SPE | SRM | UCN | UREI | Main Page

 

THE HYPOTHALAMIC-PITUITARY-ADRENAL AXIS: ITS ENDOCRINE DISORDERS AND ITS INTERACTIONS WITH THE IMMUNE SYSTEM

 

 George P. Chrousos, MD, Head, Section on Pediatric Endocrinology

Alejandro Ayala, MD, Staff Physician

Tomoshige Kino, MD, PhD, Staff Scientist

Pamela Stratton, MD, Commissioned Officer

Evangelia Charmandari, MD, Postdoctoral Fellow

Massimo De Martino, MD, Postdoctoral Fellow

Takamasha Ichijo, MD, PhD, Postdoctoral Fellow

Sujata Kelkar, PhD, Postdoctoral Fellow

Emmanouil Souvatzoglou, MD, Postdoctoral Fellow

Emmanouil Zoumakis, PhD, Postdoctoral Fellow

Ninet Sinaii, MS, Predoctoral Student

Keith Zachman, MS, Biolaboratory Technician

Salvatore Alesci, MD, Guest Researchera

Ze’ev Hochberg, MD, Guest Researcherb

Deborah Merke, MD, Guest Researcherc

Karen Winer, MD, Guest Researcherd

 

We investigate the molecular basis of the following genetic disorders: pituitary and adrenal tumors, adrenal insufficiency, ACTH resistance, congenital adrenal hyperplasia, glucocorticoid resistance/hypersensitivity, and mineralocorticoid resistance (hypothalamic-pituitary-adrenal [HPA] axis); hypothalamic hypogonadism (hypothalamic-pituitary-gonadal [HPG] axis); and metabolic and immune disorders related to the functions of the HPA and HPG axes and the autonomic system, such as premature adrenarche, eating disorders that include anorexia and bulimia nervosa and adolescent obesity, childhood asthma and rheumatoid arthritis, premenstrual tension syndrome, postpartum and climacteric depression, and autoimmunity. We also study the critical influences of early life stress on the later development of behavioral disorders (dysthymia, depression, dissociative and conduct disorders, substance abuse) and metabolic disorders (metabolic syndrome X). By identifying pathogenetic defects of the adrenocorticotropic hormone and glucocorticoid receptor genes and of genes of functionally related molecules, we have elucidated the molecular mechanisms of such hereditary diseases as congenital isolated glucocorticoid deficiency and glucocorticoid resistance syndromes and of acquired states such as glucocorticoid resistance associated with the respiratory distress syndrome or glucocorticoid hypersensitivity associated with the AIDS-related lipodystrophy and insulin resistance syndrome. We also conduct research to increase our understanding of the physiologic and pathophysiologic interactions between the endocrine and immune systems.

A natural determinant of glucocorticoid sensitivity in human tissues and its role in human pathophysiology

Kino, De Martino, Charmandari, Souvatzoglou, Alesci, Chrousos; in collaboration with Vottero

The glucocorticoid receptor gene, located in the long arm of chromosome 5, encodes two proteins produced by alternative splicing: the classic glucocorticoid receptor, designated GRalpha, and a nonligand-binding variant called GRbeta. We demonstrated that both isoforms are expressed in human tissues and that the nonligand-binding receptor is present at high concentrations in normal human tissues and exerts dominant negative effects on the classic receptor. Given that alteration in glucocorticoid receptor gene splicing produces different ratios of GRalpha to GRbeta, we hypothesized that the ratio of glucocorticoid receptor types regulates tissue sensitivity to glucocorticoids. A change in sensitivity might have major implications for human pathophysiology in that both glucocorticoid resistance and glucocorticoid hypersensitivity can produce disease states. The former causes hyperandrogenism and hypertension while the latter could be involved in metabolic disorders such as visceral obesity or syndrome X, affective disorders such as depression, and immune disorders characterized by suppression of the inflammatory/immune response and vulnerability to infectious diseases and/or tumors. We showed that glucocorticoid-resistant asthma type II is associated with a major shift in the GRalpha-to-GRbeta ratio toward GRbeta predominance, explaining the profound resistance of patients’ cells to glucocorticoids. We observed a similar shift in cells from glucocorticoid-resistant New World monkeys. We demonstrated that normal human neutrophils, which are naturally glucocorticoid-resistant, also overexpress GRbeta. Recently, we showed how the GRbeta isoform causes glucocorticoid insensitivity. This isoform forms heterodimers with GRalpha, which then inhibit interaction with the nuclear receptor p160 transcription co-activators and hence the transcription of glucocorticoid receptor–responsive genes.

