Chronic α -adrenoreceptor blockade produces age-dependent ... · Summary. In order to examine the...

9
Summary. In order to examine the influence of chronic α 1 -adrenergic receptor (α 1 -AR) blockade on the thymus structure and T-cell maturation, peripubertal and adult male rats were treated with urapidil (0.20 mg/kg BW/d; s.c.) over 15 consecutive days. Thymic structure and phenotypic characteristics of the thymocytes were assessed by stereological and flow cytometry analysis, respectively. In immature rats, treatment with urapidil reduced the body weight gain and, affecting the volume of cortical compartment and its cellularity decreased the organ size and the total number of thymocytes compared to age-matched saline-injected controls. The percentage of CD4+8- single positive (SP) thymocytes was decreased, while that of CD4-8+ was increased suggesting, most likely, a disregulation in final steps of the positively selected cells maturation. However, α 1 - AR blockade in adult rats increased the thymus weight as a consequence of increase in the cortical size and cellularity. The increased percentage of most immature CD4-8- double negative (DN) cells associated with decreased percentage of immature CD4+8+ double positive (DP) thymocytes suggests a decelerated transition from DN to DP stage of T-cell development. As in immature rats, the treatment in adult rats evoked changes in the relative numbers of SP cells, but contrary to immature animals, favoring the maturation of CD4+8- over CD4-8+ thymocytes. These results demonstrate that: i) chronic blockade of α 1 -ARs affects both the thymus structure and thymocyte differentiation, ii) these effects are age-dependent, pointing out to pharmacological manipulation of α 1 -AR-mediated signaling as potential means for modulation of the intrathymic T-cell maturation. Key words: α 1 -adrenoceptor blockade, Urapidil, Rat, Thymus structure, Thymocyte development Introduction Throughout intrathymic differentiation T cells actively migrate between stratified stromal regions, so that each thymopoietic stage is localized in anatomically and functionally distinct subregions (Shortman et al., 1990; Lind et al., 2001). The bone-marrow-derived precursors enter the thymus near the cortico-medullary junction and then they migrate toward the subcapsular epithelium where TCRß rearrangement and assembly of ß/pTα complex, leading to expression of coreceptor CD4 and CD8 molecules, and rearrangement of TCR α locus occur. As thymocytes enter this stage, they travel back through the cortex. On this route CD4+8+ double positive (DP) thymocytes are subjected to selection processes. Positively selected cells differentiate into mature CD4+ or CD8+ single positive (SP) cells, which migrate through the medulla before exiting the thymus (Lind et al., 2001). Thymocytes that are not positively selected die either of “neglect” or as result of negative selection (Chan et al., 1993). The catecholamines norepinephrine and epinephrine are believed to be important modulators of immune functions, especially following exposure to stress when they act in concert with activation of the hypothalamic pituitary axis (Cunnick et al., 1990). In vitro , catecholamines have been shown to modulate a range of immune cell functions including cell proliferation, cytokine and antibody production, lytic action and migration (Madden, 2003). It has been hypothesized that in vivo catecholamines influence immune functions by altering the responsiveness of individual immune cells (Madden, 2003) and/or by changing the number and/or subset proportion of cells that participate in a given immune response (Benschop et al., 1996). Furthermore, data from in vitro and in vivo studies have demonstrated that pharmacological manipulation of ß-adrenoceptor (AR)-mediated catecholamine action may affect the thymus structure and T-cell maturation and that these effects are age-dependent (Singh and Owen, 1976; Singh Chronic α 1 -adrenoreceptor blockade produces age-dependent changes in rat thymus structure and thymocyte differentiation B. Ple´ caˇ s-Soloravi´ c 1 , I. Hristi´ c- ˇ Zivkovi´ c 1 , K. Radojevi´ c 2 , D. Kosec 2 and G. Leposavi´ c 1 1 Department of Physiology, Faculty of Pharmacy, University of Belgrade and 2 Institute of Immunology and Virology “Torlak”, Belgrade, Serbia and Montenegro Histol Histopathol (2005) 20: 833-841 Offprint requests to: Prof. Bosiljka Ple´caˇs-Solarovi´c, Department of Physiology, Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia and Montenegro. e-mail: [email protected] http://www.hh.um.es Histology and Histopathology Cellular and Molecular Biology

Transcript of Chronic α -adrenoreceptor blockade produces age-dependent ... · Summary. In order to examine the...

