Influence of Neuroleptics on Cytotoxic Activity of Rat Natural Killer Cells

1998 ◽  
Vol 11 (2) ◽  
pp. 57-62
Author(s):  
A.J. Madej ◽  
J. Kowalski ◽  
D. Belowski ◽  
Z. S. Herman

The aim of the study was to evaluate the in vivo and in vitro effects of three neuroleptics (chlorpromazine, haloperidol, and sulpiride) on the activity of rat spleen NK cells. In the in vivo experiment, rats were injected with different intraperitoneal doses of neuroleptics given once, for 14 or 28 days. In the in vitro experiment rat spleen NK cells were cultured in medium containing two different concentrations of neuroleptics for three days. The cytotoxic activity of NK cells was evaluated by measuring 51Cr release from YAC-1 target cells after 4-hour incubation. We also measured, using fluorescein-labelled anti-NK monoclonal antibody, the percentage of NK cells in the splenocyte population before and after single intraperitoneal injections of neuroleptics. In the in vitro experiment, both haloperidol (1×10−5 M and 1×10−6 M) and sulpiride (1.5×10−3 M and 1.5×10−4 M) induced a statistically significant decrease in the cytotoxic activity of NK cells. The lower dose of chlorpromazine (6×10−6 M) decreased the cytotoxic activity of NK cells, while the higher dose (6×10−5 M) did not. In the in vivo experiment, both single and repeated doses of chlorpromazine (2 mg /kg /day), haloperidol (0.5 mg/kg/day) and sulpiride (50 mg/kg/day) increased NK cell activity. That effect reflected an increase in NK cell activity but not in the number of NK cells. The study has shown that the immunomodulatory effect of neuroleptics on NK cell activity depends mainly on drug concentrations and experimental conditions.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4122-4122
Author(s):  
Katja Sockel ◽  
Claudia Schönefeldt ◽  
Sieghart Sopper ◽  
Martin Wermke ◽  
Marc Schmitz ◽  
...  

Abstract Abstract 4122 The hypomethylating agent azacytidine (AZA) represents the standard treatment for many high-risk MDS and AML patients. While the clinical efficacy has been confirmed in several studies, the precise molecular mechanism of action has not been fully understood yet. Human NK-cells play an important role in the regulation of immune responses against malignant cells. Their function is controlled by a complex interplay of activating and inhibitory receptors - some of them being regulated by methylation of the respective genes. We, therefore explored, whether AZA modulates in vitro NK-cell function as well as in vivo during minimal-residual disease (MRD)-guided treatment of imminent relapse in MDS and AML patients treated within the prospective RELAZA trial (NCT00422890). Methods: After purifying NK-cells of healthy donors by MACS (magnetic cell sorting), NK-cells were exposed in vitro to different concentrations of AZA (100nM, 1μM, 3μM) with or without IL-2. In parallel, the NK-cell phenotype of patients (n=12) with AML or MDS, undergoing MRD-guided treatment with AZA after stem cell transplantation was monitored by FACS from peripheral blood samples on day 1, 5 and 7 of the first and second AZA cycle. All patients were still in complete haematological remission at the time of therapy. Results: In vitro, we observed a significant reduction (3,1% to 1,8% p=0.028) of the immature and cytokine-regulating CD56bright NK-cell subpopulation with increasing concentrations of AZA. There was a trend towards a reduced expression of the death-ligand TRAIL, the activating receptors NKG2D and NKp46 and for an increased expression of the inhibitory KIR CD158b1/b2, whereas we could not detect any changes in the expression of FAS-L, Perforin, Granzyme B, NKp30, NKp44, CD69, CD57, DNAM-1, CD16, and NKG2A-CD94. Confirmatory, we observed a significant decrease in the expression of TRAIL (p=0.003), NKG2D (p=0.03) and NKp46 (p=0.006) during AZA treatment in-vivo. Interestingly, these changes appeared to be reversible. The observed reduction of NK-cell activating receptors and TRAIL during AZA treatment correlated with a reduction or stable course of MRD in all analyzed patients. Conclusion: In summary these data suggest that the clinical effects of AZA are not mediated by enhancing NK-cell activity. In fact, the drug may have inhibitory effects on NK-cell function which should be considered when applying AZA in the post-transplant setting. Disclosures: Platzbecker: Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.


