiatr 8:35 1-357, 1984.

146. Nakashima T, Hori T, Kuriyama K, Kiyohara T: Naloxone blocks the interferon-alpha induced changes in hypothalamic neuronal activity. Neurosci Lett 82:332-336, 1987.

147. Farkkila M, Iivanainen M, Harkonen M, Laakso J , Mattson K, Niiranen A, Lmen TA, Cantell K: Effect on interferon-gamma on biogenic amine metabolism, electroencephalographic recordings, and transient poten- tials. Clin Neuropharmacol 11:63-67, 1988.

148. Rohatiner SZA, Prior PF, Burton AC, Smith AT, Balkwill FR, Lister TA: Central nervous toxicity of interferon. Prog J Cancer47:419-422, 1983.

149. Gockerman JP, Hood L, Bishop C, Huang AT, Triozzi P, Sedwick WD, Koren H, Borowitz H, Tso CY, Laszlo J: Phase I studies of recombi- nant gamma interferon. Proc Am Soc Clin Oncol4:232, 1985.

150. Johnson HM, Farrar WL, Torres BA: Vasopressin replacement of interleukin 2 requirement in gamma interferon production: Lymphokine activity of a neuroendocrine hormone. JImmunol129:983-986, 1982.

151. Johnson HM, Torres BA, Smith EM, Dion LD, Blalock JE: Regula- tion of lymphokine (gamma-interferon) production by corticotropin. J Immunol 132:246-250, 1984.

152. Iivainen M, Laaksonen R, Niemi M-L, Farkkila M, Bergstrom L, Mattson K, Niiranen A, Cantell K: Memory and psychomotor impair- ment following high-dose interferon treatment in amyotrophic lateral sclerosis. Acta Neurol Scand 72:475-480, 1985.

153. McDonald EM, Mann AH, Thomas HC: Interferons as mediators of psychiatric morbidity. An investigation in a trial of recombinant alpha- interferon in hepatitis-B carriers. Lancet 2.1 175-1 178, 1987.

154. Arora PK, Hanna EE, Paul SM, Skolnick P: Suppression of the immune response by benzodiazepine receptor inverse agonists. J Neuroimmunol

13~293-303, 1987.

11:52-57, 1986.

15~1-9, 1987.

Human Psychoneuroimmunology 35

176. DeLisi LE: Immunologic studies of schizophrenic patients. In Viruses, lmmuniry, andMental Disorders, E Kurstak, ZI Lipowski, PV Momzov, eds. Plenum Medical, New York, 1987, pp 271- 183.

177. Kamp HV: Nuclear changes in the white blood of patients with schiz- ophrenic reactions. J Neuropsychiatr 4: 1-3, 1962.

178. Fessel WJ, Hirata-Hibi M: Abnormal leukocytes in schizophrenia. Arch Gen Psychiatr 9:601-613, 1963.

179. DeLisi LE, Ortaldo JR, Maluish AE, Wyatt RJ: Natural killer (NK) cell activity of lymphocytes from schizophrenic patients. J Neur Transm 58:96-106, 1983.

180. DeLisi LE, Goodman S , Necker LM, Wyatt RJ: Lmphocyte subpopu- lations in schizophrenic patients. BiolPsychiatr 17: 1003-1007, 1982.

181. Kolyaskina GI: Blood lymphocytes in schizophrenia: Immunological and virological aspects. In Research on the Viral Hypothesis of Men- talDisorders, PV Morozov, ed. Basel, Karger, 1983, pp 142-149.

182. Bessler H, Eviatar J , Meshulam M, Tyano S, Djaldetti M, Sirota P: Theophylline-sensitive T-lymphocyte subpopulation in schizophrenic patients. Biol Psychiarr 22: 1025-1028, 1987.

183. DeLisi LE, Wyatt RJ: Abnormal immune regulation in schizophrenic patients. Psychopharmacol Bull 18:(4): 158-163, 1982.