  • Summary. In order to examine the influence of chronicα1-adrenergic receptor (α1-AR) blockade on the thymusstructure and T-cell maturation, peripubertal and adultmale rats were treated with urapidil (0.20 mg/kg BW/d;s.c.) over 15 consecutive days. Thymic structure andphenotypic characteristics of the thymocytes wereassessed by stereological and flow cytometry analysis,respectively. In immature rats, treatment with urapidilreduced the body weight gain and, affecting the volumeof cortical compartment and its cellularity decreased theorgan size and the total number of thymocytes comparedto age-matched saline-injected controls. The percentageof CD4+8- single positive (SP) thymocytes wasdecreased, while that of CD4-8+ was increasedsuggesting, most likely, a disregulation in final steps ofthe positively selected cells maturation. However, α1-AR blockade in adult rats increased the thymus weightas a consequence of increase in the cortical size andcellularity. The increased percentage of most immatureCD4-8- double negative (DN) cells associated withdecreased percentage of immature CD4+8+ doublepositive (DP) thymocytes suggests a deceleratedtransition from DN to DP stage of T-cell development.As in immature rats, the treatment in adult rats evokedchanges in the relative numbers of SP cells, but contraryto immature animals, favoring the maturation of CD4+8-over CD4-8+ thymocytes. These results demonstratethat: i) chronic blockade of α1-ARs affects both thethymus structure and thymocyte differentiation, ii) theseeffects are age-dependent, pointing out topharmacological manipulation of α1-AR-mediatedsignaling as potential means for modulation of theintrathymic T-cell maturation.

    Key words: α1-adrenoceptor blockade, Urapidil, Rat,Thymus structure, Thymocyte development

    Introduction

    Throughout intrathymic differentiation T cellsactively migrate between stratified stromal regions, sothat each thymopoietic stage is localized in anatomicallyand functionally distinct subregions (Shortman et al.,1990; Lind et al., 2001). The bone-marrow-derivedprecursors enter the thymus near the cortico-medullaryjunction and then they migrate toward the subcapsularepithelium where TCRß rearrangement and assembly ofß/pTα complex, leading to expression of coreceptorCD4 and CD8 molecules, and rearrangement of TCR αlocus occur. As thymocytes enter this stage, they travelback through the cortex. On this route CD4+8+ doublepositive (DP) thymocytes are subjected to selectionprocesses. Positively selected cells differentiate intomature CD4+ or CD8+ single positive (SP) cells, whichmigrate through the medulla before exiting the thymus(Lind et al., 2001). Thymocytes that are not positivelyselected die either of “neglect” or as result of negativeselection (Chan et al., 1993).

    The catecholamines norepinephrine and epinephrineare believed to be important modulators of immunefunctions, especially following exposure to stress whenthey act in concert with activation of the hypothalamicpituitary axis (Cunnick et al., 1990). In vitro,catecholamines have been shown to modulate a range ofimmune cell functions including cell proliferation,cytokine and antibody production, lytic action andmigration (Madden, 2003). It has been hypothesized thatin vivo catecholamines influence immune functions byaltering the responsiveness of individual immune cells(Madden, 2003) and/or by changing the number and/orsubset proportion of cells that participate in a givenimmune response (Benschop et al., 1996). Furthermore,data from in vitro and in vivo studies have demonstratedthat pharmacological manipulation of ß-adrenoceptor(AR)-mediated catecholamine action may affect thethymus structure and T-cell maturation and that theseeffects are age-dependent (Singh and Owen, 1976; Singh

    Chronic αα1-adrenoreceptor blockade produces age-dependent changes in rat thymus structure and thymocyte differentiationB. Plećaš-Soloravić1, I. Hristić-Živković1, K. Radojević2, D. Kosec2 and G. Leposavić11Department of Physiology, Faculty of Pharmacy, University of Belgrade and 2Institute of Immunology and Virology “Torlak”, Belgrade, Serbia and Montenegro

    Histol Histopathol (2005) 20: 833-841

    Offprint requests to: Prof. Bosiljka Plećaš-Solarović, Department ofPhysiology, Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade,Serbia and Montenegro. e-mail: [email protected]

    http://www.hh.um.es

    Histology andHistopathology

    Cellular and Molecular Biology

  • et al., 1979; Leposavić et al., 2000, Madden and Felten,2001; Rauški et al., 2003; Plećaš-Solarović et al., 2004).