2016 ◽  
Vol 17 (1) ◽  
pp. 31-40 ◽  
Author(s):  
Kaito Sakaguchi ◽  
Ming Zhong ◽  
Saeko Kawai ◽  
Yoshio Shimizu ◽  
Eiichi Gohda

A reduced number and/or reduced activity of natural killer (NK) cells, which are important for defense against a variety of cancers and viral infections, occur under various stress conditions and in patients with various diseases. In this article, we report that the 30% to 50% ethanol precipitate of oyster extract (EPOE50) dose-dependently enhanced the activity of mouse spleen NK cells in vitro and in vivo. The activity of EPOE50 was eluted with a molecular weight of about 2000 by gel filtration and was inactivated by periodate but not by proteinase K. The activity of highly purified NK cells was also augmented by EPOE50 but not by oligodeoxyribonucleotide 1585, which mimics bacterial DNA. Administration of EPOE50 to mice stimulated splenic NK cell activity without a change in splenic NK cell populations. Although the proliferation of B16 tumor cells in vitro was slightly stimulated by EPOE50, the growth of B16 melanoma in vivo was dose-dependently suppressed by administration of EPOE50. Taken together, our results indicate that EPOE50 augmented NK cell activity and that its administration to mice inhibited tumor growth presumably through the activation of NK cells and also suggest that the active substance is a sugar-containing oligomer or polymer and is not of bacterial origin.


2020 ◽  
Vol 39 (5) ◽  
pp. 477-490
Author(s):  
Attalla Farag El-kott ◽  
Ali S. Alshehri ◽  
Heba S. Khalifa ◽  
Abd-El-karim M. Abd-Lateif ◽  
Mohammad Ali Alshehri ◽  
...  

This study investigated whether the mechanism underlying the neurotoxic effects of cadmium chloride (CdCl2) in rats involves p66Shc. This study comprised an initial in vivo experiment followed by an in vitro experiment. For the in vivo experiment, male rats were orally administered saline (vehicle) or CdCl2 (0.05 mg/kg) for 30 days. Thereafter, spatial and retention memory of rats were tested and their hippocampi were used for biochemical and molecular analyses. For the in vitro experiment, control or p66Shc-deficient hippocampal cells were treated with CdCl2 (25 µM) in the presence or absence of SP600125, a c-Jun N-terminal kinase (JNK) inhibitor. Cadmium chloride impaired the spatial learning and retention memory of rats; depleted levels of glutathione and manganese superoxide dismutase; increased reactive oxygen species (ROS), tumor necrosis factor α, and interleukin 6; and induced nuclear factor kappa B activation. Cadmium chloride also decreased the number of pyramidal cells in the CA1 region and induced severe damage to the mitochondria and endoplasmic reticulum of cells in the hippocampi of rats. Moreover, CdCl2 increased the total unphosphorylated p66Shc, phosphorylated (Ser36) p66Shc, phosphorylated JNK, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, cytochrome c, and cleaved caspase-3. A dose–response increase in cell death, ROS, DNA damage, p66Shc, and NADPH oxidase was also observed in cultured hippocampal cells treated with CdCl2. Of note, all of these biochemical changes were attenuated by silencing p66Shc or inhibiting JNK with SP600125. In conclusion, CdCl2 induces hippocampal ROS generation and apoptosis by promoting the JNK-mediated activation of p66Shc.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3274-3274
Author(s):  
Luca Vago ◽  
Barbara Forno ◽  
Elisabetta Zino ◽  
Simona Di Terlizzi ◽  
Maria T. Lupo Stanghellini ◽  
...  