184. Vartanian ME, Kolyaskina GI, Lozovsky DV, Burbaeva GS, Ignatov SA: Aspects of humoral and cellular immunity in schizophrenia. In Neurochemical and Immunological Components of Schizophrenia, D Bergsma, AL Goldstein, eds. Alan R. Liss, Inc., New York, 1978,

185. Strahilevitz M, Fleishman JB, Fischer GW. Harris R, Narasimhachari N: Immunoglobulin levels in psychiatric patients. Am J Psychiatr 133:772-776, 1976.

186. Zarrabi MH, Zucker S , Miller F, Derman RM, Romano GS, Hartnett JA, Varma AO: Immunologic and coagulation disorders in chlorprom- azine treated patients. Ann Intern Med 91: 194-199, 1979.

187. Ahokas A, Koskiniemi M-L, Vaheri A, Rimon R: Altered white cell count, protein concentration and oligoclonal IgG bands in the cere- brospinal fluid of many patients with acute psychiatric disorders. Neuropsychobiology 14: 1-4, 1985.

188. DeLisi LE, Weinberger DR, Potkin SG, Neckers LM, Shilling DJ, Wyatt RJ: Quantitative determination of immunoglobulins in CSF and plasmaofchronicschizophrenicpatients. BrJPsychiatr 1395 13-518, 1981.

189. Roos RP, Davis K, Meltzer HY: Immunoglobulin studies in pa- tients with psychiatric diseases. Arch Gen Psychiatr 42: 124-128, 1985.

190. Allansmith M, McClellan B, Butterworth M: The influence of hered- ity and environment on human immunoglobulin levels. J lmmunol 102: 1504- 15 10, 1969.

191. Rimon R, Ahokas A, Ruutiainen J , Halonen P: Myelin basic protein antibodies in catatonic schizophrenia. JClin Psychiatr47:26-28, 1986.

192. Kirchbach A von, Fischer EG, Komhuber HH: Failure to detect dopa- mine receptor Ig autoantibodies in sera of schizophrenic patients. J NeurTransm 70:175-179, 1987.

193. Cappel R, Sprecher S: Are herpes viruses responsible for neuropsy- chiatric diseases? In Research on the Viral Hypothesis of Mental Dis- orders, PV Morozov, ed. Karger, Basel, 1983, pp 168-173.

194. Torrey EF, Yolker RH, Winfrey CJ: Cytomegalovirus antibody in cere- brospinal fluid of schizophrenic patients detected by enzyme immu- noassay. Science 216:892-894, 1982.

195. Crow TJ: A re-evaluation of the viral hypothesis: Is psychosis the result of retroviral integration at a site close to the cerebral dominance gene? BrJPsychiarr 145:243-253, 1984.

196. King DJ, Cooper SJ, Earle JAP, Martin SJ. McFerran NV, Wisdom GB: Serum and CSF antibody titres to seven common viruses in schiz- ophrenic patients. Br JPsychiatr 147: 145- 149, 1985.

197. DeLisi LE, Sarin P: Lack of evidence for retrovirus infection in schizo- phrenia. BrJPsychiatr 146:674, 1985.

pp 339-364.

155. Ruff MR, Pert CB: Human monocyte chemotaxis to neuropeptides. In Hypothalamic Dysfunction in Neuropsychiatric Disorders, D Nerozzi, FK Goodwin, E Costa, eds. Raven Press, New York, 1987, pp 247-260.

156. Ganguli R, Rabin B, Raghu U, Ulrich RS: T lymphocytes in schizo- phrenics and normals and the effects of varying antipsychotic dosage. In Viruses, Immunity, and Mental Disorders, E Kurstak, ZJ Lipowski, PV Morozov, eds. Plenum Medical, New York, 1987. pp 321-326.

157. Kaufmann CA, DeLisi LE, Torrey EF, Folstein SE, Smith WJ: T-lymphocyte subsets and schizophrenia. In Viruses, Immuniry. and Menral Disorders, E Kurstak, ZI Lipowski, PV Morozov, eds. Ple- num Medical, New York, 1987,307-320.

