Does the Selenium (SE) Level and Se-Dependent Enzyme Activity in Blood Plasma Correlate with Human Lymphocyte Subpopulations and Function?

1992 ◽  
Vol 5 (1) ◽  
pp. 13-21 ◽  
Author(s):  
Z. Baj ◽  
K. Zeman ◽  
Ewa Majewska ◽  
W. Wasowicz ◽  
Maria Sklodowska ◽  
...  

Recent literature data on the effects of Se on subpopulations of T lymphocytes, on autologous mixed lymphocyte reaction (AMLR) and on natural killer (NK) cell cytotoxicity are limited or poorly defined. In healthy volunteers we have estimated se levels, glutathione peroxidase (GSH-Px) activity and lipid peroxide levels in human plasma and simultaneously, the subpopulations of T lymphocytes, proliferation in AMLR, and activity of NK cells. We found a significantly positive correlation between the selenium level and GSH-Px activity. The proliferative response in AMLR significantly correlated with plasma selenium levels but not with GSH-Px activity. NK cytotoxicity, subpopulations of T lymphocytes, and lipid peroxide levels did not correlate with both selenium concentration and GSH-Px activity. We suppose that the effect of Se on the proliferation of suppressor T lymphocytes (Ts) in AMLR is not mediated through GSH-Px activity and fluctuations of Se concentration within a physiological range in healthy persons do not affect NK cytotoxicity.

Blood ◽  
2007 ◽  
Vol 109 (9) ◽  
pp. 3767-3775 ◽  
Author(s):  
Laura Chiossone ◽  
Chiara Vitale ◽  
Francesca Cottalasso ◽  
Sara Moretti ◽  
Bruno Azzarone ◽  
...  

Abstract Steroids have been shown to inhibit the function of fresh or IL-2–activated natural killer (NK) cells. Since IL-15 plays a key role in NK-cell development and function, we comparatively analyzed the effects of methylprednisolone on IL-2– or IL-15–cultured NK cells. Methylprednisolone inhibited the surface expression of the major activating receptors NKp30 and NKp44 in both conditions, whereas NK-cell proliferation and survival were sharply impaired only in IL-2–cultured NK cells. Accordingly, methylprednisolone inhibited Tyr phosphorylation of STAT1, STAT3, and STAT5 in IL-2–cultured NK cells but only marginally in IL-15–cultured NK cells, whereas JAK3 was inhibited under both conditions. Also, the NK cytotoxicity was similarly impaired in IL-2– or IL-15–cultured NK cells. This effect strictly correlated with the inhibition of ERK1/2 Tyr phosphorylation, perforin release, and cytotoxicity in a redirected killing assay against the FcRγ+ P815 target cells upon cross-linking of NKp46, NKG2D, or 2B4 receptors. In contrast, in the case of CD16, inhibition of ERK1/2 Tyr phosphorylation, perforin release, and cytotoxicity were not impaired. Our study suggests a different ability of IL-15–cultured NK cells to survive to steroid treatment, thus offering interesting clues for a correct NK-cell cytokine conditioning in adoptive immunotherapy.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 8571-8571
Author(s):  
Ines Esteves Domingues Pires Da Silva ◽  
Sonia Jimenez-Baranda ◽  
Anne Gallois ◽  
Vijay Kuchroo ◽  
Iman Osman ◽  
...  

