Laboratory of Clinical and Environmental Endocrinology and Immunology, Wadsworth Center, New York State Department of Health, Albany, New York 12201
Received January 22, 2003; accepted May 3, 2003
![]() |
ABSTRACT |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Key Words: acute cold/restraint stress; host resistance; Listeria monocytogenes; CD4/mice; nitric oxide.
![]() |
INTRODUCTION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Studies using chronic physical and psychological stress models have attempted to explore the underlying cellular and molecular mechanisms associated with stress-induced changes in host resistance. For example, daily 12-h restraint stress repeated over 7 days caused increased susceptibility to Listeria monocytogenes (LM) infection accompanied by multiple immunosuppressive effects, including inhibition of leukocyte migration into infected foci, suppression of LM-induced MHC class II expression, and down-regulation of IFN and IL-12, with concomitant up-regulation of IL-4 and IL-6 gene expression (Zhang et al., 1998
). Therefore, decreased leukocyte infiltration, reduced antigen presentation, and dysregulated Th1/Th2 balance all appear to be involved in stress-impaired host resistance. Studies with repeated restraint stress and herpes simplex virus-1 (HSV-1) infection (Bonneau et al., 1991
) demonstrated that suppression of NK cells and antigen-specific cytotoxic T cell function contributed to the chronic stress-induced susceptibility to HSV-1.
In contrast to the situation for chronic stress, there have been few reports on the effects of acute physical and psychological stressors on host resistance. Previously, others reported that inescapable tail shocks cause reduced inflammatory responses after injection of killed Escherichia coli (Deak et al., 1999). Acute restraint stress-induced leukocyte redistribution led to a significantly enhanced delayed-type hypersensitivity response due to locally accumulated lymphocytes (Dhabhar and McEwen, 1998
). In our previous studies, acute cold/restraint stress (ACRS) for 1 h significantly suppressed host resistance to LM in BALB/c mice accompanied with greater inflammatory responses (Cao and Lawrence, 2002
; Cao et al., 2002
). This inhibition was mediated through the peripheral sympathetic nervous system (SNS) via ß1-adrenergic receptors (ADR) (Cao et al., 2003
). Despite these studies, it is still uncertain how acute stress affects host resistance against infectious diseases at the cellular/molecular level. This is especially important to investigate because environmental physical and psychological stressors can have profound effects on health, and these stressors generally are greater in populations of lower socioeconomic status, in which there are increased health concerns.
Listeria monocytogenes is a gram-positive, facultative intracellular bacillus, widespread in nature (Mielke et al., 1997). Mouse LM infection is a well-established model for studying host resistance to infection (Finelli et al., 1999
; North and Conlan, 1998
). Primary immunity to LM involves both innate and adaptive immunity (mainly cell-mediated immunity) (Busch and Pamer, 1999
; Finelli et al., 1999
; North and Conlan, 1998
; Unanue, 1997b
). Innate immunity is critical for the early control of infection. Neutrophils accumulate quickly (increased within 24 h after infection) at infectious foci and phagocytose extracellular bacteria, along with tissue-fixed macrophages (e.g., Kupffer cells) and infiltrating monocytes/macrophages. Upon phagocytosis of bacteria, the macrophages become activated and produce a variety of cytokines (including pro-inflammatory cytokines IL-6, IL-1, and TNF
), chemokines, and NO; activate NK cells (innate responses) via IL-12; and begin to activate CD4+ T cells (adaptive responses) through antigen presentation and IL-12. NK cells and differentiated Th1 cells are major sources of IFN
, which can fully activate macrophages to become effective killers. In addition, Th1 cells can activate CD8+ T cells to become cytotoxic T cells and kill infected cells. Thus, CD4+ T cell activation is critical in the initiation of adaptive immune response; and macrophages, Th1, and cytotoxic T cells (mainly CD8+ T cells) are the major effector cells during host defense against LM.
Here, following our previous studies, we further investigated ACRS-induced inhibition of host resistance to LM in BALB/c mice at the cellular level and the involvement of ß-ADR. To evaluate the involvement of different cell components, we utilized CD4/ mice, examined in vivo kinetic changes in splenic leukocyte phenotypes and serum NO levels, and assessed splenic cell proliferation and cytokine production in vitro.