158. Pogady J , Libikova H: Influence upon the immune response and cyto- toxicity of psychotropic drugs. Actiw Nerv Super 24:269-270, 1982.

159. Baker GA, Santalo R, Blumenstein J: Effect of psychotropic agents upon the blastogenic response of human T-lymphocytes. Biol Psychiatr

160. Barbu V, Maziere JC, Maindrault F, Maziere C, Rampini C, Roux C, Polonovski J: Effect of AY 9944 and chlorpromazine on concanavalin A-induced stimulation of human lymphocytes. Biochem Pharmacol 36:353-356, 1987.

161. Lazarus JH, John R, Bennie EH, Chalmers RJ, Crockett G: Lithium therapy and thyroid function: A long-term study. PsycholMed 11:85-92, 1981.

162. Lazarus JH, McGregor AM, Ludgate M, Darke C, Creagh FM, Kingswood CJ: Effect of lithium carbonate therapy on thyroid immune status in manic depressive patients: A prospective study. J Aflective Disord 11:155-160, 1986.

163. Marullo S, Hoebeke J , Guillet JG, Andre C, Strosberg AD: Immuno- logic mimicry by a monoclonal antibody of the tricyclic anti-depressants’ binding site on muscarinic acetylcholine receptors. J Immunol 138524-526, 1987.

164. Bering B, Moises HW, Mueller WE: Muscarinic cholinergic recep- tors on intact human lymphocytes. Properties and subclass character- ization. BiolPsychiatr 22:1451-1458, 1987.

165. Vinar 0, Poch T, Vinarova E: Levamisol counteracts the resistance to antidepressants. Actiw N e w Super 29:239-241, 1987.

166. Cerbo R, Formisano R, Catarci T, Agnoli A: Psicofarmaci e Sistema lmmunirario (Conjnia Cephalalgica. wol. 2). EMI, Pavia, 1987.

167. Stefan0 GB, Zhao X, Bailey D, Metlay M, Leung MK: High affinity dopamine binding to mouse thymocytes and Mytilus edulis (Bivalvia) hemocytes. J Neuroimmunol2 1 :67-74, 1989.

168. Ovadia H, Abramsky 0 : Dopamine receptors on isolated membranes of rat lymphocytes. JNeurosciRes 18:70-74, 1987.

169. Mendelsohn LG, Kerchner GA, Culwell M, Ades E W Immunorrgulation by opioid peptides: I . Absence of classical opioid receptor on human mononuclear cells. J Clin Lab Immunol 16: 125-129, 1985.

170. Harbour DV, Smith EM, Blalock JE: Novel processing pathway for proopiomelanocortin in lymphocytes: Endotoxin induction of a new prohormone-cleaving enzyme. J Neurosci Res 189-101, 1987.

171. McCain HW, Bilotta J , Lamster 1B: Endorphinergic modulation of immune function: potent action of the dipeptide glycyl-L-glutamine. Life Sci 4 1 : 169- 176, 1987.

172. Plotnikoff NP: The ying-yang hypothesis of opioid peptide immuno- modulation. Psychopharmacol Bull 21:489, 1985.

173. Kehrer P, Tumill D, Dayer J-M, Muller AF, Gaillard RC: Human recom- binant interleukin-I -beta and -alpha, but not recombinant tumor necro- sis factor alpha stimulate ACTH release from rat anterior pituitary cells in vitro in a prostaglandin E2 and CAMP independent manner. Neuro- endocrinology48: 160-166, 1988.

174. Tsagarakis S. Gillies G, Rees LH, Besser M, Grossman A: Interleukin-I directly stimulates the release of corticotrophin releasing factor from rat hypothalamus. Neuroendocrinology 49:98-101, 1989.

175. Breder CD, Dinarello CA, Saper CB: Interleukin- I immunoreactive innervation of the human hypothalamus. Science 240:321-324, 1988.

12: 159- 169, 1977.