8571 Background: The concept of CD8+ T cell exhaustion in the context of metastatic cancer has been reinforced by the recent success of immunotherapies targeting the exhaustion markers CTLA-4 and PD-1 in advanced melanoma. T-cell immunoglobulin 3 (Tim-3), another exhaustion marker, is also expressed in natural killer (NK) cells, however its role is still unknown. Recent reports have shown that NK cells, innate immune cells that eliminate tumors through cytotoxicity and IFN-g production, are functionally impaired in advanced melanoma patients, although no receptor has been linked with that phenotype so far. In this study, we characterize the role of Tim-3 in NK cells, particularly in the presence of its natural ligand, Galectin-9 (Gal-9), that is known to be expressed/secreted by some tumor cells including melanoma. Methods: We compared 20 advanced melanoma donors NK cells with 40 healthy donors NK cells as it relates to Tim-3 expression (by flow cytometry) and function (cytotoxicity, IFN-γ production and proliferation). NK cells cytotoxicity was measured by lamp-1 expression, and two different target cells were used: i) K562 cells (Gal-9-) and ii) Gmel Gal-9+ and Gmel Gal-9- sorted melanoma cells. Proliferation was quantified by CFSE after 6 days in the presence of rhIL-2. Recombinant rhGal9 effect was tested in cytotoxicity and IFN-γ production. Results: Melanoma patients NK cells express higher levels of Tim-3 compared to healthy donors NK cells (p<0.05). Melanoma patients NK cells have a defect in cytotoxicity, proliferation and IFN-γ production. Tim-3 expression by itself (without engagement of specific ligands) does not negatively affect NK cell functions (p<0.05). However, when rhGal9 is added to the system, a decrease in NK cell cytotoxicity and IFN-γ production (p<0.05) was observed. Finally, the expression of Gal-9 by the target cells induces a defect in NK cell cytotoxicity (Gmel Gal-9+ vs Gmel Gal-9-). Conclusions: These data suggest that advanced melanoma patients NK cells are exhausted, although it still remains unclear if Tim-3 is involved in this phenotype. In addition,the expression/secretion of Galectin-9, immunosuppressive for NK cells, may be a possible mechanism for tumors to evade immune surveillance.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2465-2465
Author(s):  
Masashi Okamoto ◽  
Tohru Inaba ◽  
Sonoko Nakano ◽  
Kyoko Namura ◽  
Noriko Yamada ◽  
...  

Abstract Introduction: Major histocompatibility complex class-I related chain (MIC) is a ligand for NKG2D, one of the activating NK receptor. It is expressed on various cancerous cells, and mediates NK cytolysis. Myeloma cells may be susceptible to immune therapy because interferon (IFN)-alpha or thalidomide are effective for a part of myeloma patients. It is also reported that one myeloma cell line is susceptible to NK cytolysis, but another lines are not, and this difference is not depend on its KIR incompatibility. We therefore examined the expression of MIC on myeloma cells, and its role on NK cell cytotoxicity. Method: MIC expression was examined on two myeloma cell lines, U266 and RPMI8226, and fresh myeloma cells by flow cytometry (FCM) and reverse transcript polymerase chain reaction (RT-PCR). Modulation of MIC expression on these cell lines and fresh myeloma cells by the addition of various concentration of the drugs such as IFN-alpha, thalidomide, all-trans retinoic acid (ATRA), dexamethasone, and incadronate was examined by FCM and RT-PCR. The relationship between MIC expression and NK cytolysis was examine by 51Cr release assay. Result: MIC was highly expressed on these two cell lines. MIC mRNA was also detected by RT-PCR in both cell lines. IFN-alpha, thalidomide, ATRA, incadronate and dexamethasone are known to be effective for MM in vivo or in vitro, we therefore examined the effect of these drugs on the expression of MIC on two myeloma cell lines. The cell lines were treated with or without these drugs at various concentrations, and then subjected to FCM. MIC expression was not changed after treatment of thalidomide, incadronate and dexamethasone. But unexpectedly, it was down-regulated by IFN-alpha and ATRA dose-dependently on both cell lines. To examine whether MIC expression on myeloma cell lines is involved in their susceptibility to NK cells, NK cell cytotoxicity against these cell lines was analyzed by 51Cr releasing assay. A substantial cytolysis was detected in U266 cells, while RPMI8226 did not show any relevant cytolysis, though the cell line express high level of MIC. Furthermore NK cytotoxicity was not inhibited by the addition of anti-MIC antibody. Then we examined the effect of IFN-alpha on the NK cytotoxicity, because IFN-alpha down-regulated the expression of MIC on myeloma cell lines. However, NK cytotoxicity was also induced by IFN-alpha in both cell lines. To examine whether MIC is expressed on fresh myeloma cells and is involved in the effect of the various drugs against myeloma cells, fresh samples from 12 MM patients were examined on the expression of MIC using two-color flow cytometric analysis. MIC was expressed on myeloma cells in one of 12 patients examined. The treatment with IFN-alpha, thalidomide and ATRA did not up-regulated the expression of MIC in all samples. Conclusion: MIC is expressed on myeloma cell lines, but rarely expressed on fresh myelma cells. The reason of the discrepancy of MIC expression between fresh and cloned myeloma cells may be that myeloma cells are killed by host NK cells in vivo, provided the cells express MIC. In case myeloma cells express MIC in vivo, it may be that the molecule is not recognized by NKG2D on the NK cell.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2298-2298
Author(s):  
Miroslaw J. Szczepanski ◽  
Malgorzata Czystowska ◽  
Marta Szajnik ◽  
Ann Welsh ◽  
Kenneth A. Foon ◽  
...  