    Although it is reckoned that catecholamines exertthe effects on the immune system mainly via ß2-ARs(Elenkov et al., 2000; Sanders and Straub, 2002;Madden, 2003), it should be pointed out that α2-ARexpression has been demonstrated in rat lymph node andspleen cells (Fernández-López and Pazos, 1994), as wellas in macrophages (Spengler at al., 1990). In addition,functional studies clearly implicated α2-ARs not only inthe modulation of mature lymphocyte functions (Sandersand Munson, 1984; Spengler et al., 1990; Felsner et al.,1995), but also in the regulation of thymocyte apoptosis(Ćupić et al., 2003) suggesting their possible role in thefine tuning of T-cell maturation.

    More recently, it has been shown that rat quiescentlymphocytes express the genes, not only for α2-ARs, butalso for α1-ARs (Schauenstein et al., 2000). However,according to the study of Rouppe and her collaborators(2002) α1-ARs are not detected in peripheral bloodmononuclear cells of healthy humans, but humanprimary lymphoid organs do express mRNA encodingthese receptors (Kavelaars, 2002). Therefore, it has beenassumed that the human lymphocytes express α1-ARs atcertain stages of development, but that their expressionis not detectable on the mature cells entering theperipheral circulation (Kavelaars, 2002). Moreover, ithas been reported that the expression of α1-ARs byimmune system cells can be modulated byglucocorticoid, ß-AR agonist (Rouppe et al., 1999) andcytokine TNF-α and IL-1ß (Heijnen et al., 2002) action.

    Very little is known on the role of α1-ARs in themodulation of the immune response. There is evidenceto suggest that α1-ARs are involved in the regulation ofhematopoiesis (Maestroni and Conti, 1994), anddendritic cell maturation and migration (Maestroni,2000). Moreover, it has been shown that the treatment offetal thymic organ culture with phenylephrine, an α1-ARagonist, evokes an increase in the thymocyteproliferative activity, and consequently, in cell yield(Singh, 1979).

    Since there are data indicating the possible role ofα1-AR signalling in the regulation of T-cell maturation,in the present study we examined the effects of chronicurapidil-induced α1-AR blockade on the rat thymusstructure and T-cell development during the peripubertalperiod, when the thymus in rats peaks in size and beginsto involute (Marchetti et al., 1990; Morale et al., 1991),and during the early adult period of life. Urapidil isknown as a postsynaptic α1-AR blocker with apharmacodynamic profile similar to prazosin (Langtry etal., 1989). Structural characteristics of the thymus afterchronic urapidil treatment were followed by sterologicalanalysis and its influence on thymocyte differentiationwas assessed by determining the relative proportion ofmain thymocyte subsets delineated by expression ofCD4/CD8 coreceptor molecules and TCRαß by two-colour and one-colour flow cytometric analysis,respectively.

    Material and methods

    Animals and treatment

    Male Wistar rats aged either 21 (BW: 70-80 g) or 75days (BW: 250-300 g) at the beginning of experimentwere maintained in our animal room under a standard12-h photoperiod, at 21±2°C, with free access to foodand water. The animals from both age groups wereassigned randomly (n=5/group) to receive urapidil orsaline. Over 15 consecutive days, they were injectedsubcutaneously (s.c.) with 0.20 mg/kg BW/day ofurapidil (Ebrantil, Byk Gulden, Germany) diluted insaline or an equivalent volume (1 ml/kg BW/day) ofsaline. The dose chosen was based on doses reported tobe effective at blocking other α1-adrenergic inducedeffects in vivo (Plećaš et al., 1996; Ittner et al., 2002).One hour after the final injection the animals wereindividually removed from their cages and anaesthesiawas induced in bell-jar using diethyl ether (LEK,Slovenia). After decapitation, the thymus was removed,dissected free of parathymic lymph nodes and adherentmembranous tissue and two lobes were divided andindividually weighed. The right lobus was used foranalysis of phenotypic characteristics of thymocytes,while the left lobus was processed for stereologicalanalysis.