36 Biondi and Kotzalidis

198. Preble OT, Torrey EF: Serum interferon in patients with psychosis. Am JPsychiatr 142:1184-1186, 1985.

199. Doha FC: Cereals and schizophrenia data and hypothesis. Acta Psychiutr Scand42: 125- 152. 1966.

200. Rix KJB, Ditchfield J , Freed DLJ, Goldberg DP. Hillier VF: Food antibodies in acute psychoses. Psycho1 Med 15:347-354, 1985.

201. Goldin LR, Gershon ES: Association and linkage studies of genetic marker loci in major psychiatric disorders. Psychiarr Dev 4:387-418, 1983.

202. McGuffin P, Festenstein H, Murray R: A family study of HLA anti- gens and other genetic markers in schizophrenia. Psychol Med 13:31-43, 1983.

203. Metzer WS, Newton JEO, Steele RW, Claybrook M, Paige SR, McMil- Ian DE, Hays S: HLA antigens and haplotypes in schizophrenia. Neuropsychiatr Neuropsychol Behav Neurol 1 :39-46. 1988.

204. Rotstein E, Mishra RK, Singal DP, Barone D: Lymphocyte 3H-spiro- peridol binding in schizophrenia: Preliminary findings. Prog Neuro- psyrhopharmacol Biol Psychiatr 7:729-732, 1983.

205. Bondy B, Ackenheil M: 3H-Spiperone binding sites in lymphocytes as possible vulnerability marker in schizophrenia. J Psychiatr Res 2 I52 1-529, 1987.

206. Schleifer SJ, Keller SE. Meyerson AT, Raskin MJ, Davis KL, Stein M: Lymphocyte function in major depressive disorder. Arch Gen Psychiatr 4 1 :484-486, 1984.

207. Kronfol 2, Turner R, Nasrdllah H, Winokur G: Leukocyte regulation indepression and schizophrenia. PsychiatrRes 13:13-18, 1984.

208. Schleifer SJ, Keller SE, Siris SG, Davis KL, Stein M: Depression and immunity. Lymphocyte function in ambulatory depressed patients, hos- pitalized schizophrenic patients, and patients hospitalized for hernior- rhaphy. Arch Gen Psychiatr42: 129-133, 1985.

209. Murphy D, Gardner R, Greden JF, Carroll BJ: Lymphocyte numbers in endogenous depression. Psychol Med 17:381-385, 1987.

210. Kronfol 2, Quinn J , House JD, Nasrallah HA: Immunologic abnor- malities in depressive illness. In Viruses, ImmuniQ, and Mental Dis- orders, E Kurstak, ZJ Lipowski, PV Morozov, eds. Plenum Medical, New York. 1987, pp 363-367.

211. Calabrese JR, Skwerer RG, Barna B, Gulledge AD, Valenzuela R, Butkus A, Subichin S, Krupp NE: Depression, immunocompetence, and prostaglandins of the E series. Psychiatr Res 17:41-47, 1986.

212. Kronfol 2, Turner R, House JD, Winokur G: Elevated blood neutro- phi1 concentration in mania. J Clin Psychiatr 47:63-65, 1986.

213. Kronfol 2, Silva J Jr, Greden J , Dembinski S, Gardner R, Carroll B: Impaired lymphocyte function in depressive illness. Lifi.Sci 33:241-247, 1983.

214. Kronfol 2. House JD: Depression, hypothalamic-pituitary-adreno- cortical activity, and lymphocyte function. Psychopharmacol Bull 21:476-478, 1985.

215. Kronfol 2, House JD, Silava J Jr. Grreden J , Carroll BJ: Depression, urinary free cortisol excretion and lymphocyte function. Br J Psychiarr 148:70-73, 1986.

216. Darko DF, Lucas AH, Gillin JC: Replication of lower lymphocyte blas- togenesis in depression (lett). Am JPsychiatr 143: 1492- 1493, 1986.

217. Cosyns P, Maes M, Vandewoude M, Stevens WJ, De Clerck LS, Schotte C: Impaired mitogen-induced lymphocyte responses and the hypothalamic-pituitary-adrenal axis in depressive disorders. J Afec- rive Disord 16:41-48, 1989.