![]() |
MATERIALS AND METHODS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
For ACRS, as described previously (Cao and Lawrence, 2002; Cao et al., 2002
), 10-week-old mice were individually restrained in well-ventilated plastic 60-ml syringes (Sherwood Medical Company, St. Louis, MO) at 4°C for 1 h. Mice can move forward and backward in the syringe but cannot turn head to tail. The ACRS was always performed between 10 A.M. and noon. ACRS is considered a physical and psychological stressor. Control mice were left in their original cages undisturbed during the same time period. The day at which mice were given ACRS was considered day 0 for all experiments. All animal procedures were approved by the IACUC of the Wadsworth Center (protocol 00-278).
Drug administrations.
Both ß-ADR antagonists, atenolol and propranolol, were purchased from Sigma (St. Louis, MO). Drugs were dissolved in sterile saline and injected ip according to individual body weight (BW). The nonselective ß-ADR antagonist propranolol was given at 20 mg/kg BW, and the selective ß1-ADR antagonist atenolol was given at 20 mg/kg BW. Either drug or vehicle control were injected into the experimental mice immediately before ACRS. The doses were chosen based on our previous studies (Cao et al., 2003), in which both drugs significantly eliminated the inhibition of host resistance by ACRS. In addition, both propranolol (cross-bloodbrain barrier; BBB) and nadolol (not cross-BBB) were used in the previous study. No difference in host resistance (i.e., organ LM levels at day 3 after infection) was seen between these two drugs, indicating that the ACRS-induced inhibition of host resistance is mediated through peripheral but not central ß-ADRs. Thus, either drug can be used in this study. We chose propranolol here. No toxic effects were evident after any drug treatments.
LM infection and determination of viable LM.
Listeria monocytogenes was originally isolated from an LM meningitis patient and was passaged in mice for multiple (>9) generations before LM stocks were prepared, as described previously (Cao et al., 2002). Mice were intravenously injected with 56 x 103 colony-forming units (CFU) LM/mouse on day 0 immediately after ACRS. Mice were sacrificed at the indicated times postinfection. Upon sacrifice, blood was collected after CO2 anesthesia by cardiac puncture, and sera were obtained by centrifugation and kept at 20°C for later cytokine detection by enzyme-linked immunosorbent assay (ELISA) or NO determination. Spleens and livers were removed aseptically and homogenized for enumeration of viable LM, as described before (Cao and Lawrence, 2002
; Cao et al., 2002
). LM burdens in the organs are presented as viable LM per organ. Spleen homogenates were further centrifuged after addition of 1% NP-40 (Sigma), and the supernatants were kept at 20°C until cytokine analysis.
Analysis of splenic leukocyte phenotype by flow cytometry.
Following ACRS and/or infection as described above, at the indicated time, mice were anesthetized with CO2, and spleens were removed aseptically and weighed. Single-cell suspensions were prepared by grinding the tissue between two frosted microscope slides (Erie Scientific Company, Portsmouth, NH). Red blood cells were lysed with RBC-lysing buffer (0.15 M NH4Cl, 10 mM KHCO3, and 0.1 mM Na2EDTA, pH 7.27.4). Cells were washed twice with PBS, then total cell numbers were determined with a Coulter Counter ZM (Coulter Corp., Miami, FL). Cells (2 x 106 per tube) were used for further staining. All monoclonal antibodies were purchased from PharMingen (San Diego, CA): purified antimouse CD16/CD32 Fc receptor, PerCP-antimouse CD3, PE-antimouse CD69, PE-antimouse CD19, PE-antimouse CD11b, FITC-antimouse CD8, FITC-antimouse panNK, and FITC-antimouse I-Ad. Briefly, cells were first treated with antimouse CD16/CD32 Fc receptor (1 µg/tube, in 100 µl staining buffer: 2% FBS and 0.09% NaN3 in PBS) for 15 min on ice to block Fc receptors, then stained with a combination of two or three antimouse monoclonal antibodies (1 µg/tube for each) for 30 min on ice. After washing twice with PBS, cells were analyzed by a FACScan flow cytometer with Cell Quest software (Becton Dickinson, San Jose, CA). The following combinations of antibodies were used: (tube 1) CD3, CD69, and CD8, with CD3+/CD8 representing CD4+ T cells, CD3+/CD8+ representing CD8+ T cells, and CD69+ (expressed on activated T, B, NK cells and macrophages) used to identify activation of the T cell subsets and CD3 cells; (tube 2) CD3, CD19, and panNK, with CD3-/CD19+ representing B cells and CD3/panNK+ representing NK cells; and (tube 3) CD11b and I-Ad, with CD11b+/I-Ad+ cells representing macrophages, CD11b+/I-Ad cells representing neutrophils, and the intensity of I-Ad on macrophages representing macrophage activation. Cell numbers of each cell population were calculated based on the total cell numbers and percentage of each cell population. Activation of T cells and of CD3 cells was expressed as the percentage of CD69+ cells within an indicated cell population.
