Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Br J Anaesth ; 120(1): 188-196, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29397129

ABSTRACT

BACKGROUND: Perioperative strategies can significantly influence long-term cancer outcomes. Dexmedetomidine, an α2-adrenoceptor agonist, is increasingly used perioperatively for its sedative, analgesic, anxiolytic, and sympatholytic effects. Such actions might attenuate the perioperative promotion of metastases, but other findings suggest opposite effects on primary tumour progression. We tested the effects of dexmedetomidine in clinically relevant models of dexmedetomidine use on cancer metastatic progression. METHODS: Dexmedetomidine was given to induce sub-hypnotic to sedative effects for 6-12 h, and its effects on metastasis formation, using various cancer types, were studied in naïve animals and in the context of stress and surgery. RESULTS: Dexmedetomidine increased tumour-cell retention and growth of metastases of a mammary adenocarcinoma (MADB 106) in F344 rats, Lewis lung carcinoma (3LL) in C57BL/6 mice, and colon adenocarcinoma (CT26) in BALB/c mice. The metastatic burden increased in both sexes and in all organs tested, including lung, liver, and kidney, as well as in brain employing a novel external carotid-artery inoculation approach. These effects were mediated through α2-adrenergic, but not α1-adrenergic, receptors. Low sub-hypnotic doses of dexmedetomidine were moderately beneficial in attenuating the deleterious effects of one stress paradigm, but not of the surgery or other stressors. CONCLUSIONS: The findings call for mechanistic translational studies to understand these deleterious effects of dexmedetomidine, and warrant prospective clinical trials to assess the impact of perioperative dexmedetomidine use on outcomes in cancer patients.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/toxicity , Colonic Neoplasms/pathology , Dexmedetomidine/toxicity , Hypnotics and Sedatives/toxicity , Lung Neoplasms/pathology , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis , Neoplasms, Experimental/pathology , Adenocarcinoma/pathology , Animals , Carcinoma, Lewis Lung/pathology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Rats, Inbred F344 , Receptors, Adrenergic, alpha-2/drug effects
2.
BMC Obes ; 4: 24, 2017.
Article in English | MEDLINE | ID: mdl-28690853

ABSTRACT

BACKGROUND: Obesity was identified as a major risk factor for malignant diseases, but underlying mechanisms remain unclear. Natural killer (NK) cells, a pivotal aspect of innate immunity, are capable of identifying and killing virally infected and tumor cells. Previous studies have shown altered NK cell functions in obesity, and the current study aimed to investigate the relationship between altered NK cell functions and increased cancer risk in obesity. METHODS: To induce obesity male F344-rats received a high-fat diet (34% fat) or a control diet (4% fat). Thereafter, syngeneic mammary adenocarcinoma cells (MADB106) or a vehicle were intravenously (i.v.) injected. 15 min after injection, half of each group of rats were killed, lungs removed and immunohistochemically stained. Numbers of NK cells, MADB106 cells and NK cell-tumor cell interactions were quantified. Twenty-one days after tumor-cell injection the other half group of rats was killed and lung metastases were counted and relative mRNA concentrations of different NK cell receptors were determined. RESULTS: After short-term MADB106-challenge, DIO fed animals showed significantly decreased NK cell numbers in the blood and NK cell-tumor cell interactions in the lung as compared to their control littermates. Twenty-one days after MADB106 injection, the lungs of the DIO fed rats showed significantly more lung metastases compared to control animals, accompanied by reduced relative mRNA concentrations of the activating NK cell receptor NKG2D. CONCLUSIONS: We conclude that induction of obesity in F344-rats leads to reduced lung NK cell function against tumor cells and results in significantly enhanced lung metastasis as compared to lean animals. It can be hypothesized that obesity-induced altered NK cell functions play an important role in cancer growth and metastasis.

3.
Brain Behav Immun ; 56: 209-220, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26944000

ABSTRACT

We recently reported that immune stimulation can be compromised if animals are simultaneously subjected to stressful conditions. To test the generalizability of these findings, and to elucidate neuroendocrine mediating mechanisms, we herein employed CpG-C, a novel TLR-9 immune-stimulating agent. Animals were subjected to ongoing stress (20-h of wet cage exposure) during CpG-C treatment, and antagonists to glucocorticoids, ß-adrenoceptor, COX2, or opioids were employed (RU486, nadolol, etodolac, naltrexone). In F344 rats, marginating-pulmonary NK cell numbers and cytotoxicity were studied, and the NK-sensitive MADB106 experimental metastasis model was used. In Balb/C mice, experimental hepatic metastases of the CT-26 colon tumor were studied; and in C57BL/6J mice, survival rates following excision of B16 melanoma was assessed - both mouse tumor models involved surgical stress. The findings indicated that simultaneous blockade of glucocorticoid and ß-adrenergic receptors improved CpG-C efficacy against MADB106 metastasis. In mice bearing B16 melanoma, long-term survival rate was improved by CpG-C only when employed simultaneously with blockers of glucocorticoids, catecholamines, and prostaglandins. Prolonged stress impaired CpG-C efficacy in potentiating NK activity, and in resisting MADB106 metastasis in both sexes, as also supported by in vitro studies. This latter effect was not blocked by any of the antagonists or by adrenalectomy. In the CT26 model, prolonged stress only partially reduced the efficacy of CpG-C. Overall, our findings indicate that ongoing behavioral stress and surgery can jeopardize immune-stimulatory interventions and abolish their beneficial metastasis-reducing impacts. These findings have implications for the clinical setting, which often involve psychological and physiological stress responses during immune-stimulation.