De Martino M, Bhattacharyya N, Alesci S, Fabbri A, Chrousos GP, Kino T. The glucocorticoid receptor (GR) and the orphan nuclear receptor chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) interact with and mutually affect each other’s transcriptional activities: implications for intermediary metabolism. Mol Endocrinol 2004;18:820-833.

Galon J, Franchimont D, Hiroi N, Boettner A, Ehrhart-Bornstein M, Chrousos GP, Bornstein S. Gene profiling reveals unknown enhancing and suppressive actions of glucocorticoids on immune cells. FASEB J 2002;16:61-71.

Hauk PJ, Goleva E, Strickland I, Vottero A, Chrousos GP, Kisich KO, Leung DY. Increased glucocorticoid receptor beta expression converts mouse hybridoma cells to a corticosteroid-insensitive phenotype. Am J Respir Cell Mol Biol 2002;27:361-367.

Kino T, Chrousos GP. Tissue-specific glucocorticoid resistance-hypersensitivity syndromes: multifactorial states of clinical importance. J Allergy Clin Immunol 2002;109:609-613.

Viral determinants of glucocorticoid hypersensitivity in patients with AIDS coupled with AIDS-related lipodystrophy and insulin resistance syndrome

Kino, De Martino, Charmandari, Souvatzoglou, Alesci, Chrousos; in collaboration with Vottero

Recently, we described a human example of a glucocorticoid hypersensitivity state: Vpr, one of the accessory proteins of HIV-1, acts as a co-regulator of the glucocorticoid receptor, altering the sensitivity of human cells to glucocorticoids so that viral replication is favored. Such hypersensitivity explains several manifestations of AIDS, including innate and T helper 1 immunosuppression, T helper 2 shift, myopathy and muscle atrophy, and visceral obesity–related insulin resistance with its metabolic manifestations and cardiovascular consequences. Vpr co-activates the glucocorticoid receptor by binding to it and to the co-integrator molecule p300/CBP and by stabilizing the transcription initiation complex. Recently, we identified Tat, a second HIV-1 accessory protein, that also acts as a co-activator of the glucocorticoid receptor. This protein binds to nuclear receptor p160 co-activators and cooperates with Vpr in potentiating the activity of glucocorticoids on transcription of responsive genes. It appears that the HIV-1 LTR promoter also uses p300/CBP and p160 co-activators for its own activation. The ability of HIV-1 proteins such as Vpr to influence the activity of nuclear receptor co-activators extends beyond the glucocorticoid signaling system. We recently identified the PPAR-gamma and the insulin signaling systems as targets of Vpr in the host cell. Indeed, Vpr causes resistance to PPAR-gamma and insulin by directly interfering with the actions of the former on transcription and by indirectly blocking insulin actions on the activity of the fork-head transcription factor, a major modulator of insulin actions.

Kino T, Gragerov A, Slobodskaya O, Tsopanomihalou-Nglotsou M, Chrousos GP, Pavlakis G. Human immunodeficiency virus type 1 (HIV-1) accessory protein Vpr induces transcription of the HIV-1 and glucocorticoid-responsive promoters by binding directly to p300/CBP coactivators. J Virol 2002;76:9724-9734.

Kino T, Slobodskaya O, Pavlakis N, Chrousos GP. Nuclear receptor coactivator p160 proteins enhance the human immunodeficiency virus (HIV) type 1 long terminal repeat promoter by bridging promoter bound factors and the Tat/pTEB complex. J Biol Chem 2002;277:2396-2405.

Kino T, Souvatzoglou E, De Martino MU, Tsopanomihalou M, Wan Y, Chrousos GP. Protein 14-3-3 sigma interacts with and favors cytoplasmic subcellular localization of the glucocorticoid receptor, acting as a negative regulator of the glucocorticoid signaling pathway. J Biol Chem 2003;252:32-44.