In Vitro spleen cell culture and proliferation assay.
Spleen single-cell suspensions were prepared from mice immediately after nonstressed control or ACRS time period but without LM infection, as described in the preceding section. Heat-killed LM (HKLM) was prepared by heating living LM at 70°C for 1 h, and the number of colonies was determined by culturing of a small portion of living LM before heat inactivation as described. Unfractionated single-cell splenic leukocyte preparations (2 x 105/well/200 µl) were stimulated with HKLM at various concentrations (0, 105, 106, and 107 CFU/well) in complete RPMI1640 media for 72 h in a low-O2 incubator (37°C, 7% CO2, and 5% O2). During the last 6 h of incubation, cells were incubated with 0.5 µCi [3H]-thymidine (25 µl/well; Perkin Elmer, Boston, MA). After incubation, cells were harvested with a semiautomatic cell harvester (Skatron, AS, Norway), and radioactive signals were then analyzed with a FUJIX BAS2000 phosphorimager (Fuji, Minamiashigara City, Japan). The average of the photostimulating luminescence (PSL, manufacturers units for the intensity of radioactivity) minus background PSL (PSL-Bkg) of triplicate wells was calculated. The proliferation was expressed as the percentage of the average PSL-Bkg of treated wells, relative to the average PSL-Bkg of nonstimulated wells, which was set at 100%. In addition, similar spleen cell cultures were set up at the same time for cytokine analysis. Following incubation (72 h), the supernatants were collected after centrifugation and were stored at 20°C until quantification of cytokines by ELISA.
Cytokine ELISA.
IL-6 was determined by DuoSet ELISA development kit (R&D, Minneapolis, MN) according to manufacturers protocols. The sensitivity of the kit is 3 pg/ml. IFN
and IL-4 were detected with antibody pairs (Caltag, Burlingame, CA) following a modified protocol (Heo et al., 1996
). Briefly, 96-well easy-wash EIA/RIA plates (Costar, Corning, NY) were coated with 2 µg/ml capture antibody (50 µl/well) in coating buffer (0.1 M NaHCO3, pH 8.2) and incubated at 4°C overnight. The next day, plates were washed with PBS/0.05% Tween 20, and nonspecific binding was blocked with 10% FBS/PBS (200 µl/well) for 2 h at room temperature. Then after washing, diluted samples and standards were loaded, and plates were incubated at 4° C overnight. On the third day, detection antibody was added at 1.5 µg/µl in 10% FBS/PBS (50 µl/well) after washing of the plates. Following 45 min incubation at room temperature, avidin-HRP (Sigma) was added (1:400, 50 µl/well), followed by 30 min incubation at room temperature. Next, plates were washed again, and TMB substrate (PharMingen) was added (50 µl/well). After 10 min development in the dark, 2 N H2SO4 stop solution was added (25 µl/well), and plates were read at 450 nm with an EL310 ELISA reader (Bio-Tek, Burlington, VT). IFN
or IL-4 concentrations were calculated, based on the standard curve. The sensitivity of these assays is 75 pg/ml for IFN
and 15 pg/ml for IL-4. Culture supernatant and serum cytokine levels were expressed in pg/ml, and splenic cytokines were normalized based on individual protein concentration (i.e., pg/mg protein), which is determined by BCA protein assay reagents (Pierce, Rockford, IL) following manufacturers protocol.
Serum NO determination.
Serum collected from nonstressed control and ACRS mice at various time points after LM infection were analyzed for NO content with an NOA280 nitric oxide analyzer (Sievers Instruments, Inc., Boulder, CO) following manufacturers operation manual. Serum samples were deproteinized by precipitating with cold absolute ethanol (sample/ethanol = 1:3 in volume). The supernatants were used for NO analysis. Nitrate standards were prepared in a range from 0 to 10 mM. The detection limit is 1 pM. The results were calculated from the standards and presented as µM of NO for each sample.