Subject(s)
Catecholamines/antagonists & inhibitors , Glucocorticoids/antagonists & inhibitors , Immunologic Factors/pharmacology , Killer Cells, Natural , Neoplasm Metastasis/prevention & control , Neoplasms/drug therapy , Oligodeoxyribonucleotides/pharmacology , Prostaglandin Antagonists/pharmacology , Stress, Psychological/immunology , Animals , Disease Models, Animal , Female , Immunologic Factors/administration & dosage , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oligodeoxyribonucleotides/administration & dosage , Rats , Rats, Inbred F344
4.
Brain Behav Immun ; 28: 128-38, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23153554

ABSTRACT

Surgery can suppress in vivo levels of NK cell cytotoxicity (NKCC) through various mechanisms, including catecholamine-, glucocorticoid (CORT)-, and prostaglandin (PG)-mediated responses. However, PGs are synthesized locally following tissue damage, driving proinflammatory and CORT responses, while their systemic levels are often unaffected. Thus, we herein studied the role of adrenal factors in mediating in vivo effects of PGs on NKCC, using adrenalectomized and sham-operated F344 rats subjected to surgery or PGE(2) administration. In vivo and ex vivo approaches were employed, based on intravenous administration of the NK-sensitive MADB106 tumor line, and based on ex vivo assessment of YAC-1 and MADB106 target-line lysis. Additionally, in vitro studies assessed the kinetics of the impact of epinephrine, CORT, and PGE(2) on NKCC. The results indicated that suppression of NKCC by epinephrine and PGE(2) are short lasting, and cannot be evident when these compounds are removed from the in vitro assay milieu, or in the context of ex vivo assessment of NKCC. In contrast, the effects of CORT are long-lasting and are reflected in both conditions even after its removal. Marginating-pulmonary NKCC was less susceptible to suppression than circulating NKCC, when tested against the xenogeneic YAC-1 target line, but not against the syngeneic MADB106 line, which seems to involve different cytotoxicity mechanisms. Overall, these findings indicate that elevated systemic PG levels can directly suppress NKCC in vivo, but following laparotomy adrenal hormones mediate most of the effects of endogenously-released PGs. Additionally, the ex vivo approach seems limited in reflecting the short-lasting NK-suppressive effects of catecholamines and PGs.


Subject(s)
Adrenal Cortex Hormones/physiology , Dinoprostone/physiology , Killer Cells, Natural/physiology , Adrenalectomy , Animals , Cell Line, Tumor , Epinephrine/pharmacology , Female , Flow Cytometry , Glucocorticoids/pharmacology , Killer Cells, Natural/drug effects , Laparotomy/adverse effects , Male , Rats , Rats, Inbred F344
5.
Brain Behav Immun ; 26(6): 996-1005, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22659252

ABSTRACT

Interleukin-12 (IL-12) is a major pro-inflammatory cytokine, which promotes cell-mediated immunity and T(H)1 differentiation. In vitro studies indicated suppression of IL-12 production by several stress-related factors, but no effects of behavioral stress were shown on plasma IL-12 levels. Therefore, in the current study we (i) examined the in vivo effects of various behavioral and pharmacological stress paradigms on baseline plasma IL-12 levels; (ii) compared these in vivo findings to those obtained following in vitro stimulation of leukocytes from the same rats; and (iii) assessed potential sexual dimorphism in these outcomes. The findings indicated that plasma IL-12 levels were significantly reduced by social confrontation, wet-cage exposure, surgery, and the administration of corticosterone, epinephrine, or prostaglandin-E(2). Notably, most in vivo impacts on plasma levels were not evident when assessed in vitro. The IL-12-reducing effects of wet-cage exposure, and of corticosterone and epinephrine administration, were significantly greater in males than in females, although females exhibited greater total corticosterone levels following stress. The duration of acute stressors predicted the degree of IL-12 reduction, but more prolonged stressors did not. Furthermore, seven days of alternating behavioral stressors reduced plasma IL-12 levels more than 14 days. These findings suggest animals' behavioral habituation to stress conditions, or a specific immune mechanism restricting the duration of IL-12 reduction. Overall, our findings indicate a generic and robust stress-induced reduction in plasma IL-12 levels, and suggest epinephrine, corticosterone, and prostaglandin-E(2), as potential mediators that should be scrutinized in vivo in the context of natural physiological stress responses.