Kino T, Tsukamoto M, Chrousos GP. Transcription factor TFIIH components enhance the GR coactivator activity but not the cell cycle-arresting activity of the human immunodeficiency virus type-1 protein Vpr. Biochem Biophys Res Commun 2002;298:17-23.

Mirani M, Elenkov I, Volpi S, Hiroi N, Chrousos GP, Kino T. HIV-1 protein Vpr suppresses IL-12 production from human monocytes by enhancing glucocorticoid action: potential implications of Vpr coactivator activity for the innate and cellular immunity deficits observed in HIV-1 infection. J Immunol 2002;169:6361-6368.

Familial and sporadic glucocorticoid resistance syndromes and inflammation-induced glucocorticoid resistance

Kino, De Martino, Charmandari, Souvatzoglou, Alesci, Chrousos; in collaboration with Latronico, Meduri, Vottero

Our group recently analyzed patients with the familial/sporadic glucocorticoid resistance syndrome, which is caused by mutations of the glucocorticoid receptor gene. Two novel glucocorticoid receptor mutations, one replacing isoleucine (I) 559 with aspartic acid (N) and another in which isoleucine (I) 747 was replaced by methionine (M), both cause glucocorticoid resistance in the heterozygote state, acting in a transdominant negative fashion by forming function-deficient dimers with wild-type GRalpha. We examined the cause of the transdominant activity of these mutant receptors and found that GRI559N has a nuclear localization sequence defect, which interferes with the translocation of wild-type GRalpha from the cytoplasm into the nucleus, while GRI747M has a defective Activation Function–2 (AF-2) domain, which prevents proper interaction of the wild-type receptor with the p160 nuclear receptor co-activators. Recently, we studied a newborn female infant with severe sexual ambiguity, an unusual presentation of the glucocorticoid resistance syndrome. In this patient, the GRalpha gene bore a homozygous mutation that decreased the receptor’s affinity for glucocorticoids and its ability to activate glucocorticoid-responsive genes. We are studying an American kindred in which the proposita has a heterozygote amino acid substitution in its C-terminus that disturbs AF-2–mediated interaction with p160 nuclear receptor co-activators.

We have further studied the mechanisms by which severe inflammation causes resistance of immune and immune-associated cells to glucocorticoids, a particularly important issue in pediatrics and medicine. We have determined that the key mechanisms through which inflammation causes resistance is an inflammatory cytokine-induced signaling cascade in which the TNFalpha and Fas systems (1) activate NF-kappaB by phosphorylation and nuclear translocation and (2) block the p160 glucocorticoid receptor p160 co-activators from interacting properly with the glucocorticoid receptor through activation of an intermediate cytoplasmic protein called FLASH.

Charmandari E, Kino T, Souvatzoglou E, Vottero A, Bhattacharayya N, Chrousos GP. Natural glucocorticoid receptor mutants causing glucocorticoid resistance: molecular genotype, genetic transmission and clinical phenotype. J Clin Endocrinol Metab 2004;89:1939-1949.

Franchimont D, Kino T, Galon J, Meduri GU, Chrousos GP. Glucocorticoids and inflammation revisited: the state of the art. NIH Clinical Staff Conference. Neuroimmunomodulation 2003;10:247-260.

Kino T, Chrousos GP. Tumor necrosis factor alpha receptor- and Fas-associated FLASH inhibit transcriptional activity of the glucocorticoid receptor by binding to and interfering with its interaction with p160 type nuclear receptor coactivators. J Biol Chem 2003;278:3023-3029.

Mendonca BB, Leite MV, deCastro M, Kino T, Elias LL, Bachega TAS, Billerbeck AE, Arnhold IAJP, Chrousos GP, Latronico AC. Female pseudohermaphroditism caused by a novel homozygous missense mutation of the glucocorticoid receptor gene. J Clin Endocrinol Metab 2002;87:1805-1809.