Statistical analysis.
For comparison of multiple groups, appropriate ANOVA was used initially. If a significant main effect or interaction was identified by the ANOVA analysis (p < 0.05), respective group means were compared using the Student-Newman-Keuls post hoc test; p < 0.05 was considered a significant difference.
![]() |
RESULTS |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
|
|
|
|
|
|
|
![]() |
DISCUSSION |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
|
On the other hand, the absence of ACRS-induced suppression in CD4/ mice may suggest that CD4+ T cells themselves or other immune responses affected by activated CD4+ T cells are inhibitory under ACRS conditions. T cell, especially helper T cell, functions were then further investigated. No propranolol-induced changes were observed with CD4/ mice, suggesting that this ß-ADR blocker may act either directly or indirectly on CD4+ T cells or on a downstream immune component following CD4+ T cell activation. These accumulative findings suggest a negative or at least nonpositive role for CD4+ T cells. CD4+ T cells appear to be required for inhibition of host resistance by ACRS, possibly via inhibition of effector CD8+ T cells. Alternatively, the ACRS-induced suppression may be dependent on the CD4/CD8 ratio or the absolute number of CD8+ T cells throughout the host, in that host resistance to LM is suppressed when the numbers of CD8+ T cells are low (WT mice) but not when they are elevated (CD4/ mice).
To evaluate helper T cell functions further (Fig. 8, right side), we examined the CD4+ T cell phenotype after infection in vivo and the in vitro HKLM-stimulated splenocyte proliferation and cytokine production by cells from nonstressed and ACRS mice. Equal expansion and enhanced activation (represented by CD69 expression) in vivo were observed in ACRS mice. In agreement, splenocyte proliferation and elevated IFN
production were observed in vitro with cells from ACRS mice. Altogether, these indicate that CD4+ T cells from ACRS mice can be activated, can proliferate, and can together with NK cells release reasonable amounts of IFN
to activate macrophages further. Therefore, lack of activation of helper T cells and/or lack of IFN
do not appear to contribute to the ACRS-inhibited host resistance. In addition, the ß-ADR antagonists propranolol and atenolol blocked enhancement of CD4+ T cell activation in ACRS mice, consistent with the idea that the SNS is responsible for the ACRS-impaired host resistance via ß1-ADRs (Cao et al., 2002
, 2003
). The association between inhibited host resistance and increased CD4+ T cell activation possibly could be due to: (1) ACRS-induced preferential development of Th2 cells, leading to predominant humoral responses and inhibition of the cell-mediated responses needed to defend against LM infection or (2) regulatory/suppressor T cell (CD4+CD25+ T cells) activity (Thornton and Shevach, 1998
) enhanced by ACRS. However, either of these possibilities requires further direct investigation.
In terms of the major effector cells, macrophages, and cytotoxic T cells (effectors downstream of CD4+ [helper] T cell activation, Fig. 8), there was similar CD8+ T cell expansion, higher activation of CD8+ T cells, no reduction of MHC class II expression on splenic macrophages, and no suppression of NO production in ACRS mice, in comparison with the nonstressed mice. These outcomes suggest normal activation of macrophages and cytotoxic CD8+ T cells, and comparable NO-induced bactericidal activity by macrophages. In fact, both CD8+ T cell activation and serum NO levels were higher in ACRS mice, correlating with the enhanced CD4+ T cell activation discussed earlier. Thus, these studies have detected no significant dysfunction in either of these effector cells under ACRS conditions. ACRS-induced inhibition of host resistance does not seem to be due to qualitative depression of cell-mediated defenses.
Use of CD8/ mice or cytotoxic T cell activity assays will help to evaluate further the cytotoxic T cell function in ACRS-induced changes of host resistance, but other possible defects in bacteria killing also need to be considered. Studies using MHC class I-deficient mice demonstrated that MHC class I peptide recognition is critical for cytotoxic T cell-specific killing of infected cells (Ladel et al., 1994; Seaman et al., 1999
). Interestingly, MHC class I has also been associated with nonimmune functions, such as central nervous system development (Corriveau et al., 1998
). Moreover, MHC class I exhibited molecular associations with several hormone receptors, including insulin receptors, epidermal growth factor receptors, ß-ADRs, and muscarinic cholinergic receptors. Antibodies directed against MHC class I could stimulate ß-ADRs and could induce the downstream intracellular secondary messengers, resulting in physiological changes in the targeted cells (Gremaschi and Sterin-Borda, 1994
). Therefore, we speculate that ACRS-induced activation of ß-ADRs could affect MHC class I expression on LM infected cells, and lack of effective targeting on infected cells could lead to an inefficient elimination of infected bacteria by cytotoxic T cells and, thus, a higher LM burden in ACRS mice. However, with this scenario it is not obvious why CD4+ T cells would be needed to observe the ACRS-induced inhibition of host resistance.