Subject(s)
Interleukin-12/blood , Stress, Psychological/blood , Animals , Corticosterone/blood , Corticosterone/pharmacology , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Environment , Enzyme-Linked Immunosorbent Assay , Epinephrine/pharmacology , Female , Housing, Animal , Laparotomy , Male , Oligodeoxyribonucleotides/pharmacology , Rats , Rats, Inbred F344 , Rats, Long-Evans , Restraint, Physical , Sex Characteristics , Social Environment , Stress, Physiological/physiology , Swimming/psychology
6.
Int J Clin Pharmacol Ther ; 49(9): 545-54, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21888867

ABSTRACT

OBJECTIVE: COX inhibitors and ß-adrenergic blockers were recently shown to reduce cancer progression in animal models through various mechanisms. These include the prevention of immune suppression during the critical perioperative period, and the preclusion of direct promoting effects of catecholamines and prostaglandins on malignant tissue growth. To assess the safety of such pharmacological treatments in the context of oncologic surgery, the current study evaluates wound healing efficacy in the skin, muscle, and colon tissues in rats undergoing colonic anastomosis. METHODS: F344 rats were treated daily with a COX-2 inhibitor (etodolac), a ß-adrenergic blocker (propranolol), both drugs or vehicles. All rats underwent skin punch biopsy, and half were also subjected to laparotomy and colonic anastomosis. Tensile strength of the abdominal wall and colonic bursting pressure were assessed on Days 3, 7, and 30 postoperatively, and skin biopsy site healing was scored on Days 2, 4, and 6 postoperatively. RESULTS: None of the drug treatments produced any deleterious effects along the expected course of tissue healing. On Day 30, colon bursting pressure showed an abnormal strengthening in animals undergoing anastomosis compared to non-operated animals, across all drug treatments. This abnormal strengthening was attenuated by etodolac. In the skin, surgery reduced healing rate, irrespective of drug treatments. CONCLUSIONS: Effective doses of etodolac and propranolol caused no negative effects on wound healing processes in rats. The apparent safety of such treatments, together with their potential clinical benefits, suggests the incorporation of these treatments in oncologic patients undergoing curative tumor resection.


Subject(s)
Abdominal Wall/physiopathology , Adrenergic beta-Antagonists/pharmacology , Anastomosis, Surgical , Colon/surgery , Cyclooxygenase 2 Inhibitors/pharmacology , Etodolac/pharmacology , Laparotomy , Propranolol/pharmacology , Skin/physiopathology , Wound Healing/drug effects , Anastomosis, Surgical/adverse effects , Anastomosis, Surgical/mortality , Animals , Colon/physiopathology , Female , Laparotomy/adverse effects , Laparotomy/mortality , Male , Postoperative Complications/epidemiology , Rats , Rats, Inbred F344 , Tensile Strength , Weight Loss
7.
Acta Anaesthesiol Scand ; 50(10): 1223-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17067322

ABSTRACT

BACKGROUND: Anesthesiologists are a population at high risk of alcohol and drug abuse, depression, suicide, and psychiatric hospitalization. The impact of their working milieu on specific immune indices has scarcely been studied, and it is assumed that immune perturbations may contribute to some of the above risks. This study took advantage of an unplanned, 3-month long strike of anesthesiologists, and explored its relations to specific immune measures. METHODS: We assessed induced cytokine production and lymphocytes proliferative responses in blood samples taken from 10 anesthesiologists just before the strike and at its end, after a long period of markedly reduced workload. RESULTS: The results indicated that the proliferative responses to phytohemagglutinin (PHA) and concanavalin A (Con A) were significantly lower at the end of the strike. At this time point, we observed a significant decrease in the production of interleukin-6 (IL-6), IL-10 and IL1ra levels, and a significant increase in IL-2 production. A strong trend towards a decline in tumor necrosis factor-alpha (TNF-alpha) levels was evident, while levels of IL-1beta were unchanged. CONCLUSION: These findings suggest that the working conditions of anesthesiologists are associated with specific immune alterations, including a shift towards a Th2 cytokines' dominance, and an elevated pro-inflammatory cytokine response. A reduced Th1 profile has been related to increased susceptibility to infections, and high pro-inflammatory cytokine levels were recently proposed as etiological factors in cardiovascular diseases and in depression.


Subject(s)
Anesthesiology/methods , Anesthetics/pharmacology , Cytokines/biosynthesis , Lymphocyte Activation/drug effects , Th2 Cells/immunology , Adult , Female , Humans , Israel , Male , Middle Aged , Surveys and Questionnaires , Th2 Cells/drug effects
8.
Breast Dis ; 26: 99-114, 2006.
Article in English | MEDLINE | ID: mdl-17473369

ABSTRACT

Surgical resection of the primary tumor is a necessary and effective treatment for breast cancer patients. For various reasons discussed, we believe that the short postoperative period is critical for eliminating minimal residual disease (MRD), thus markedly impacting long term survival. Unfortunately, both animal and human studies have shown that surgery induces suppression of anti-metastatic cell-mediated immunity (CMI) at this critical period, which is suggested to worsen patients' prognosis. In this review we examine different aspects of the surgical procedure that cause immunosuppression (e.g., anesthesia and tissue damage), discuss their mediating humoral and cellular mechanisms, and suggest prophylactic interventions feasible in cancer patients to avoid postoperative suppression of CMI. The use of the suggested interventions has been shown to significantly reduce postoperative metastasis in animal models, including mammary adenocarcinoma, and initial data suggest similar efficacy in breast cancer patients. We believe that our recommended prophylactic interventions can easily be applied by health-care practitioners and hold promise in reducing long-term recurrence and metastasis in cancer patients.