Vottero A, Kino T, Combe H, Lecomte P, Chrousos GP. A novel, C-terminal dominant negative mutation of the glucocorticoid receptor causes familial glucocorticoid resistance through abnormal interactions with p160 steroid receptor coactivators. J Clin Endocrinol Metab 2002;87:2658-2667.

Pathophysiology and therapy of adrenal hyperfunction–congenital adrenal hyperplasia

Merke, Charmandari, Chrousos

The goal of our research is to understand the mechanisms involved in diseases caused by adrenal hyperandrogenism. We use insights into the mechanism to generate hypotheses for new approaches to treatment. Deborah Merke is principal investigator for a clinical trial that is testing a new medical treatment for children with classic congenital adrenal hyperplasia (CAH). Preliminary results based on two years of combined therapy of an anti-androgen and an aromatase inhibitor showed that children receiving the new treatment regimen exhibited normalization of growth velocity and bone maturation. Thus, the regimen represents a promising treatment approach for children with classic 21-hydroxylase deficiency. Reproductive, metabolic, and other morbidities, including risk of tumor formation, particularly testicular tumors, are currently under investigation in the severe forms of the disease. Bilateral adrenalectomy performed on a severe case revealed massive lymphocytic infiltration and histologic signs of adrenal carcinoma, leading to new insights regarding lymphocyte-adrenal-cell interaction and adrenocortical tumor formation. Recent clinical studies led to findings of adrenomedullary dysplasia and hypofunction in patients with CAH, results that have important clinical implications. The protective function of the adrenal medulla may be absent in the most severe cases. Studies of hypoglycemia and exercise tolerance in patients are under way. It appears that the lack of a proper epinephrine response to exercise prevents an appropriate glucose response in CAH. Recently, children with CAH were found to be resistant to insulin, a finding with negative repercussions for these patients (Charmandari et al., J Clin Endocrinol Metab 2002;87:2139). Studies of healthy children are also under way, and preliminary results reveal sex and age differences in catecholamine secretion. Clinical initiatives include studies of the pathophysiology of premature adrenarche and the effects of androgens and other hormones on the brain. Psychological testing and head MRI studies are being conducted with children suffering from endocrine disorders, including CAH, Cushing’s syndrome, and familial male precocious puberty. The studies will provide insight into hormonal influences on the brain during growth and development.

Charmandari E, Eisenhofer G, Mehlinger S, Carlson A, Wesley R, Keil M, Chrousos GP, New MI, Merke DP. Adrenomedullary function may predict phenotype and genotype in classic 21-hydroxylase deficiency. J Clin Endocrinol Metab 2002;87:3031-3037.

Charmandari E, Merke D, Negro PJ, Keil M, Martinez PE, Haim A, Gold PW, Chrousos GP. Endocrinologic and psychologic evaluation of 21-hydroxylase deficiency carriers and matched normal subjects: evidence for physical and/or psychologic vulnerability to stress. J Clin Endocrinol Metab 2004;89:2228-2236.

Merke DP, Fields JD, Keil MF, Vaituzis AC, Chrousos GP, Giedd JN. Children with classic congenital adrenal hyperplasia have decreased amygdala volume: potential prenatal and postnatal hormonal effects. J Clin Endocrinol Metab 2003;88:1760-1765.

Weise M, Drinkard B, Mehlinger SL, Eisenhofer G, Chrousos GP, Merke DP. Stress dose of hydrocortisone is not beneficial in patients with classic congenital adrenal hyperplasia undergoing short-term high-intensity exercise. J Clin Endocrinol Metab 2004;89:3679-3684.

Weise M, Mehlinger S, Rawson E, Charmandari E, Eisenhofer G, Yanovski J, Chrousos GP, Merke D. Patients with classic congenital adrenal hyperplasia have decreased epinephrine reserve and defective glucose elevation in response to high intensity exercise. J Clin Endocrinol Metab 2004;89:591-597.