    Preparation of thymic cell suspensions

    The thymic lobes were excised and placed inindividual Petri dishes containing ice-cold phosphate-buffered saline (PBS, pH 7.3). The thymocytesuspension was prepared by grinding the thymic tissuebetween the frosted ends of microscope slides andpassing the resultant suspension through a fine nylonmesh. Thus obtained single-cell suspension was washedthree times in ice-cold PBS containing 2% fetal calfserum (Gibco, Grand Island, N.Y., USA) and 0.01%sodium azide (PS medium). The cells were then countedin an improved Neubauer haemacytometer and celldensity was adjusted to 107 cells/ml by addition of PSmedium. The viability of such cell preparations, asdetermined by Trypan blue exclusion, was routinelygreater than 95%.

    Flow cytometry (FCA)

    Immunofluorescence staining of thymocytes wasperformed using two independent systems, including (a)direct two-colour staining with fluorescein isothicyanateFITC-conjugated anti CD4 (clone W3/25, Serotec,Oxford, UK) and phycoerythrin (PE)-conjugated anti-CD8 (clone MRC OX-8, Serotec) monoclonal antibodies(mAbs) and (b) indirect one-colour staining with biotin-conjugated mAb, directed at a constant determinant ofthe rat TCR (clone R73, Serotec), as first-step reagent,followed by FITC-conjugated streptavidin (BectonDickinson, Mountain View, Calif. USA), as second-step

    834

    Rat thymus and α1-adrenoceptor blockade

  • reagent. The immunostaining was carried out as it hasbeen previously reported (Leposavić et al., 2000).

    All samples were analyzed on the same day on aFACScan flow cytometer (Becton Dickinson). Deadcells and debris were excluded from analysis byselective gating based on anterior and right-angle scatter.104 flow cytometric events for the two-colour and 5x103flow cytometric events for the one-colour FCA wereanalyzed. The analyses were carried out with respect toappropriate isotypic and fluorochrome-matched controls,with Consort 30 and Lysis software (Becton Dickinson).

    Stereological analysis

    After fixation in Bouin’s solution, dehydratation in agraded series of ethyl alcohol and embedding in paraffin,the thymic tissue was serially cut at 5 µm thick sectionsand the sections were stained with haematoxylin andeosin. Every 40th section (20 sections per organ) wassubjected to analysis of different stereologicalparameters.

    Stereological measurements were performed bypoint and intersection counting method (Weibel, 1979;Karapetrović et al., 1995; Pejčić-Karapetrović et al.,2001; Plećaš-Solarović et al., 2004) using image analysissoftware (Micro Image Version 4.0, OLYMPUS). Thetest areas were randomly chosen, and each image,acquired using a digital camera, was saved, overlaid withthe corresponding grid and analyzed.

    Absolute volumes of the thymic cortex and medullawere estimated from the volume of the processed andembedded organ and volume density (Vv) ofcorresponding compartment. The volume of fixedthymic tissue was calculated from the fresh tissueweight, specific gravity (Casley-Smith, 1988) and thepercentage of tissue shrinkage (approximately 34%),which was determined stereologically (Plećaš-Solarovićet al., 2004). Thus, all morphometrical data refer to fixedthymic tissue. Vv of each thymic compartment wasdetermined under magnification of x40, using anorthogonal test grid with 100 points, and according tothe equation: Vv=Pf/Pt, where Pf is the number of testpoints hitting the analyzed structure and Pt is totalnumber of test points falling on the organ. Total numberof analyzed test-areas was 100 per animal.

    The total number of thymocytes in the thymiccompartment was calculated from numerical density(Nv) of thymocytes and the absolute volume of thatcompartment. The Nv of thymocytes, representing thenumber of cells per unite volume, was estimated underimmersion magnification, using a grid that correspondsto the multipurpose M42 test-system (Weibel, 1979).The test grid was placed randomly, but positionedparallel to, and just touching, the capsule for the outercortex and the cortico-medullary junction for the deepcortex analysis, respectively (Kendall and Al-Shawaf,1991). For estimating Nv of medullary thymocytes, thegrid was placed randomly throughout the medulla. Foreach thymic compartment 60 test-areas per animal were