Subject(s)
Breast Neoplasms/immunology , Breast Neoplasms/surgery , Immune Tolerance , Adjuvants, Immunologic/therapeutic use , Anesthetics/adverse effects , Breast Neoplasms/pathology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Female , Humans , Menstrual Cycle/immunology , Neoplasm Metastasis/immunology , Neoplasm Metastasis/prevention & control , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Postoperative Period , Risk Factors , Stress, Physiological/immunology , Stress, Physiological/prevention & control
9.
J Endocrinol Invest ; 26(10): 1013-22, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14759076

ABSTRACT

Among inbred strains of rats, the Fischer 344 (F344) is commonly used in immunological and behavioral studies. However, little is known about patterns of sex hormones and corticosterone (CORT) secretion throughout the estrous cycle in this strain, which is characterized by a marked CORT response to stress and variable length of cycles. In the current study, using radioimmunoassays, we assessed serum levels of progesterone, estradiol, LH, testosterone, prolactin and CORT, at 1-h intervals throughout the estrous cycle in F344 female rats with 4- and 5-day cycles, as well as in males. Vaginal smears were obtained from 268 females for 15 consecutive days to determine individual length of the estrous cycle and the exact estrous phase upon blood withdrawal, which was conducted once in each rat on the 12th day of smearing. The results indicated that both 4- and 5-day cyclers have two distinct and marked surges of progesterone, one on proestrus day and the other on diestrous-1 day. Testosterone levels in 5-day cyclers peaked on diestrus-3, one day earlier than in 4-day cyclers. Daily peak levels of CORT gradually increased from estrus day to proestrous day, whereas daily nadir levels of CORT remained unchanged. To simulate the natural kinetics of specific sex hormones in ovariectomized females, different doses of estradiol, progesterone, testosterone, prolactin or CORT were injected s.c. or i.p., or 90-day sustained release pellets containing different doses of estradiol or progesterone were implanted. The findings indicated dose- and time-dependent effects, suggesting regimens for modeling the estrous cycle or replacement therapy.


Subject(s)
Corticosterone/blood , Estrous Cycle/physiology , Gonadal Steroid Hormones/blood , Animals , Estradiol/blood , Female , Kinetics , Luteinizing Hormone/blood , Male , Ovariectomy , Progesterone/blood , Prolactin/blood , Rats , Rats, Inbred F344 , Testosterone/blood
10.
Int J Cancer ; 94(3): 401-6, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11745421

ABSTRACT

Natural killer cell cytotoxicity (NKCC) was reported to manifest a circadian rhythm, peaking during wakefulness in both human blood and rat spleen. Using F344 rats, we investigated whether such fluctuations (i) reflect changes in NK cell numbers or in cytotoxicity per cell; (ii) coincide in the blood and spleen; (iii) correspond with clearance of NK-sensitive tumor cells from the lungs and (iv) influence formation of lung metastases. Two rat groups were housed in opposite 12:12 hr lighting regimens. Two hours after the onset of light or dark, both groups were either sacrificed or intravenously inoculated with tumor cells to study the following indices: NKCC and NK cell numbers in the spleen (n = 29) and blood (n = 79), lung clearance of tumor cells (n = 142) and lung metastasis (n = 69). The tumor employed, MADB106, is an NK-sensitive mammary adenocarcinoma that metastasizes only to the lungs. The results indicated that, during the dark phase, splenic NKCC increased (37% higher lytic unit [LU](50)) mostly due to a 28.9% higher percentage of NK cells in the spleen. In contrast, blood NKCC decreased by 42.5% (LU(20)) and this decline was independent of circulating NK cell numbers, which remained constant. Lung tumor clearance increased in the dark (up to 42% lower retention 9 hr after inoculation), but no corresponding changes in the number of metastases were observed 3 weeks later. We conclude that diurnal changes in rats' NKCC are organ-specific, involve changes in both cell distribution and activity and may affect short-term in vivo indices of NK tumoricidal activity.


Subject(s)
Blood Cells , Killer Cells, Natural/cytology , Lung Neoplasms/metabolism , Spleen/cytology , Animals , Dose-Response Relationship, Drug , Female , Flow Cytometry , Humans , Light , Lung Neoplasms/secondary , Male , Neoplasm Metastasis , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Sex Factors , Spleen/metabolism , Time Factors
11.
Int Immunopharmacol ; 1(11): 1947-56, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11606026

ABSTRACT

The effects of anti-cancer number one (ACNO), a 19-herb Chinese formula used to treat cancer patients, were studied in F344 rats. In the first study, the number and activity of circulating NK cells were evaluated following 18 days of oral consumption of 0.1, 0.5, or 2 g/kg/day. The second study assessed the effect of ACNO on resistance to metastasis of the MADB106 tumor line, a syngeneic mammary adenocarcinoma that metastasizes only to the lungs and is highly sensitive to NK activity (NKA) in vivo. Resistance to metastasis was assessed under baseline conditions and following the administration of a beta-adrenergic agonist, metaproterenol (MP). MP was used to simulate sympathetic response to stressful conditions, and was previously shown to suppress resistance to MADB 106 metastasis. The results of the first study indicated a dose-dependent increase in NKA per ml of blood and per NK cell, with no significant changes in blood concentration of NK cells. In the second study, whereas MP caused a 4.5-fold increase in the number of metastases in untreated rats, only a 2.3-fold increase occurred in rats treated with ACNO. No significant improvement in baseline levels of resistance to metastasis was observed. These findings indicate the importance of studying ACNO under stressful conditions in patients with potentially metastasizing tumors. This may prove particularly important during the perioperative period, spanning from the detection of the primary tumor to postoperative treatment. During this critical period, psychological and physiological stress responses are known to cause massive immunosuppression, which was suggested to promote metastatic development.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drugs, Chinese Herbal/pharmacology , Killer Cells, Natural/drug effects , Neoplasm Metastasis/prevention & control , Adenocarcinoma/pathology , Adrenergic beta-Agonists/toxicity , Animals , Cell Count , Cytotoxicity Tests, Immunologic , Dose-Response Relationship, Drug , Female , Flow Cytometry , Killer Cells, Natural/immunology , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Metaproterenol/antagonists & inhibitors , Metaproterenol/toxicity , Neoplasm Metastasis/immunology , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Tumor Cells, Cultured , Weight Gain/drug effects
12.
Anesthesiology ; 94(6): 1066-73, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11465599