Role of corticotropin releasing hormone in behavior and behavioral/developmental/sleep disorders

Charmandari, Chrousos; in collaboration with Cizza, Deuster, Dorn, Gold, Susman

We have used corticotropin releasing hormone (CRH) to probe central mechanisms in patients with atypical seasonal depression, the chronic fatigue/fibromyalgia syndromes, and the postpartum blues/depression syndromes. The patients’ responses to CRH are indicative of mild, sustained hypocortisolism of central nervous system etiology, suggesting that a spectrum of diseases exists that are characterized by hypofunction of the central stress system. These syndromes contrast with melancholic depression, panic anxiety, and anorexia nervosa, for which we earlier demonstrated hyperactivity of the CRH system. As we have shown, the behavioral similarities between patients with Cushing’s syndrome and atypical depression probably stem from a similar pathologic decrease in CRH secretion. Recently, we demonstrated that preadolescent and adolescent girls with a history of sexual abuse exhibited an HPA axis response to CRH similar to that of patients with melancholic depression or recovering anorexics. The findings suggest that major stress in children may have long-term effects on the HPA axis accompanied by effects on the psyche. In a social nonhuman primate model, we demonstrated effects of parental abuse not only on the HPA axis but also on growth.

Individuals who are carriers of a 21-hydroxylase deficiency, such as parents of patients with CAH, are presumably totally asymptomatic. We studied the mood and HPA axis of such subjects, testing the hypothesis that a compensatory elevation of CRH secretion would be associated with indices of increased anxiety. We found that, indeed, compensatory changes in the activity of the HPA axis are apparent in a standard CRH test and that these changes correlate positively with anxiety. As a group, the carriers had higher anxiety scores than appropriate controls. Studies in healthy normal volunteers demonstrated that, in middle age, the sleep centers in the brain are sensitized to the activational effects of CRH and glucocorticoids, a finding that explains the increasing vulnerability to stress- or caffeine-induced insomnia with advancing age. Studies in young and middle-aged patients with idiopathic insomnia showed that such patients have significant evening activation of their HPA axis and an overall activation of the axis that correlates positively with indices of poor-quality sleep, such as decrements in amount of deep sleep and increments in wakefulness (Vgontzas et al., Endocrinol Metab Clin North Am 2002;31:15). We also recently showed that antalarmin, a nonpeptide CRH antagonist that crosses the blood barrier, inhibits the stress-mimicking properties of CRH, including its effects on the HPA axis and behavior. This drug thus represents a new class of pharmacologic agents for the potential treatment of diseases characterized by CRH hypersecretion, such as melancholic depression, chronic anxiety disorder, anorexia nervosa, and idiopathic insomnia.

Ayala AR, Pushkas J, Higley JD, Ronsaville D, Gold PW, Chrousos GP, Pacak K, Calis KA, Gerald M, Lindell S, Rice KC, Cizza C. Behavioral, adrenal and sympathetic responses to long term administration of an oral corticotropin-releasing hormone receptor antagonist in a primate stress paradigm. J Clin Endocrinol Metab 2004;89:5729-5737.

Gold PW, Chrousos GP. Organization of the stress system and its dysregulation in melancholic and atypical depression: high vs. low CRH/NE states. Mol Psychiatry 2002;7:254-275.

Vgontzas A, Zoumakis E, Bixler EO, Lin H-M, Follett H, Kales A, Chrousos GP. Adverse effects of modest sleep restriction on sleepiness, performance, and inflammatory cytokines. J Clin Endocrinol Metab 2004;89:2119-2126.

Ward AM, Syddall HE, Wood PJ, Chrousos GP, Phillips DI. Fetal programming of the hypothalamic-pituitary-adrenal (HPA) axis: low birth weight and central HPA regulation. J Clin Endocrinol Metab 2004;89:1227-1233.

Yuhas Y, Weizman A, Chrousos GP, Ovadia H, Ashkenazi S. Involvement of the neuropeptide corticotropin-releasing hormone in an animal model of shigella-related seizures. J Neuroimmunol 2004;153:36-39.