    measured. Nv was calculated according to the equation:Nv=NA/(D+Go) where NA, as the number ofthymocytes per surface unit of test area, was estimatedfrom the relation: NA=N/At. N is the number of cells pertest area and At is actual size of the test area. D, themean caliper diameter of thymocytes, was calculated as:D=6Vv/Sv. Vv in this equation refers to volume densityof thymocytes, while Sv is the surface density, calculatedas: Sv=2If/Lt. If is the number of intersections of the testlines with the plasma membrane of thymocytes and Lt isthe total length of test lines. The depth sharpness (Go)was determined from the equation: Go=λ/(n+Na)2, λbeing the wavelength of light, n is the coefficient ofdiffraction of the immersion oil and Na, the numericalaperture of the objective lens.

    Statistical analysis

    The results are expressed as means ± SEM. Groupdifferences in the stereological parameters of the thymusas well as in the relative proportion of thymocytesubpopulations were analyzed by the nonparametricWilcoxon test, using Statistical Package for SocialScience (SPSS) Version 7.5. Significance was set atp

  • The decrease in thymus size in urapidil-treatedimmature rats reflected a significant (p

  • 837

    Rat thymus and α1-adrenoceptor blockade

    Fig. 5. Relative proportions of CD4-8- (A), CD4+8+ (B), CD4+8- (C) and CD4-8+ (C) thymocytes in sexually immature and adult rats injected withurapidil (striped bars) or saline (open bars) for 15 consecutive days. Results are means ± SEM for 5 animals in each group. Error bar less than 0.03% isomitted. * p

  • subsets of thymocytes were delineated as: 1) cells withlow level of TCRαß expression (TCRαßlow), 2) cellswith high level of TCRαß expression (TCRαßhigh) and3) cells expressing TCRαß at not detectable level(TCRαß-) (Fig. 6). In immature rats urapidilsignificantly influenced neither the percentage ofTCRαß- cells nor the relative proportions of the cellsexpressing detectable levels of TCRα (TCRαßlow andTCRαßhigh cells) (Fig. 7).

    Adult rats

    Thymus weight and total thymus cellularity

    In contrast to immature rats, the 15-day-long

    treatment with urapidil in adult rats did not affect thebody weight, but significantly (p

  • cortex. The enlargement of cortical volume in theseanimals was due to increased cellularity of thiscompartment. The augmented cellularity of thymuscortex may be the result of 1) increased entrance of pro-thymocytes and/or thymocyte proliferation and 2)decreased apoptosis and/or decelerated corticaldifferentiation. The data indicating that α1-ARs areinvolved in regulation of hematopoiesis (Maestroni andConti, 1994) may suggest an altered T-cell precursorimmigration into the thymus of animals subjected to α1-AR blockade. The reduced size of the corticalthymocytes does not speak in favour of an increasedthymocyte proliferation rate. As it has been describedthat chronic exposure of adult rats to norepinephrineenhanced thymocyte apoptosis via α-AR (Stevenson etal., 2001), it may be assumed that a reduced apoptosis is,at least partly, responsible for the increased thymus sizeand cellularity in adult urapidil-treated rats.

    Furthermore, the treatment with α1-AR blockersubstantially affected the distribution of the mainthymocyte subsets delineated by expression of CD4/8.The over-representation of the CD4-8- DNsubpopulation and the under-representation of theCD4+8+ DP subpopulation suggest the occurrence ofsuppression in thymocyte transition from DN to DPstage of development. In accordance with thisassumption is a reduction of the percentage of TCRαßlowthymocytes that are shown to be mainly CD4+8+ DPcells ready to enter into the positive selection process(Tscuchida et al., 1994). Additionally, it has been shownthat the treatment, compared with immature rats, in adultrats evoked the opposite effect on the relative proportionof SP cells, most likely, favouring the maturation ofCD4+8- SP cells over CD4-8+ thymocytes. This findingis consistent with the hypothesis that mature CD4+ cells,whether newly generated in the thymus or re-entrantsfrom periphery, exert a negative feed-back on the CD4-8- DN to CD4+8+ DP thymocyte transition and positivefeed-back on the maturation of DP cells toward CD4+8-SP cells (Mehr et al., 1997).