ABSTRACT

BACKGROUND: The perioperative period is characterized by a state of immunosuppression, which was shown in animal studies to underlie the promotion of tumor metastasis by surgery. As this immunosuppression is partly ascribed to the neuroendocrine stress response, the authors hypothesized that spinal blockade, known to attenuate this response, may reduce the tumor-promoting effect of surgery. METHODS: Fischer-344 rats were subjected to a laparotomy during general halothane anesthesia alone or combined with either systemic morphine (10 mg/kg) or spinal block using bupivacaine (50 microg) with morphine (10 microg). Control groups were either anesthetized or undisturbed. Blood was drawn 5 h after surgery to assess number and activity of natural killer cells, or rats were inoculated intravenously with MADB106 adenocarcinoma cells, which metastasize only to the lungs. Metastatic development was assessed by quantifying lung retention of tumor cells 24 h after inoculation or by counting pulmonary metastases 3 weeks later. RESULTS: Laparotomy conducted during general anesthesia alone increased lung tumor retention up to 17-fold. The addition of spinal block reduced this effect by 70%. The number of metastases increased from 16.7 +/- 10.5 (mean +/- SD) in the control group to 37.2 +/- 24.4 after surgery and was reduced to 10.5 +/- 4.7 during spinal block. Systemic morphine also reduced the effects of surgery, but to a lesser degree. Natural killer cell activity was suppressed to a similar extent by surgery and by anesthesia alone. CONCLUSIONS: The addition of spinal blockade to general halothane anesthesia markedly attenuates the promotion of metastasis by surgery.


Subject(s)
Anesthesia, Spinal , Laparotomy/adverse effects , Neoplasms/pathology , Nerve Block , Analgesics, Opioid/pharmacology , Anesthesia, General , Animals , Flow Cytometry , Killer Cells, Natural/drug effects , Lung/pathology , Male , Morphine/pharmacology , Neoplasm Metastasis/pathology , Pain Measurement/drug effects , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
13.
Gynecol Oncol ; 81(2): 254-62, 2001 May.
Article in English | MEDLINE | ID: mdl-11330959

ABSTRACT

OBJECTIVES: The aim of this study was to study the impact of sex, the menstrual cycle, and the use of oral contraceptives (OC) on the number and activity of natural killer (NK) cells. METHODS: Both the number and the activity of NK cells were assessed per milliliter of blood, and NK activity (NKA) per NK cell and per lymphocyte was calculated. NKA was measured in each subject using a whole blood assay, which preserves the plasma and all blood cells, and using a washed blood assay, in which plasma is replaced with an artificial medium. The subjects were young (20-29 years old) women with a regular menstrual cycle (n = 39; 26 tested on both the follicular and the luteal phases), age-matched women who use OC (n = 26), and age-matched men (n = 20). RESULTS: Men showed markedly and significantly higher NKA than women with regular menstrual cycles or women using OC, who had the lowest levels of NKA. No significant differences in blood concentration of NK cell were found. Differences in NKA were of similar magnitude in the whole and washed blood assays per milliliter of blood, per NK cell, or per lymphocyte. The menstrual cycle had no significant effect on activity levels of NK cells, but during the periovulatory phase, the number of NK cells per milliliter of blood increased significantly. CONCLUSIONS: The observed differences are independent of the presence of serum factors during the in vitro assessment of NKA, but may be related to chronic exposure to sex steroids and to fluctuation in the NK cell expression of beta-adrenoceptors.


Subject(s)
Contraceptives, Oral, Synthetic/pharmacology , Killer Cells, Natural/immunology , Menstrual Cycle/immunology , Adult , Contraceptives, Oral, Synthetic/immunology , Cytotoxicity, Immunologic/drug effects , Estradiol/blood , Female , Follicular Phase/blood , Granulocytes/cytology , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Leukocyte Count , Leukocytes/cytology , Luteal Phase/blood , Luteinizing Hormone/blood , Lymphocytes/cytology , Male , Menstrual Cycle/blood , Progesterone/blood , Sex Factors
14.
Pain ; 90(1-2): 191-9, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11166986