Cytokines and the inflammatory response in endocrine hyper- and hyposecretion states

Chrousos; in collaboration with Elenkov, Gold, Sternberg, Vgontzas

Several cytokines, including interleukin 1, tumor necrosis factor-alpha (TNF-alpha), and interleukin-6 (IL-6), as well as several lipid mediators of inflammation, including platelet-activating factor and several eicosanoids, cause profound stimulation of the HPA axis, primarily by activating the CRH neuron and, through it, by suppressing and restraining the inflammatory/immune response. We recently studied IL-6 in humans and found that this cytokine is extremely potent in activating the HPA axis and that, at high doses, causes systemic vasopressin secretion. At all doses, it is not noxious, unlike TNF-alpha or interleukin-1, which cause hypotension and vascular leakage. We demonstrated that IL-6 is a potent stimulator of ACTH release in normal subjects and that catecholamines and glucocorticoids regulate IL-6 secretion. IL-6 levels are normal in patients with Cushing’s syndrome but increase markedly after the surgical cure of hypercortisolism. The rebound elevation in circulating IL-6 levels immediately after surgical cure of hypercortisolism may be the cause of the systemic symptoms typical of the glucocorticoid withdrawal syndrome in these patients. Thus, IL-6 participates in an immune-endocrine feedback loop involving CRH, ACTH, and cortisol. We found that the stress hormones glucocorticoids and catecholamines inhibit the secretion of interleukin-12 and stimulate the secretion of IL-10 by monocytes/macrophages, leading to a shift from T helper 1– to T helper 2–directed immunity. We discovered that, in the transition from pregnancy to the postpartum period, the hormonal changes that take place cause a T helper 2– to T helper 1–shift, which explains the vulnerability of postpartum women to autoimmune disorders of the T helper 1 type, such as Hashimoto thyroiditis, multiple sclerosis, and rheumatoid arthritis.

Bader T, Zoumakis E, Friedberg M, Chrousos GP, Hochberg Z. Human adipose tissue under in vitro inhibition of 11beta-hydroxysteroid dehydrogenase type 1: differentiation and metabolism changes. Horm Metab Res 2002;34:752-757.

Vgontzas AN, Zoumakis M, Papanicolaou DA, Bixler E, Prolo P, Lin HM, Vela-Bueno A, Kales MD, Chrousos GP. Chronic insomnia is associated with a shift of IL-6 and TNFalpha secretion from nighttime to daytime. Metabolism 2002;29:1252-1261.

Leptin and cytokines in obesity, polycystic ovary syndrome, and sleep disorders

Ayala, Zoumakis, Bornstein, Chrousos; in collaboration with Vgontzas

We recently demonstrated that, in addition to leptin, human fat secretes large amounts of TNF-alpha and Il-6 and that the secretion of adipose tissue cytokines is regulated by both food intake and the sympathetic nervous system. The secretion of these cytokines, e.g., Il-6, has a circadian rhythm that is influenced by sleep while circulating levels increase in proportion to BMI and are further elevated by visceral adiposity. Sleep deprivation is associated with elevations of daytime IL-6, whose levels correlate with somnolence and fatigue. This holds true for patients with insomnia who, during the day, have elevated levels not only of cortisol but also of IL-6. Interestingly, IL-6 concentrations in plasma are proportional to body fat, and viscerally obese subjects have further elevations that correlate with sleep apnea measures. Women with polycystic ovary syndrome exhibit high cytokine and leptin levels and are at markedly increased risk for sleep apnea while girls with premature adrenarche show features of visceral adiposity such as high fasting leptin and insulin, predictors of later development of polycystic ovary syndrome and sleep apnea. The elevation of inflammatory cytokines in obesity and its correlation with many pathologic manifestations of this condition have led us to conduct clinical studies with biological antagonists of such cytokines.

Kalantaridou SN, Naka KK, Papanikolaou E, Kazakos N, Kravariti M, Calis KA, Paraskevaidis EA, Sideris DA, Tsatsoulis A, Chrousos GP, Michalis LK. Impaired endothelial function in young women with premature ovarian failure: normalization with hormone therapy. J Clin Endocrinol Metab 2004;89:3907-3913.

Vgontzas AN, Bixler EO, Chrousos GP. Metabolic disturbances in obesity versus sleep apnoea: the importance of visceral obesity and insulin resistance. J Intern Med 2003;254:32-44.

Vgontzas AN, Zoumakis E, Lin H-M, Bixler EO, Trakada G, Chrousos GP. Marked decrease of sleepiness in patients with sleep apnea by etanercept, a TNFalpha antagonist. J Clin Endocrinol Metab 2004;89:4409-4413.