An alternative scenario could be that there is increased growth of LM in response to the stress hormone NE, leading to the higher LM burdens in ACRS mice. Certain bacteria, such as E. coli, have been reported to respond to NE, showing an enhanced growth (Freestone et al., 2000; Lyte and Ernst, 1992
). However, our previous study showed similar NE levels in spleen and liver in nonstressed and ACRS mice (Cao et al., 2002
), and other studies have shown that NE has no effects on LM growth rate in vitro (Rice et al., 2001
). Furthermore, it is not obvious why such a direct effect would occur only between days 2 and 3 after infection (Cao and Lawrence, 2002
).
In summary, ACRS inhibition of host resistance does not seem to be due to suppressive effects on innate immunity, inhibition of the bactericidal activity of macrophages, lack of T cell activation, or inhibited IFN production. However, the suppression of host resistance by ACRS is strongly associated with the presence of CD4+ T cells. The major defense pathway following CD4+ T cell activation needs to be fully investigated in order to uncover the mechanisms by which ACRS inhibits host resistance. It is most interesting that ß-ADR antagonists (in WT mice) and absence of CD4+ T cells eliminated the suppressive effects of ACRS on host resistance to LM. Furthermore, blockage of ß-ADR signaling lowered the number of LM-activated (CD69+) lymphocytes in nonstressed and ACRS-treated mice while improving host resistance. Clearly, activated lymphocytes are needed to defend against infection, suggesting that the mechanisms responsible for the ACRS-induced suppression must involve regulatory mechanisms still unresolved; however, CD4+ T cells and ß-ADRs are implicated.
![]() |
ACKNOWLEDGMENTS |
---|
![]() |
NOTES |
---|
2 Present address: Department of Medicine, SUNY Upstate Medical School, Syracuse, NY 13210.
3 To whom correspondence should be addressed at Wadsworth Center, New York State Department of Health, Empire State Plaza, P.O. Box 509, Albany, NY 12201-0509. Fax: (518) 474-1412. E-mail: david.lawrence{at}wadsworth.org.
![]() |
REFERENCES |
---|
![]() ![]() ![]() ![]() ![]() ![]() ![]() |
---|
Bhardwaj, V., Kanagawa, O., Swanson, P. E., and Unanue, E. R. (1998). Chronic Listeria infection in SCID mice: Requirements for the carrier state and the dual role of T cells in transferring protection or suppression. J. Immunol. 160, 376384.
Biondi, M., and Zannino, L.-G. (1997). Psychological stress, neuroimmunomodulation, and susceptibility to infectious diseases in animals and man: A review. Psychother. Psychosom. 66, 326.[ISI][Medline]
Bonneau, R. H., Sheridan, J. F., Feng, N., and Glaser, R. (1991). Stress-induced suppression of herpes simplex virus (HSV)-specific cytotoxic T lymphocyte and natural killer cell activity and enhancement of acute pathogenesis following local HSV infection. Brain Behav. Immun. 5, 170192.[ISI][Medline]
Brown, D. H., and Zwilling, B. S. (1994). Activation of the hypothalamic-pituitary-adrenal axis differentially affects the anti-mycobacterial activity of macrophages from BCG-resistant and susceptible mice. J. Neuroimmunol. 53, 181187.[CrossRef][ISI][Medline]
Busch, D. H., Kerksiek, K. M., and Pamer, E. G. (2000). Differing roles of inflammation and antigen in T cell proliferation and memory generation. J. Immunol. 164, 40634070.