ABSTRACT

We have previously shown in rats that the provision of analgesic doses of morphine significantly reduces the tumor-promoting effects of undergoing and recovering from surgery. Because morphine had no effect in non-operated animals, and because a single preoperative dose given hours before tumor inoculation was effective, we have suggested that it is the pain-relieving effects of the drug that underlies its beneficial impact. To support and strengthen this suggestion, two different regimens of analgesia were employed, the systemic administration of the more selective mu-agonist, fentanyl, and the intrathecal (i.t.) administration of bupivacaine plus morphine. To assess host resistance against metastasis, we used a lung clearance assay of the MADB106 mammary adenocarcinoma, a natural killer (NK)-sensitive syngeneic cell line that metastasizes only to the lungs. Female and male Fischer 344 rats were randomly assigned to one of four groups using a 2x2 experimental design: experimental laparotomy under halothane anesthesia versus anesthesia alone, by drug treatment versus vehicle. In the first in vivo experiment, fentanyl was administered 20 min before surgery (40 microg/kg subcutaneously (s.c.)), and at the end of surgery in a slow-release suspension (20 microg/kg s.c.). In the second in vivo experiment, bupivacaine (10 microg) plus morphine (20 microg) in 50 microl was administered i.t. before surgery. Surgery resulted in a 3- to 4-fold increase in the lung retention of MADB106 cells in both males and females, and the observed surgery-induced increase in lung tumor retention was reduced by more than 65% in the fentanyl-treated animals and more than 45% in the animals receiving i.t. bupivacaine plus morphine. Neither drug regimen exerted effects in the anesthesia only animals. Surgery also resulted in a significant suppression of whole blood NK activity assessed at 5 h postoperatively, the same time point at which MADB106 tumor cells were inoculated in the in vivo studies. Unlike the in vivo study, fentanyl suppressed NK activity at this time point in non-operated rats, but had no effect in operated rats. Taken together, these findings strengthen the suggestion that the management of perioperative pain is a critical factor in preventing surgery-induced decreases in host resistance against metastasis. If similar relationships between pain and metastasis occur in humans, then pain control must become a priority in the postoperative care of individuals with cancer.


Subject(s)
Analgesics, Opioid/administration & dosage , Anesthetics, Local/administration & dosage , Bupivacaine/administration & dosage , Fentanyl/administration & dosage , Lung Neoplasms/secondary , Morphine/administration & dosage , Pain, Postoperative/immunology , Surgical Procedures, Operative/adverse effects , Adenocarcinoma , Analgesics, Opioid/pharmacology , Animals , Antiviral Agents , Exploratory Behavior/drug effects , Female , Idoxuridine , Injections, Spinal , Killer Cells, Natural/metabolism , Laparotomy , Lung Neoplasms/blood , Lung Neoplasms/immunology , Male , Pain, Postoperative/blood , Pain, Postoperative/drug therapy , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
15.
Neuroimmunomodulation ; 8(3): 154-64, 2000.
Article in English | MEDLINE | ID: mdl-11124582

ABSTRACT

Although acute stress has been reported to suppress natural killer cell activity (NKA) and host resistance to metastasis, it is unclear whether the sympathetic nervous system (SNS) has a role in these effects. The current study in Fischer 344 rats assessed the involvement of adrenal catecholamines and beta(1)- and beta(2)-adrenoceptors in mediating these deleterious effects of swim stress. In addition to assessing the number and activity of NK cells following swim stress, we used a tumor model based on the MADB106 mammary adenocarcinoma line: this syngeneic tumor metastasizes only to the lungs, and its lung tumor retention (LTR) and metastatic colonization are highly sensitive to NKA. The findings indicate that stress increased both LTR, assessed 24 h after inoculation, and the number of lung metastases, counted 3 weeks later. These effects were attenuated or completely abolished by the ganglionic blocker chlorisondamine (3 mg/kg i.p.), by adrenal demedullation, by a selective beta-adrenergic antagonist (nadolol, 0.4 mg/kg), and additively by a selective beta(1)- (atenolol, 1-6 mg/kg) and a selective beta(2)-antagonist (either butoxamine 4-32 mg/kg or ICI-118,551 0.3-8 mg/kg). Stress also suppressed NKA, and adrenal demedullation prevented this suppression. Administration of adrenaline (0.1-1 mg/kg) or of a beta-adrenergic agonist (metaproterenol, 0.8 mg/kg), in physiologically relevant doses, suppressed NKA in a dose-dependent manner, and increased LTR to levels characteristic of swim stress. Taken together, these findings suggest that acute stress, by releasing catecholamines from the adrenal glands and activating beta(1)- and beta(2)-adrenoceptors, suppresses NKA and consequently compromises resistance to NK-sensitive metastasis.


Subject(s)
Adrenal Medulla/physiology , Catecholamines/physiology , Killer Cells, Natural/immunology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/immunology , Receptors, Adrenergic, beta-1/physiology , Receptors, Adrenergic, beta-2/physiology , Stress, Physiological/immunology , Adrenal Medulla/immunology , Adrenergic beta-1 Receptor Antagonists , Adrenergic beta-2 Receptor Antagonists , Animals , Disease Models, Animal , Epinephrine/metabolism , Female , Killer Cells, Natural/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Male , Mammary Neoplasms, Experimental/metabolism , Metaproterenol/metabolism , Rats , Rats, Inbred F344 , Stress, Physiological/metabolism , Swimming , Sympathetic Nervous System/metabolism , Sympathomimetics/metabolism , Tumor Cells, Cultured/transplantation
16.
Br J Cancer ; 83(12): 1630-6, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11104557