218. Schleifer SJ, Keller SE, Bond RN, Cohen J, Stein M: Major depres- sive disorder and immunity. Role of age, sex, severity, and hospital- ization. Arch Gen Psychiatr 463-87 , 1989.

219. Denman AM: Immunity anddepression. BrMedJ293:464-465, 1986. 220. Kronfol Z. House JD: Immune function in mania. Biol Psychiatr

221. Irwin H, Daniels M, Weiner H, Bloom E: Depression and changes in 24:34 I -343. 1988.

T cell subpopulations (abstr). Psychosom Med48:303-304, 1986.

222. Roy BF, Rose JW, McFarland HF, McFarlin DE, Murphy DL: Anti- beta-endorphin immunoglobulin G in humans. Proc Nafl AcadSci USA 83:8739-8743. 1986.

223. Syvalahti E, Eskola J , Ruuskanen 0, Laine T: Nonsuppression of cor- tisol in depression and immune function. Prog Neuropsychopharmacol Biol Psvchiatr 9:413-422, 1985.

224. Harbour DV, Anderson A, Farrington J , Wassef A, Smith EM, Meyer WJ 111: Decreased mononuclear leukocyte TSH responsiveness in patients with major depression. BiolPsychiatr 23:797-806, 1988.

225. Gormley GJ, Lowy MT, Reder AG, Hostelhorn VD, Antel JP, Meltzer HY: Glucocorticoid receptors in depression: Relationship to the dexa- methasone suppression test. Am J Psychiatr 142: 1278-1284, 1985.

226. Schlechte JA, Sheman B: Lymphocyte glucocorticoid receptor bind- ing in depressed patients with hypercortisolemia. Psychoneuroendo- crinology 10:469-474, 1985.

227. Hunter R, Dick H, Christie JE, Goodwin GM. Fink G: Lymphocyte glucocorticoid receptor binding in depression: Normal values follow- ing recovery. JAffective Disord 14:155-159, 1988.

228. Extein I . Tallman J , Smith CC, Goodwin FK: Changes in lymphocyte beta-adrenergic receptors in depression and mania. P sychiatr Res 1:191-197, 1979.

229. Wood K, Whiting K , Coppen A: Lymphocyte beta-adrenergic recep- tor density of patients with recurrent affective illness. JAffective Disord 10:3-8, 1986.

230. Berrettini WH, Cappelari CB, Nurnberger JI Jr, Gershon ES: Beta- adrenergic receptors on lyniphoblasts. A study of manic-depressive illness. Neuropsychohiology 17: 15-18, 1987.

231. Halper JP, Brown RP, Sweeney JA, Kocsis JH, Peters A, Mann JJ: Blunted beta-adrenergic responsivity of peripheral blood mononuclear cells in endogenous depression. Isoproterenol dose-response studies. Arclz Gen Psvchiatr 45:241-244, 1988.

232. Anisimov VN: Pineal gland, aging and carcinogenesis (abstr). Neuroendocrinol Lett 9:28 I , 1987.

233. Angeli A, Gatti G, Sartori ML, Del Ponte D, Carignola R: Effect of exogenous melatonin on human natural killer (NK) cell activity. An approach to the immunomodulatory role of the pineal gland (abstr). Neuroendocrinol Lett 9:286, 1987.

234. Karasek M, Kunert-Radek J , Stepien H, Pawlikowski M: Melatonin inhibits the proliferation of estrogen-induced rat pituitary tumor cells in vitro. Neuroendocrinol Lett 10:135-140, 1988.

235. Halberg F, Sanchez de la Pena S, Wetterberg L, Halberg J , Halberg Francine, Wrba H, Hermida Dominguez RC: Chronobiology as a tool for research, notably on melatonin and tumor development. In Bio- rhythms and Stress in the Physiopathology of Reproduction, P Pancheri, LZichella, eds. Hemisphere, New York, 1988, pp 131-175.