    Differental effect of the treatment on the thymusstructure and distribution of main thymocyte subsets inperipubertal and adult rats may be related to thesubstantial differences in the morphometriccharacteristics of thymuses and phenotypic profile ofthymocytes observed in intact rats of these ages(Leposavić et al., 1996; Plećaš-Solarović et al., 2004).

    Furthermore, as autoradiographic studiesdemonstrating that, not only density of ß-adrenoceptors,but also their distribution within the thymus,significantly changes with sexual maturation (Marchettiet al., 1990), it may be speculated that density and/ordistribution of α1-adrenoceptors are subjected todevelopmentally-induced changes which, in turn, may beresponsible for differential effects of α1-signaling insexually immature and mature rats. Finally, the presentfindings are consistent with data showing: a) differentialconcentration of the hormones influencing thymicstructure and function (i.e., gonadal steroids,

    839

    Rat thymus and α1-adrenoceptor blockade

    reduction (p

  • gonadotropins, GnRH) in peripubertal and adult rats andii) differential effects of these hormones on themorphometric characteristics of the thymus and on thethymopoiesis in these two groups of rats (Karapetrovićet al., 1995; Leposavić et al., 1996, 2005).

    These are the first data to show that systemic α1-ARblockade affects the structure of the rat thymus andthymocyte differentiation, the effects being age-dependent and, most likely, mediated through differentmechanisms in immature and adult animals. A moredetailed assessment of additional thymocytedifferentiation markers in combination with functionalmarkers of proliferation and apoptosis will help furthercharacterisation of the α1-AR blockade-induced age-dependent changes in the thymus. These data may be ofclinical importance as there are circumstances underwhich increased naïve T-cell generation by thymus isnecessary for successful recovery of the host, such asfollowing iatrogenic and disease-induced T-celldepletion (McFarland et al., 2001).

    Acknowledgements. This work is supported by The Ministry of Science,Development and Technology of Serbia, Grant No. 1239.

    References

    Benschop R.J., Rodriguez-Feuerhahn M. and Schedlowski M. (1996).Catecholamine-induced leukocytosis: early observations, currentresearch, and future directions. Brain Behav. Immun. 10, 77-91.

    Casley-Smith J.R. (1988). Expressing stereological results “per cm3” isnot enough. J. Pathol. 156, 263-265.

    Chan S.H., Cosgrove D., Waltzinger C., Benoist C. and Mathis D.(1993). Another view of the selective model of thymocyte selection.Cell 73, 225-236.

    Cunnick J.E, Lysle D.T., Kucinski B.J. and Rabin B.S. (1990). Evidencethat shock-induced immune suppression is mediated by adrenalhormones and peripheral beta-adrenergic receptors. Pharmacol.Biochem. Behav. 36, 645-651.

    Ćupić V., Čolić M., Jandrić D., Milojković B. and Varagić V.M. (2003).Xylazine, an alpha 2-adrenergic agonist, induces apoptosis of ratthymocytes and a thymocyte hybridoma line in vitro. Methods Find.Exp. Clin. Pharmacol. 25, 5-10.

    Elenkov I.J., Wilder R.L., Chrousos G.P. and Vizi E.S. (2000). Thesympathetic nerve - an integrative interface between twosupersystems: The brain and the immune system. Pharmacol. Rev.52, 595-638.

    Felsner P., Hofer D., Rinner I., Porta S., Korsatko W. and SchauensteinK. (1995). Adrenergic suppression of peripheral blood T cellreactivity in the rat is due activation of peripheral α2-receptors. J.Neuroimmunol. 57, 27-34.

    Fernández-López A. and Pazos A. (1994). Identification of alpha 2-adrenoceptors in rat lymph nodes and spleen: an autoradiographicstudy. Eur. J. Pharmacol. 252, 333-336.

    Heijnen C.J., Rouppe van der Voort C., Van de Pol M. and Kavelaars A.(2002). Cytokines regulate α1-adrenergic receptor expression inhuman monocytes and endothelial cells. J. Neuroimmunol. 125, 66-72.

    Ittner K.P., Bucher M., Zimmermann M., Grobecker H.E., Kramer B.K.

    and Taeger K. (2002). Effect of three different doses of urapidil onblood glucose concentrations in the streptozotocin diabetic rats. Eur.J. Anaesthesiol. 19, 504-509.