Busch, D. H., and Pamer, E. G. (1999). T lymphocyte dynamics during Listeria monocytogenes infection. Immunol. Lett. 65, 9398.[CrossRef][ISI][Medline]
Cao, L., Filipov, N. M., and Lawrence, D. A. (2002). Sympathetic nervous system plays a major role in acute cold/restraint stress inhibition of host resistance to Listeria monocytogenes. J. Neuroimmunol. 125, 94102.[CrossRef][ISI][Medline]
Cao, L, Hudson, C. A., and Lawrence, D. A. (2003). Acute cold/resistant stress inhibits host resistance to Listeria monocytogenes via ß1-adrenergic receptors. Brain Behav. Immun. 17, 121133.[CrossRef][ISI][Medline]
Cao, L., and Lawrence, D. A. (2002). Suppression of host resistance to Listeria monocytogenes by acute cold/restraint stress: Lack of direct IL-6 involvement. J. Neuroimmunol. 133, 132143.[CrossRef][ISI][Medline]
Corriveau, R. A., Huh, G. S., and Shatz, C. J. (1998). Regulation of class I MHC gene expression in the developing and mature CNS by neuronal activity. Neuron 21, 505520.[ISI][Medline]
Deak, T., Nguyen, K. T., Fleshner, M., Watkins, L. R., and Maier, S. F. (1999). Acute stress may facilitate recovery from a subcutaneous bacterial challenge. Neuroimmunomodulation 6, 344354.[CrossRef][ISI][Medline]
Dhabhar, F. S., and McEwen, B. S. (1998). Enhancing versus suppressive effects of stress hormones on skin immune function. Proc. Natl. Acad. Sci. U.S.A. 96, 10591064.
Finelli, A., Kerksiek, K. M., Allen, S. E., Marshall, N., Mercado, R., Pilip, I., Busch, D. H., and Pamer, E. G. (1999). MHC class I restricted T cell responses to Listeria monocytogenes, an intracellular bacteria pathogen. Immunol. Res. 19, 211223.[ISI][Medline]
Freestone, P. P., Lyte, M., Neal, C. P., Maggs, A. F., Haigh, R. D., and Williams, P. H. (2000). The mammalian neuroendocrine hormone norepinephrine supplies iron for bacterial growth in the presence of transferrin or lactoferrin. J. Bacteriol. 182, 60916098.
Fukui, Y., Sudo, N., Yu, X.-N., Nukina, H., Sogawa, H., and Kubo, C. (1997). The restraint stress-induced reduction in lymphocyte cell number in lymphoid organs correlates with the suppression of in vivo antibody production. J. Neuroimmunol. 79, 211217.[CrossRef][ISI][Medline]
Gremaschi, G. A., and Sterin-Borda, L. (1994). MHC class Ihormone receptor associations: Still a physiological enigma? Acta. Physiol. Pharmacol. Ther. Latinoam. 44, 5564.[Medline]
Heo, Y., Parsons, P. J., and Lawrence, D. A. (1996). Lead differentially modifies cytokine production in vitro and in vivo. Toxicol. Appl. Pharmacol. 138, 149157.[CrossRef][ISI][Medline]
Iwakabe, K., Shimada, M., Ohta, A., Yahata, T., Ohmi, Y., Habu, S., and Nishimura, T. (1998). The restraint stress drives a shift in Th1/Th2 balance toward Th2-dominant immunity in mice. Immunol. Lett. 62, 3943.[CrossRef][ISI][Medline]
Kizaki, T., Oh-Ishi, S., and Ohna, H. (1996). Acute cold stress induces suppressor macrophages in mice. J. Appl. Physiol. 81, 393399.
Komori, T., Fujiwara, R., Shizuya, K., Miyahara, S., and Nomura, J. (1996). The influence of physical restraint or fasting on plaque-forming cell response in mice. Psychiatry Clin. Neurosci. 50, 295298.[ISI][Medline]
Ladel, C. H., Flesch, I. E., Arnoldi, J., and Kaufmann, S. H. (1994). Studies with MHC-deficient knock-out mice reveal impact of both MHC I- and MHC II-dependent T cell responses on Listeria monocytogenes infection. J. Immunol. 153, 31163122.