236. Gatti G, Sartori M, Cavallo R, Del Ponte D, Carignola R, Salvadori A, Masera R, Angeli A: Circadian variations of human natural killer (NK) cell response to positive modulation by immune interferon and negative modulation by cortisol. Chronobiologia 14: 178- 179, 1987.

237. Maestroni GJM, Conti A, Pierpaoli W: Role of the pineal gland in immunity. Circadian synthesis and release of melatonin modulates the antibody response and antagonizes the immunosuppressive effect of corticosterone. JNeuroimmunol 13:19-30, 1986.

238. Brainard GC, Knobler RL, Podolin PL, Lavasa M, Lublin FD: Neuroimmunology: Modulation of the hamster immune system by pho- toperiod. LifeSci40:1319-1326, 1987.

239. Brainard GC, Watson-Whitmeyer M, Knobler RL. Lublin FD: Neuro- endocrine regulation of immune parameters. Photoperiod control of the spleen in Syrian hamsters. Ann NYAcadSci 540:704-706, 1988.

240. Jacobsen FM, Wehr TA, Rosenthal NE: Pineal and seasonal reproduc- tion in seasonal affective disorder. In Biorhythms and Stress in the Patho- phy~iology ofReproduction. P Pancheri, L Zichella, eds. Hemisphere, New York, 1988, pp241-250.

241. Sanchez de la Pena S, Halberg F, Ungar F, Lakatua D: Ex vivo hierar-

Human Psychoneuroimmunology 37

handedness: evidence from individuals with diabetes, Crohn’s disease and ulcerative colitis. Neuropsychologia 25:367-374, 1987.

264. Renoux G, Biziere K, Renoux M, Guillaumin JM: Le cortex cerebral regle les responses immunes des souris. CR Acad Sci Paris 290: 719-722, 1980.

265. Bardos P, Degenne D, Lebranchu Y, Biziere K, Renoux G: Neocorti- cal lateralisation of NK activity in mice. Scand J Immunol 13: 609-611, 1981.

266. Renoux G, Biziere K , Renoux M, Guillaumin JM, Degenne D: A bal- anced brain asymmetry modulates T cell-mediated events. J Neuro- imrnunol5:227-237, 1983.

267. Renoux G, Biziere K, Renoux M, Guillaumin JM: The production of T-cell inducing factors in mice is controlled by the brain neocortex. Scand JImmunol 17:45-50, 1983.

268. Neveu PJ, Taghzouti K, Dantzer R, Simon H, Le Moal M: Modula- tion of mitogen-induced lymphoproliferation by cerebral neocortex. L @ Sci 38: 1907-1913, 1986.

269. Bameoud P, Neveu PJ, Vitiello S, Le Moal M: Functional heterogene- ity of the right and left cerebral neocortex in the modulation of the immune system. Physiol Behav 41525-530. 1987.

270. Gur RE, Resnick SM, Alavi A, Cur RC, Caroff S , Dann R, Silver FL, Saykin AJ, Chawluk JB, Kushner M, Reivich M: Regional brain function in schizophrenia. I. A positron emission tomography study. Arch Gen Psychiatr 44:119-125, 1987.

271. Bracha HS: Asymmetric rotational (circling) behavior, a dopamine- related asymmetry: preliminary findings in unmedicated and never- medicated schizophrenic patients. Biol Psychiarr 22:995- 1003, 1987.

272. Crow TJ, Deakin JFW: Neurohumoral transmission, behaviour and mental disorder. In Handbook of Psychiatry Vol. 5 : The Scientific Foun- dations of Psychiatry, M Shepherd, ed. Cambridge University Press, Cambridge, England, 1985, pp 137-182.

273. Birch RE, Rosenthal AK, Polmar SH: Pharmacological modification of immunoregulatory T lymphocytes. 11. Modulation of T lymphocute cell surface characteristics. Clin Exp lmmunol48:23 1-238, 1982.

274. Jankov BD: From immunoneurology to immunopsychiatry: Neuro- modulating activity of anti-brain antibodies. In t Rev Neurobiol 26:249-314, 1985.