    Karapetrović B., Mićić M. and Leposavić G. (1995). Stereologicalanalysis of the sexually mature rat thymus after orchiectomy. IndianJ. Med. Res. 102, 42-48.

    Kavelaars A. (2002). Regulated expression of α-1 adrenergic receptorsin the immune system. Brain Behav. Immun. 16, 799-807.

    Kendall M.D. and Al-Shawaf A.A. (1991). Innervation of the rat thymusgland. Brain Behav. Immun. 5, 9-28.

    Langtry H.D., Mammen G.J. and Sorkin E.M. (1989). Urapidil. A reviewof its pharmacodynamic and pharmacokinetic properties, andtherapeutic potential in the treatment of hypertension. Drugs 38,900-940.

    Latta K., Krieg Jr. R.J., Carbajo-Pérez E., Carbajo S. and Chan J.C.M.(2002). Effects of deflazacort and cortisone on cellular proliferationin the rat thymus. Life Sci. 71, 1951-1960.

    Leposavić G., Karapetrović B., Obradović S., Vidić-Danković B. andKosec D. (1996). Differential effects of gonadectomy on thethymocyte phenotypic profile in male and female rats. Pharmacol.Biochem. Beh. 54, 269-276.

    Leposavić G., Plećaš B. and Kosec D. (2000). Differential effects ofchronic propranolol treatment on the phenotypic profi le ofthymocytes from immature and adult rats. Immunopharmacology 46,79-87.

    Leposavić G., Pekić S. and Kosec D. (2005). Gonadotropin-releasinghormone agonist administration affects the thymopoiesis in adultfemale rats independently on gonadal hormone production. Am. J.Reprod. Immunol. 53, 1-13.

    Li L., Hsu H.-C., William G.E., Stockard C.R., Ho K.-J., Lott P., Yang P.-A., Zhang H.-G. and Mountz J.D. (2003). Cellular mechanism ofthymic involution. Scand. J. Immunol. 57, 410-422.

    Lind E.F., Prockop S.E., Porritt H.E. and Petrie H.T. (2001). Mappingprecursor movement through the postnatal thymus reveals specificmicroenvironments supporting defined stages of early lymphoiddevelopment. J. Exp. Med. 194, 127-134.

    Madden K.S. (2003). Catecholamines, sympathetic innervation, andimmunity. Brain Behav. Immun. 17, S5-S10.

    Madden K.S. and Felten D.L. (2001). ß-Adrenoceptor blockade altersthymocyte differentiation in aged mice. Cell. Mol. Biol. 47, 189-196.

    Maestroni G.J.M. (2000). Dendritic cell migration controlled by α1b-adrenergic receptors. J. Immunol. 165, 6743-6747.

    Maestroni G.J. and Conti A. (1994). Noradrenergic modulation oflymphohematopoiesis. Int. J. Immunopharmacol. 16, 117-122.

    Marchetti B., Morale M.C. and Palletier G. (1990). Sympathetic nervoussystem control of rat thymus gland maturation: autoradiographiclocalization of the ß2-adrenergic receptor in the thymus andpresence of sexual dimorphism during ontogeny. Prog.NeuroEndocrinImmun. 3, 103-115.

    McFarland R.D., Picker L.J., Koup A.R. and Douek D.C. (2001).Recently identified measures of human thymic function. Clin. Appl.Immun. Rev. 2, 65-73.

    Mehr R., Perelson A.S., Fridkis-Hareli M. and Globerson A. (1997).Regulatory feedback pathways in the thymus. Immunol. Today 18,581-585.

    Morale M.C., Batticane N., Bartoloni G., Guarcello V., Farinella Z.,Galasso M.G. and Marchetti B. (1991). Blockade of central andperipheral luteinizing hormone-releasing hormone (LHRH) receptorsin neonatal rats with a potent LHRH-antagonist inhibits the

    840

    Rat thymus and α1-adrenoceptor blockade

  • morphofunctional development of the thymus and maturation of thecell-mediated and humoral immune responses. Endocrinology 128,1073-1085.

    Müller E.E., Locatelli V. and Cocchi D. (1999). Neuroendocrine controlof growth hormone secretion. Physiol. Rev. 79, 511-607.