Abstract Haploidentical Hematopoietic Stem Cell Transplantation (haplo-HSCT) is a promising therapeutic option for patients lacking a fully compatible donor. Due to extensive T cell depletion, Natural Killer (NK) cell activity represents the only immunological protection against disease relapse for the first months after haplo-HSCT. Clinical studies have associated donor-recipient incompatibility for Human Leukocyte Antigen (HLA) ligands of Killer Immunoglobulin-like Receptors (KIR), with a marked anti-leukemic activity. Alloreactive donor NK cells carrying a single KIR whose ligand is missing in the recipient mediate a potent graft vs. leukemia effect, resulting in reduced incidence of relapse and increased Overall Survival (OS). These exciting results have recently been challenged by conflicting clinical and biological data from different groups. In the present study, we have characterized reconstitution of NK cells, in particular of alloreactive single-KIR+ NK cells, in 58 patients who received CD34+ selected haplo-HSCT for high-risk hematologic malignancies. One month after haplo-HSCT CD56bright/CD56dim NK cell subsets were subverted in their proportions and phenotypic features, accounting for enrichment in maturation intermediates. We show that CD25 and CD117 deregulation by CD56bright, and NKG2A and CD62L by CD56dim, are intrinsic to NK cell physiologic differentiation and support a sequential CD56bright-to-CD56dim NK cell maturation. Consistently, the in vitro functional potential of these maturation intermediates against leukemic blasts was heavily impaired, both in terms of cytotoxicity and of cytokine release. Full mature receptor repertoire reconstitution took at least three months. Alloreactive single-KIR+ NK cells had highly variable frequency ranging from less than 1% to more than 30% of NK cells circulating at 90–120 days after transplantation, independently from predicted NK alloreactivity. Importantly, out of three patients with predicted NK alloreactivity, none had a relative expansion of alloreactive single-KIR+ cells, accounting for less than 1% of circulating NK cells in two of them. As demonstrated by flow cytometric analysis of NK cell CD107a mobilization in response to the HLA class I negative target 721.221, single-KIR+ NK cells at three months after haplo-HSCT showed a not yet fully developed functional reactivity, which was recovered to donor-levels only at later time-points. In line with these observations, clinical outcome of haplo-HSCT was not affected in any way by the presence of donor NK alloreactivity. The incidence of relapse was virtually identical in patients transplanted from alloreactive or non-alloreactive donors. Taken together, our data shed new light onto the kinetics of NK cell differentiation in vivo and suggest that NK alloreactivity could be best exploited by the use of mature donor single-KIR+ selected alloreactive NK cells.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 210-210 ◽  
Author(s):  
Chen Xilin ◽  
Jianfeng Han ◽  
Chu Jianhong ◽  
Walter Meisen ◽  
Zhang Jianying ◽  
...  