275. Edelman G: CAMS and Igs: Cell adhesion and the evolutionary ori- gins of immunity. ImmunolRev 100:11-15, 1987.

276. Garb R, Kutz I , Bleich A, Solomon Z: Varieties of combat stress reaction. An immunological metaphor. Br J Psychiatr 151:248-25 I , 1987.

277. Ader R, Cohen N: Behaviorally conditioned immunosuppression. Psvchosom Med 37:333-340, 1975.

278. Rogers MP, Reich P, Strom TB, Carpenter CB: Behaviorally condi- tioned immunosuppression: Replication of a recent study. Psychosom Med 38:447-451, 1976.

279. Cohen N, Ader R, Green N, Bovbjerg D: Conditioned suppression of a thymus-independent antibody response. Psychosom Med 41: 487-491, 1979.

280. Bovbjerg D , Ader R, Cohen N: Behaviorally conditioned suppres- sion of a graft-versus-host response. Proc Nut1 Acad Sci USA 79583-585, 1982.

281, Gorczynski RN, Kennedy M: Associative learning and regulation of immune responses. f r o g Neuropsychopharmacol Biol Psychiatr 8593-600, 1984.

282. McCoy DF, Roszman TL, Miller JS, Kelly KS, Titus MJ: Some param- eters of conditioned immunosuppression: Species difference and CS-US delay. Physiol Behav 36:731-736, 1986.

283. Gorczynski RM, Macrae S . Kennedy M: Conditioned immune re- sponse associated with allogenic skin grafts in mice. J Immune/ 129:704-709, 1982.

284. Djuric VJ, Markovic BM, Lazarevic M, Jankovic BD: Pavlovian con- ditioning to environmental stimuli as a cause of increased resistance

chy ofcircadian-infradian rhythmic pineal-pituitary-adrenal intermodu- lations in rodents. In Biorhythms and Stress in the Physiopathology of Reproduction. P Pancheri, L Zichella, eds. Hemisphere, New York, 1988, pp 177-214.

242. Covelli V, Grassi PP, Maffione AB, Catino A, Jirillo E: Impairment of non-specific immunity in patients with anxiety disorders (phobic disorders). N Trend Exp Clin Psychiatr 4:37-45, 1988.

243. Todd RD, Hickock JM, Anderson GM, Cohen DJ: Antibrain antibod- ies in infantile autism. Biol Psychiatr 23:644-647, 1988.

244. Brown S-L, Charney DS, Woods SW, Heninger GR, Tallman J: Lym- phocyte beta-adrenergic receptor binding in panic disorder. Psycho- pharmacology 94:24-28, 1988.

245. Lymphocyte beta-adrenergic receptor modification in bulimia. Arch Gen Psychiatr 45:479-482, 1988.

246. Dafny N, Pellis NR: Evidence that opiate addiction is in part an immune response. Destruction of the immune system by irradiation-altered opi- ate withdrawal. Neuropharmacology 252 15-8 18, 1986.

247. Dougherty PM, Drath DB, Dafny N: Evidence of an immune system to brain communication axis that affects central opioid functions: Muramyl peptides attenuate opiate withdrawal. Eur J Pharmacol 141:253-260, 1987.

248. Kubitz KA, Peavey BS, Moore BS: The effect of daily hassles of humoral immunity: An interaction moderated by locus of control. Biofredback SelfRegul11:115-123, 1986.

249. Kunzendorf RG, Butler W: Personality and immunity: Depressive tend- encies versus manic and schizophrenic tendencies. Psycho1 Rep 59:622, 1986.

250. Heisel JS, Locke SE, Kraus LJ, Williams RM: Natural killer cell activ- ity and MMPI scores of a cohort of medical students. Am J Psychiatr 143: 1382- 1386, 1986.

25 1. Melnechuk T: Why has psychoneuroimmunology been controversial? Proponents and skeptics at a scientific conference. Advances 2(4):22-38, 1985.

252. Ruff MR, Pert CB, Weber RJ, Wahl LM, Wahl SM, Paul SM: Benzodiazepine receptor mediates chemotaxis of human monocytes. Science 229:1281-1283, 1985.