Vgontzas AN, Zoumakis M, Bixler EO, Lin HM, Prolo P, Vela-Bueno A, Kales A, Chrousos GP. Impaired nighttime sleep in healthy old versus young adults is associated with elevated plasma interleukin-6 and cortisol levels: physiologic and therapeutic implications. J Clin Endocrinol Metab 2003;88:2087-2095.

Immune and reproductive CRH and the role of CRH antagonists

Zoumakis, Chrousos; in collaboration with Dorn, Gold, Gravanis, Sano

We demonstrated that CRH is produced locally at sites of inflammation and has profound pro-inflammatory effects at an autocrine/paracrine level. CRH is a potent degranulator of mast cells, a phenomenon that can be inhibited by antalarmin, the nonpeptide CRH antagonist specific for type 1 receptors. The antagonist has marked systemic anti-inflammatory actions and has ameliorated the disease in adjuvant-induced arthritis, a model of rheumatoid arthritis, suggesting that CRH antagonists may be useful in the treatment of inflammatory disorders in man. CRH was also found in the ovary and endometrium, where it may participate in the inflammatory phenomena of ovulation, luteolysis, blastocyst implantation, and menstruation. Antalarmin blocked implantation in rats and labor in sheep, suggesting that CRH antagonists may have clinical applications in reproductive medicine.

Webster EL, Barrientos RM, Contoreggi C, Isaac MG, Ligier S, Habib K, Chrousos GP, McCarthy EF, Rice KC, Gold PW, Sternberg EM. Corticotropin-releasing hormone (CRH) antagonist attenuates adjuvant-induced arthritis: evidence supporting major role for CRH in peripheral inflammation. J Rheumatol 2002;29:1252-1261.

aVisiting Associate, NIMH, Bethesda, MD

bTechnion-Israel Institute of Technology, Haifa, Israel

cStaff Physician, Warren Grant Magnusson Clinical Center, Bethesda, MD

dEndocrinology, Nutrition and Growth Branch, NICHD, Bethesda, MD

COLLABORATORS

Stefan Bornstein, MD, University of Düsseldorf, Germany

Giovanni Cizza, MD, PhD, Clinical Neuroendocrinology Branch, NIMH, Bethesda, MD

Patricia A. Deuster, PhD, Uniformed Services University of the Health Sciences, Bethesda, MD

Lorah Dorn, RN, University of Pittsburgh, Pittsburgh, PA

Ilia Elenkov, MD, PhD, Georgetown University, Washington, DC

Lucia Ghizzoni, MD, University of Parma, Italy

Philip W. Gold, MD, Clinical Neuroendocrinology Branch, NIMH, Bethesda, MD

Achilles Gravanis, PhD, University of Crete, Heraklion, Greece

Ana-Claudia Latronico, MD, University of São Paulo, Brazil

Donald Y. Leung, MD, University of Colorado Health Sciences Center, Denver, CO

Maria A. Magiakou, MD, Athens University Medical School, Greece

Andrew Margioris, MD, PhD, University of Crete, Heraklion, Greece

George Mastorakos, MD, Athens University, Greece

G. Umberto Meduri, MD, University of Tennessee Medical Center, Memphis, TN

Edward H. Oldfield, MD, Surgical Neurology Branch, NINDS, Bethesda, MD

Martin Reincke, MD, Universität München, Germany

Hajime Sano, MD, Kyoto University, Japan

Roger Smith, MD, University of Newcastle Medical School, Australia

Esther M. Sternberg, MD, PsychoNeuroImmunology Program, NIMH, Bethesda, MD

Elisabeth Susman, PhD, Pennsylvania State University, University Park, PA

Constantine Tsigos, MD, Athens University, Greece

Alexander Vgontzas, MD, Pennsylvania State University, Hershey, PA

Alessandra Vottero, MD, University of Parma, Italy

Jack A. Yanovski, MD, PhD, Developmental Endocrinology Branch, NICHD, Bethesda

 

For further information, contact chrousog@mail.nih.gov