ABSTRACT

Physiological responses that involve adrenergic mechanisms, such as stress-induced changes in cardiovascular indices, were reported to fluctuate along the menstrual cycle. Metastatic development following surgery was also reported to vary according to the menstrual phase during which a primary breast tumour was removed. Natural killer (NK) cells are believed to play an important role in controlling metastases. Our recent studies in rats demonstrated that adrenergic suppression of NK activity and of resistance to metastasis is more profound during oestrous phases characterized by high levels of oestradiol. In the current study in humans, we examined the in vitro impact of a beta-adrenergic agonist, metaproterenol (MP), on NK activity, comparing blood drawn from (a) women tested at 3-4 different phases of their menstrual cycle (n = 10), (b) women using oral contraceptives (OC) (n = 10), and (c) men (n = 7). NK activity in each blood sample was assessed in the presence of 5 different concentrations of MP (10(-8)M to 10(-6)M), and in its absence (baseline). The results indicated marked group differences in the magnitude of NK suppression by MP: EC(50)was 2. 6-fold lower in the luteal phase compared to the follicular phase, and 1.8-fold lower in OC users compared to men, who were least susceptible to the effects of MP. No significant group differences or menstrual effects in baseline levels of NK activity were evident. These findings provide the first empirical evidence for menstrual regulation of adrenergic impact on cellular immune competence. Relevance of these findings to the relation between the timing of breast cancer excision within the menstrual cycle and survival rates is discussed.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Contraceptives, Oral/pharmacology , Killer Cells, Natural/drug effects , Menstrual Cycle/physiology , Metaproterenol/pharmacology , Adult , Analysis of Variance , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Fibrinolysis/drug effects , Humans , K562 Cells , Killer Cells, Natural/immunology , Male , Middle Aged , Sex Factors , Time Factors
17.
Br J Cancer ; 83(12): 1747-54, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11104576

ABSTRACT

Clinical observations suggest that the rate of metastatic development and long-term mortality following surgery in breast cancer patients is influenced by the menstrual phase during which surgery is conducted. The menstrual cycle is known to modulate various physiological responses and medical conditions that involve adrenergic mechanisms (e.g., asthma). Natural killer activity (NKA), an immune function controlling metastasis, is suppressed following surgery, and in vitro by adrenaline. We therefore hypothesize that the clinical observation may be partly attributable to surgery-induced adrenergic suppression of NK-dependent resistance to metastasis, a suppression that depends on menstrual phase during surgery. To test this hypothesis in rats, 140 F344 females at different phases of their oestrous cycle were injected with a beta-adrenergic agonist, metaproterenol (MP) (0.4 or 0.8 mg kg(-1), s.c.), or with vehicle, before i.v. inoculation with MADB106 tumour cells. This syngeneic mammary adenocarcinoma line metastasizes only to the lungs, and is highly sensitive to NKA. In a second experiment, the suppression of NKA by MP was studied in vitro in blood drawn at different phases of the oestrous cycle (n = 36). Finally, the effects of stress on the number and activity of NK cells were assessed along the oestrous cycle (n = 71). The findings indicate that the suppressive effects of MP on resistance to metastasis and on NKA, are significantly greater during the oestrous phase characterized by high oestradiol levels (D3/proestrus/oestrus). Similarly, NKA per cell was suppressed by stress only during this phase. In untreated animals, in which inadvertent stress was minimized, no effects of the oestrous cycle on NKA or on resistance to metastasis were evident. These findings indicate that the oestrous cycle modulates adrenergic suppression of NKA and of resistance to metastasis. The relevance of these findings to the above clinical observation, as well as that of our related findings in women from a parallel study, is discussed.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Estrus/physiology , Killer Cells, Natural/drug effects , Metaproterenol/pharmacology , Animals , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Drug , Estradiol/pharmacology , Female , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis/prevention & control , Rats , Rats, Inbred F344 , Swimming , Time Factors , Tumor Cells, Cultured
18.
Brain Behav Immun ; 14(3): 153-69, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10970677

ABSTRACT

Schizophrenia has been associated with altered immunity and reduced occurrence of autoimmune diseases and malignancies. A few studies in schizophrenic patients have assessed natural killer cell activity (NKA), but no consistent findings have emerged. However, NKA was assessed using standard procedures and in the absence of autologous serum and the various cytokines that modulate NKA and appear to be abnormal in schizophrenic patients. In the current study, therefore, the number of NK cells and the activity of the individual NK cell were assessed in whole blood shortly after blood withdrawal, in both the presence and the absence of autologous serum. Twenty-nine schizophrenic patients (11 nonmedicated), 8 nonschizophrenic control patients (bipolar and personality disorders), and 31 age-matched healthy controls were studied. Schizophrenic patients showed higher NKA per NK cell than controls and nonschizophrenic patients. This difference remained significant even when the nonmedicated schizophrenics, who showed the highest levels of NKA, were excluded. However, the increase in NKA was more pronounced in the presence of serum and was reduced to an insignificant level when serum was removed from the same samples. In both schizophrenic patients and controls, smokers and women showed lower NKA. Numbers of NK cells did not differ among groups, although medication affected blood concentration of other leukocytes. These findings indicate that the effects of serum factors, psychiatric medication, gender, and smoking should be considered when assessing NKA in schizophrenic patients. The observed higher NKA may help explain the surprising reports of low incidence of lung cancer and other malignancies in schizophrenic patients, despite their higher rate of smoking.