Abstract Natural killer (NK) cells are innate lymphocytes that can rapidly eradicate tumor cells, especially those lacking MHC Class I molecules. NK cells can also rapidly eradicate herpes virus-infected cells. We designed an oncolytic herpes virus (oHSV) to selectively infect, replicate within, and lyse glioblastoma (GBM), a devastating brain tumor with a median survival of only 15 months following diagnosis. We have shown that the rapid influx of NK cells limits oHSV efficacy in GBM as they impede oHSV replication and spread [Alvarez-Breckenridge et al., Nat Med, 2012, 18(12):1827-34]. In the current study, we developed NK cell-based novel GBM therapies by decreasing the brain influx of NK cells to enhance the efficacy of oHSV, while arming NK cells in the brain with a chimeric antigen receptor (CAR) that targets both the wild-type EGFR and its mutant form EGFRvIII, two GBM tumor-associated antigens. We then investigated the synergistic effects between EGFR-CAR NK cells and oHSV. Transforming growth factor (TGF)-β is a potent immunosuppressive cytokine of NK cells [Yu et al, Immunity, 2006, 24(5):575-90]. We first determined if oHSV efficacy for treatment of GBM would be augmented by inhibiting anti-oHSV activity of NK cells with TGF-β pre-treatment. In vitro, NK cells pre-treated with TGF-β displayed less cytolytic capacity against oHSV-infected GBM cell lines and patient-derived GBM stem-like cells. In viral replication assays, co-culturing oHSV-infected GBM cells with NK cells pre-treated with TGF-β significantly increased virus titers. In an immunocompetent syngeneic GBM mouse model,administration of TGF-β to GBM-bearing mice prior to oHSV injection significantly inhibited intracranial infiltration and activation of NK cells (P < 0.05). In orthotopic human GBM xenograft mouse models and in syngeneic GBM mouse models, TGF-β treatment in vivo prior to oHSV therapy resulted in inhibition of NK cell infiltration, suppression of tumor growth and significantly prolonged survival of GBM-bearing mice (P < 0.05). Furthermore, depletion of NK cells incompletely blocked the positive effects of in vivo treatment of GBM with TGF-β on survival, suggesting that TGF-β may also directly act on other innate immune cells such as macrophages/microglia. These data demonstrate a single dose of TGF-β prior to oHSV administration enhances anti-tumor efficacy for GBM at least in part through the transient inhibition of the innate immune responses to oHSV infection. We next investigated whether NK cell activity could be enhanced to more directly target brain tumors while sparing eradication of oHSV. We therefore infected both human NK-92 cells and primary human NK cells to express the second generation CAR targeting both EGFR and EGFRvIII that we designed. Further, we asked if the treatment with EGFR-CAR NK cells plus oHSV could create a therapeutic synergy for the treatment to brain tumors. In vitro, compared with mock-transduced CAR-NK-cells, EGFR-CAR NK cells exhibited significantly higher cytotoxicity and IFN-γ production when co-cultured with tumor cells, for both NK-92 and primary NK cells (P < 0.01). Further, significantly higher cytolytic activity against tumor cells was obtained when CAR NK cells were combined with oHSV-1 infection of tumor cells, compared to either of the monotherapies alone (P < 0.05). In mice, to avoid oHSV clearance by the EGFR-CAR NK cells following the inoculation of the mouse with tumor cells, we administered these two agents sequentially; administering EGFR-CAR NK cells directly into the tumor first as a single injection of 2 × 106 cells, followed by intracranial infection with 2 × 105 plaque-forming units oHSV five days later, presumably after EGFR-CAR NK survival has diminished. Compared to vehicle controls, intracranial administration of either EGFR-CAR NK cells or oHSV blunted tumor growth. However, the combination of EGFR-CAR NK cells followed by oHSV infection resulted in significantly more efficient killing of tumor cells (P < 0.05) and significantly longer survival for tumor-bearing mice when compared to either monotherapy alone. Collectively, our studies demonstrate that in animal tumor models, we can combine novel NK cell and oHSV therapies to significantly improve survival. Disclosures No relevant conflicts of interest to declare.


1993 ◽  
Vol 74 (3) ◽  
pp. 1100-1106 ◽  
Author(s):  
M. Klokker ◽  
A. Kharazmi ◽  
H. Galbo ◽  
I. Bygbjerg ◽  
B. K. Pedersen

We have investigated the effects of short-term hypoxia in vivo on the human cellular immune system. Seven young healthy volunteers were placed in a decompression chamber (380 Torr) for 20 min with or without supplemental O2. The leukocyte concentration increased during hypobaric conditions because of an increased concentration of lymphocytes. The absolute and relative concentration of CD16+ natural killer (NK) cells increased markedly during hypoxia and returned to pretest values after 2 h of recovery. The NK cell activity of blood mononuclear cells (BMNC, %lysis/fixed no. of BMNC) boosted with interferon-alpha, interleukin-2 (IL-2), and indomethacin rose in parallel with unboosted NK cell activity during hypoxia. The percentage of CD4+ and CD8+ cells declined during hypoxia, whereas the absolute concentration of both CD8+ cells and CD14+ monocytes increased. Although the BMNC composition varied, the proliferative responses of BMNC after stimulation with phytohemagglutinin, purified derivative of tuberculin, and IL-2 did not change significantly. The in vitro production of interleukin-1 beta and IL-2 in supernatants obtained after stimulation of BMNC with phytohemagglutinin or lipopolysaccharide was not affected. The chemiluminescence response of neutrocytes increased 2 h after hypoxia. It was concluded that acute hypoxia induced marked alterations in the immune system and that the NK cells are especially sensitive to the hypoxic stimulus.