Li, T., Harada, M., Tamada, K., Abe, K., and Komoto, K. (1997). Repeated restraint stress impairs the antitumor T cell response through its suppressive effect on Th1-type CD4+ T cells. Anticancer Res. 17, 42594268.[ISI][Medline]
Lu, Z. W., Song, C., Ravindran, A. V., Merali, Z., and Anisman, H. (1998). Influence of a psychogenic and neurogenic stressor on several indices of immune function in different strains of mice. Brain Behav. Immun. 12, 722.[CrossRef][ISI][Medline]
Lyte, M., and Ernst, S. (1992). Catecholamine induced growth of gram-negative bacteria. Life Sci. 50, 203212.[CrossRef][ISI][Medline]
Mielke, M. E. A., Held, T. K., and Unger, M. (1997). Listeriosis. In Pathology of Infectious Diseases (D. H. Connor, F. W. Chandler, D. A. Schwartz, H. J. Manz, and E. E. Lack, Eds.), vol. 1, pp. 621633. Appleton & Lange, Stamford, CT.
North, R. J., and Conlan, J. W. (1998). Immunity to Listeria monocytogenes. Chem. Immunol. 70, 120.[ISI][Medline]
Okimura, T., Ogawa, M., Yamauchi, T., and Sasaki, Y. (1986). Stress and immune responses IV. Adrenal involvement in the alteration of antibody responses in restraint-stressed mice. Jpn. J. Pharmacol. 41, 237245.[ISI][Medline]
Rahemtulla, A., Fung-Leung, W. P., Schilham, M. W., Kundig, T. M., Sambhara, S. R., Narendran, A., Arabian, A., Wakeham, A., Paige, C. J., Zinkernagel, R. M., et al. (1991). Decreased helper cell activity in mice lacking CD4. Nature 353, 180184.[CrossRef][ISI][Medline]
Rice, P. A., Boehm, G. W., Moynihan, J. A., Bellinger, D. L., and Stevens, S. Y. (2001). Chemical sympathectomy increases the innate immune response and decreases the specific immune response in the spleen to infection with Listeria monocytogenes. J. Neuroimmunol. 114, 1927.[CrossRef][ISI][Medline]
Saperstein, A., Brand, H., Audhya, T., Nabriski, D., Hutchinson, B., Rosenzweig, S., and Hollander, C. S. (1992). Interleukin 1ß mediates stress-induced immunosuppression via corticotropin-releasing factor. Endocrinology 130, 152158.[Abstract]
Seaman, M. S., Perarnau, B., Lindahl, K. F., Lemonnier, F. A., and Forman, J. (1999). Response to Listeria monocytogenes in mice lack of MHC class Ia molecules. J. Immunol. 162, 54295436.
Thornton, A. M., and Shevach, E. M. (1998). CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J. Exp. Med. 188, 287296.
Unanue, E. R. (1997a). Inter-relationship among macrophages, natural killer cells and neutrophils in early stages of Listeria resistance. Curr. Opin. Immunol. 9, 3543.[CrossRef][ISI][Medline]
Unanue, E. R. (1997b). Studies in Listeriosis show the strong symbiosis between the innate cellular system and the T cell response. Immunol. Rev. 158, 1125.[ISI][Medline]
Wu, W., Yamaura, T., Murakami, K., Murata, J., Matsumoto, K., Watanabe, H., and Saiki, I. (2000). Social isolation stress enhanced liver metastasis of murine colon 26-L5 carcinoma cells by suppressing immune responses in mice. Life Sci. 66, 18271838.[CrossRef][ISI][Medline]
Yang, E. V., and Glaser, R. (2000). Stress-induced immunomodulation: Impact on immune defenses against infectious disease. Biomed. Pharmacother. 54, 245250.[CrossRef][ISI][Medline]
Zhang, D., Kishihara, K., Wang, B., Mizobe, K., Kubo, C., and Nomoto, K. (1998). Restraint stress-induced immunosuppression by inhibiting leukocyte migration and Th1 cytokine expression during the intraperitoneal infection of Listeria monocytogenes. J. Neuroimmunol. 92, 139151.[CrossRef][ISI][Medline]
Zwilling, B. S., Brown, D. H., and Pearl, D. (1992). Induction of major histocompatibility complex class II glycoproteins by interferon-: Attenuation of the effects of restraint stress. J. Neuroimmunol. 37, 115122.[CrossRef][ISI][Medline]
Zwilling, B. S., Dinkins, M., Christner, R., Faris, M., Griffin, A., Hilburger, M., McPeek, M., and Pearl, D. (1990). Restraint stress-induced suppression of major histocompatibility complex class II expression by murine peritoneal macrophages. J. Neuroimmunol. 29, 125130.[CrossRef][ISI][Medline]
|