    Pejčić-Karapetrović B., Kosec D. and Leposavić G. (2001). Differentialeffects of male and female gonadal hormones on the intrathymic Tcell maturation. Dev. Immunol. 8, 305-317.

    Plećaš B., Popović A., Glavaški A., Hristić M. and Solarović T. (1996).Stereologic study on the effect of prolonged urapidil-treatment onthe rat ventral prostate. Acta Vet. (Beograd) 46, 73-80.

    Plećaš-Solarović B., Lalić Lj. and Leposavić G. (2004). Age-dependentmorphometrical changes in the thymus of male propranolol-treatedrats. Ann. Anat. 186, 141-147.

    Rauški A., Kosec D., Vidić-Danković B., Radojević K., Plećaš-SolarovićB. and Leposavić G. (2003). Thymopoiesis following chronicblockade of beta-adrenoceptors. Immunopharm. Immunotox. 25,513-528.

    Rouppe van der Voort C, Heijnen C.J., Wulffraat N., Kuis W. andKavelaars A. (2002). Stress induces increases in IL-6 production byleucocytes of patients with the chronic inflammatory disease juvenilerheumatoid arthritis: a putative role for α1-adrenergic receptors. J.Neuroimmunol. 110, 223-229.

    Rouppe van der Voort C., Kavelaars A., van de Pol M. and Heijnen C.J.(1999). Neuroendocrine mediators up-regulate α1b- and α1d-adrenergic receptor subtypes in human monocytes. J.Neuroimmunol. 95, 165-73.

    Sanders V.M. and Munson A.E. (1984). Beta adrenoceptor mediation ofthe enhancing effect of norepinephrine on the murine primaryantibody response in vitro. J. Pharmacol. Exp. Ther. 230, 183-192.

    Sanders V.M. and Straub R.H. (2002). Norepinephrine, the ß-adrenergicreceptor, and immunity. Brain Behav. Immun. 16, 290-332.

    Schauenstein K., Felsner P., Rinner I., Liebmann P.M., Stevenson J.R.,

    Westermann J., Haas H.S., Cohen R.L. and Chambers D.A. (2000).In vivo immunomodulation by peripheral adrenergic and cholinergicagonists/antagonists in rat and mouse models. Ann. NY Acad. Sci.917, 618-627.

    Singh U. (1979). Effect of catecholamines on lymphopoiesis in fetalmouse thymic explants. J. Anat. 129, 279-292.

    Singh U., Millson D.S., Smith P.A. and Owen J.J. (1979). Identificationof ß-adrenoceptors during thymocyte ontogeny in mice. Eur. J.Immunol. 9, 31-35.

    Singh U. and Owen J.J. (1976). Studies on maturation of thymus stemcells. The effects of catecholamines, histamine, and peptidehormones on the expression of T cell alloantigens. Eur. J. Immunol.6, 59-62.

    Shortman K., Egerton M., Spangrude G.J. and Scollay R. (1990). Thegeneration and fate of thymocytes. Semin. Immunol. 2, 3-12.

    Spengler R.N., Allen R.M., Remick D.G., Strieter R.M. and Kunkel S.L.(1990) Stimulation of alpha-adrenergic receptor augments theproduction of macrophage-derived tumor necrosis factor. J.Immunol. 145, 1430-1434.

    Stachowiak A. and Malendowicz L.K. (1993). Stereologic studies on thelong-term effect of prazosin on rat adrenal cortex. Exp. Toxicol.Pathol. 45, 109-112.

    Stevenson J.R., Westermann J., Liebmann P.M., Hörtner M., Rinner I.,Felsner P., Wölfler A. and Schauenstein K. (2001). Prolonged alpha-adrenergic stimulation causes changes in leukocyte distribution andlymphocyte apoptosis in the rat. J. Neuroimmunol. 120, 50-57.

    Tsuchida M., Konishi M., Jojima K., Naito K., Fujikura Y. and FukumotoT. (1994). Analysis of cell surface antigens on glucocorticoid-treatedrat thymocytes with monoclonal antibodies. Immunol. Lett. 39, 209-217.

    Weibel E.R. (1979). Stereological methods. Practical methods forbiological morphometry. Vol 1. Academic Press. London. pp 1-415.

    Accepted March 24, 2005

    841

    Rat thymus and α1-adrenoceptor blockade