Subject(s)
Antipsychotic Agents/therapeutic use , Killer Cells, Natural/physiology , Schizophrenia/blood , Schizophrenia/physiopathology , Smoking/adverse effects , Adult , Bipolar Disorder/blood , Bipolar Disorder/drug therapy , Bipolar Disorder/physiopathology , Cell Count/drug effects , Female , Humans , Killer Cells, Natural/pathology , Leukocytes/pathology , Male , Middle Aged , Personality Disorders/blood , Personality Disorders/drug therapy , Personality Disorders/physiopathology , Schizophrenia/drug therapy , Sex Characteristics , Tumor Cells, Cultured
19.
J Cell Biochem ; 79(3): 407-15, 2000 Sep 07.
Article in English | MEDLINE | ID: mdl-10972978

ABSTRACT

In this study, we used a rat model to investigate the effects of gonad hormones and replacement therapy on bone structure and the immune system. In the first phase of the study, 3- and 11-month-old F344 rats underwent ovariectomy (OVX) or were sham operated. Three months later, severe osteopenia was histologically observed in OVX rats of both age groups. The changes in the bone marrow structure of OVX rats included deterioration of cancellous bone that was associated with a remarkable increase of adipocyte cells. Furthermore, differential analyses for the expression of cell surface antigens by lymph-myeloid cells was studied using flow cytometry (FACS). The number of myeloid cells expressing ED-9(+) or CD-44(+) was similar in both age groups, and unaffected by OVX. However, an augmentation of T-lymphoid cells expressing CD4(+), CD5(+), or both, were observed with age, as well as after OVX. In the second phase of the study, 11-month-old rats were divided into five experimental groups: sham-operated, OVX, and OVX treated with sustained-release pellets of 17beta-estradiol (OVX-E), progesterone (OVX-P), or both (OVX-E/P). Hormone replacement therapy maintained low physiological levels, and rats were tested 12 weeks after treatment initiation. Administration of 17beta-E, with or without the addition of progesterone, prevented the rise of T lymphoid cells observed in OVX rats, whereas progesterone alone had no effect. In agreement with findings from the first phase, neither OVX nor replacement therapy affected the myeloid cells expression of ED-9 or CD-44. In summary, the cellular changes in the bone marrow of OVX rats were associated with an increase in adipocytes that was correlated with bone atrophy. An augmentation of T-lymphopoiesis was noted with increase in age or after OVX. This increase was reversed to baseline levels by 17beta-E treatment.


Subject(s)
Bone Marrow Cells/pathology , Bone and Bones/pathology , Estradiol/pharmacology , Estrogen Replacement Therapy , Osteoporosis, Postmenopausal/pathology , Progesterone/pharmacology , Adipocytes/pathology , Animals , Antigens, Differentiation/analysis , Antigens, Surface/analysis , Atrophy , Bone Marrow Cells/chemistry , Bone Marrow Cells/drug effects , Bone and Bones/chemistry , Bone and Bones/drug effects , Delayed-Action Preparations , Estradiol/therapeutic use , Female , Femur/chemistry , Femur/drug effects , Femur/pathology , Flow Cytometry , Hematopoiesis/drug effects , Humans , Male , Models, Animal , Myeloid Cells/chemistry , Myeloid Cells/drug effects , Myeloid Cells/pathology , Osteoporosis, Postmenopausal/metabolism , Osteoporosis, Postmenopausal/therapy , Ovariectomy/adverse effects , Progesterone/therapeutic use , Rats , Rats, Inbred F344 , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/pathology
20.
J Immunol Methods ; 239(1-2): 25-34, 2000 May 26.
Article in English | MEDLINE | ID: mdl-10821944

ABSTRACT

The function of natural killer (NK) cells is often studied by assessing in vitro levels of NK cell mediated lysis of target cells, or by assessing in vivo levels of lung tumor cell retention or metastatic colonization of intravenously injected tumor cells. However, these methods do not permit direct quantification and visualization of NK cells and their targets in vivo and in situ. Here, a new approach is described to visualize effector-to-target interactions as well as to estimate total numbers of targets in the lung, in vivo and in situ. MADB106 tumor cells were vitally labeled using carboxyfluorescein (CFSE) and intravenously (i.v.) injected into Fischer 344 rats (10(6) cells/rat). This mammary adenocarcinoma derived cell line is syngeneic to the inbred Fischer 344 rat and highly sensitive to NK cell activity in vivo. Effector-to-target interactions were visualized by immunostaining. Using the optical fractionator method, total numbers of CFSE-labeled MADB106 tumor cells were estimated in the left lung of the animals 5 min after tumor inoculation. To further demonstrate the usefulness of this approach in reflecting in vivo processes, rats were inoculated with MADB106 cells and simultaneously with a single i.v. bolus of either 1 microg/kg adrenaline or saline. Both lungs were removed 5 min later. Adrenaline caused a significant 80% reduction in the total number of lung CFSE-labeled MADB106 tumor cells, suggesting a rapid modulation of metastasis by stress hormones. This new approach facilitates the monitoring of effector-to-target interactions and the quantification of immune cell function or tumor adhesion in vivo and in situ.


Subject(s)
Adenocarcinoma/immunology , Fluoresceins , Fluorescent Dyes , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Animals , Cell Count , Injections, Intravenous , Lung Neoplasms/secondary , Male , Neoplasm Transplantation , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...