253. Yehuda S, Shredny B, Kalechman Y: Effects of DSIP, 5-HTP and sero- tonin on the lymphokine system: A preliminary study. Int J Neurosci 33: 185- 197, 1987.

254. Cam DJJ: Receptors for neuroendocrine peptides on cells of the immune system. f r o g Neuroendocrinimmunol 1 :(3):20-21, 1988.

255. Goldstein AL, Guha A, Zatz MM, White A: Purification and biologi- cal activity of thymosin, a hormone of the thymus gland. Proc Nut1 AcadSci USA 69:1800-1803, 1972.

256. Geenen V, Legros J - J , Franchimont P, Baudrihaye M, Defresne M-P, Boniver J : The neuroendocrine thymus: Coexistence of oxy- tocin and neurophysin in the human thymus. Science 232508-51 I , 1986.

257. Molinas FC, Wietzerbin J , Falcoff E: Human platelets possess recep- tors for a lymphokine: demonstration of high specific receptors for HuIFNgammal. JImmunol 138:802-806, 1987.

258. Dougherty P: Gilda. The wisecracking actress laughs in the face of cancer. Life 11(3):7-78, 1988.

259. Flor-Henry P: Lateralized temporal-limbic dysfunction and psychopa- thology. Ann NY Acad Sci 280:777-797, 1976.

260. Memn EL: Motor and sighting dominance in chronic schizophrenics. Relationships to social competence, age at first admission, and clini- cal course. Br J Psychiatr 145:401-406, 1984.

26 1 . Dvirskii AE: Functional asymmetry in the cerebral hemispheres in clini- cal types of schizophrenia. Neurosci Behav Physiol7:236-239, 1976.

262. Geschwind N, Behan P: Left-handedness: Association with immune disease, migraine and developmental learning disorder. Proc Nut1 Acad Sci USA 795097-5100, 1982.

263. Searlman A, Fugagli AK: Suspected autoimmune disorders and left-

38 Biondi and Kotzalidis

to repeated anaphylactic shock in the rat (abstr). Ninrh European Immu- noloxy Meefing, Rome, September 1988.

285. Freed S: Comparative biology of mnemonic processes: Conditioned defensive reflexes and conditioned immune reactions. In? J Neurosci 23:281-285, 1984.

286. Pfenninger KH: Of nerve growth cones, leukocytes and memory: Sec- ond messenger systems and growth-regulated proteins. Trend Neurosci

287. Post RM, Weiss SRB, Rubinow DR: Recurrent affective disorders: Les- sons from limbic kindling. In Neuroendocrinology ofMood, D Ganten, D Pfaff, eds. Springer-Verlag, Berlin, 1988, pp 91-1 15.

288. Goddard GV, Morel1 F Chronic progressive epileptogenesis induced by focal electrical stimulation of brain (abstr). Neurology 21: 393, 1971.

289. Wada JA, Sata M: Generalized convulsive seizures induced by daily

91562-565, 1986.

electrical stimulation of the amygdala in cats. Correlative electrographic and behavioral features. Neurology 24565-574, 1974.

290. Ballenger JC, Post RM: Kindling as a model for alcohol withdrawal symptoms. BrJPsychiafr 133:l-14, 1978.

291. Post RM, Uhde TW, Putnam FW, Ballenger JC, Berrettini WH: Kin- dling and carbamazepine in affective illness. J Nerv Men? Dis 170:717-731, 1982.

292. Post RM, Kopanda RT: Cocaine, kindling, and psychosis. Am JPsychiurr 133:627-634, 1976.

293. Kotzalidis GD, Riccio L, Pancheri L: Trattamento della schizofrenia con peptidi. 11. Ormoni gastroenteropancreatici. A. L'insulina ed il coma ipoglicemico. Schizofrenia (in press).

294. Sutula T, Xiao-Xian H, Cavazos J , Scott G: Synaptic reorganization in the hippocampus induced by abnormal functional activity. Science 239:1147-1150, 1988.