1981 ◽  
Vol 154 (2) ◽  
pp. 306-317 ◽  
Author(s):  
J A Lust ◽  
V Kumar ◽  
R C Burton ◽  
S P Bartlett ◽  
M Bennett

Mice were treated with the bone-seeking isotope, 89Sr, cyclophosphamide, and short-term lethal irradiation in vivo, and murine spleen cells are treated with anti-Nk-1.2 plus complement (C) in vitro. Fresh spleen cell suspensions from the above groups and from beige and neonatal mice were subsequently tested for natural killer (NK) cell activity against a panel of lymphoid and nonlymphoid tumor cell target. NK cell reactivities against YAC-1, MPC-11, and Cl.18 tumors were markedly and consistently reduced in (a) mice treated with 89Sr, (b) spleen cells treated with anti-Nk-1.2 plus C, and (c) C57BL/6 bg/bg mice. In contrast, NK activities against FLD-3 and WEHI-164.1 tumors were usually normal in mice treated with 89Sr, in beige mutant mice, and in spleen cells after treatment with anti-Nk-1.2 antibody and C. It appears, therefore, that two major groups of NK cells exist in fresh mouse spleen cells suspensions. NK-A cells are marrow dependent, Nk antigen positive, and deficient in beige mice; these lyse YAC-1, MPC-11, and Cl.18 tumors. NK-B cells, which are responsible for the lysis of WEHI-164.1 and FLD-3, are Nk antigen negative, marrow independent, and unaffected by the bg/bg mutation. Other features of NK-B cells, suggest that these NK cells, although they share the characteristics mentioned above, differ among themselves especially with respect to age of maturation and susceptibility to cyclophosphamide and total body irradiation. The NK-B group may therefore induce subsets that remain to be defined.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A670-A670
Author(s):  
Elisa Toffoli ◽  
Abdolkarim Sheikhi ◽  
Roeland Lameris ◽  
Lisa King ◽  
Jurriaan Tuynman ◽  
...  

BackgroundThe ability to kill tumor cells with an acceptable toxicity profile, makes Natural Killer (NK) cells promising assets for cancer therapy. However, strategies to enhance the preferential accumulation and activation of NK cells in the tumor microenvironment would likely increase the efficacy of NK cell-based therapies.MethodsIn this study, we show a novel bispecific nanobody-based construct (biVHH) targeting both CD16A (low-affinity Fc receptor: FcRγIIIA) on NK cells and EGFR on tumors of epithelial origins.ResultsHigher levels of NK cell activity and subsequent tumor cell lysis were found in vitro in the presence of the biVHH and were dependent on the expression of both CD16A and EGFR while they were independent of the KRAS mutational status of the tumor. Increased NK cell activity was found in NK cells derived from colorectal cancer (CRC) patients when co-cultured with the biVHH and EGFR expressing tumor cells. Finally, higher levels of cytotoxicity were found against patient-derived metastatic CRC cells in the presence of the biVHH and autologous peripheral blood mononuclear cells or allogeneic NK cells.ConclusionsBased on our results, the bispecific CD16A and EGFR targeting VHH construct could be a useful tool in combination with various NK cell-based therapies.


2015 ◽  
Vol 2015 ◽  
pp. 1-11 ◽  
Author(s):  
Kazuyoshi Takeda ◽  
Ko Okumura

Panax ginsengextracts are used in traditional herbal medicines, particularly in eastern Asia, but their effect on natural killer (NK) cell activity is not completely understood. This study aimed to examine the effects ofP. ginsengextracts on the cytotoxic activity of NK cells. We orally administeredP. ginsengextracts or ginsenosides to wild-type (WT) C57BL/6 (B6) and BALB/c mice and to B6 mice deficient in either recombination activating gene 2 (RAG-2) or interferon-γ(IFN-γ). We then tested the cytotoxic activity of NK cells (of spleen and liver mononuclear cells) against NK-sensitive YAC-1 cells. Oral administration ofP. ginsengaqueous extract augmented the cytotoxicity of NK cells in WT B6 and BALB/c mice and in RAG-2-deficient B6 mice, but not in IFN-γ-deficient B6 mice. This effect was only observed with the aqueous extract ofP. ginseng. Interestingly, the ginsenosides Rb1 and Rg1 did not augment NK cell cytotoxicity. These results demonstrated that the aqueousP. ginsengextract augmented NK cell activationin vivovia an IFN-γ-dependent pathway.


Sign in / Sign up

Export Citation Format

Share Document