Abstract Natural killer (NK) cells lyse malignant cells without prior antigen-specific priming and play a critical role in the innate immune response. A balance of signals from activating and inhibiting receptors expressed on each NK cell controls its activity. The growth, differentiation and survival of NK cells have been found to be dependent on interleukin-15 (IL-15). Using multicolor flow cytometry we investigated the receptor repertoire and also measured the NK cell activity in twenty three patients with newly diagnosed acute myeloid leukemia (AML) prior to any treatment. Further, we investigated the ex-vivo effect of IL-15 on the NK cell repertoire and NK cell cytotoxicity. The percentage of circulating NK cells was lower (p&lt;0.0001) in the AML patients (6%± 0.7, range 1–17%) compared to the NK cells of healthy donors (12%± 1, range 9–17%). The expression of the activating natural cytotoxicity (NCR) receptors NKp30 and NKp46 and the C-type lectin receptors NKG2D and NKG2C was significantly decreased in the AML patients compared to the NK cells of healthy donors: NKp30 24 vs 51% p&lt;0.0001, NKp46 32 vs 73% p&lt;0.0001, NKG2D 43 vs 83% p&lt;0.0001, NKG2C 17 vs 28% p&lt;0.03. In addition, the receptor expression (mean fluorescence intensity, MFI) was also significantly lower in AML patients compared to healthy donors. No significant differences in the expression of the NCR NKp44 and the NK- cell inhibitory receptors were observed. Furthermore, the NK cytotoxicity in the AML patients at diagnosis was significantly lower (p&lt;0.0003) compared to the NK cytotoxicity of healthy donors (4 vs 75 LU). When NK cells obtained from AML patients were cultured with IL-15, significant increases in the expression of the NK- cell activating receptors (Table 1) were observed. The upregulation of the activating receptors was associated with a concomitant significant increase (p&lt;0.001) of the NK cell cytotoxicity (4 vs 70 LU). The data suggest that IL-15, a homeostatic NK cell cytokine, can upregulate the expression of activating receptors and concomitantly increase the NK lytic activity. The use of IL-15 as a platform for NK- based therapies for AML patients should be considered in the future. Table 1. The effect of IL-15 on the receptor expression in AML patients NK cell Activating Receptors NK cells in AML pts at diagnosis NK cells in AML pts after IL-15 stimulation p value % positive, MFI % positive, MFI NKp30 24, 2.6 84, 6.0 0.001 NKp44 4, 2.4 71, 6.7 0.001 NKp46 32, 2.6 83, 7 0.002 NKG2C 17, 2.2 58, 6.9 0.005 NKG2D 43, 2.5 87, 7.8 0.01


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 553-553
Author(s):  
Chitra Hosing ◽  
Zachary Braunstein ◽  
Alaa M Ali ◽  
Benigno C. Valdez ◽  
Borje S. Andersson ◽  
...  

Abstract Background: Allo-SCT is the only curative option for patients with high risk and relapsed/refractory T-cell malignancies. Even among allo-SCT recipients, survival is less than 50% and relapse rates are 55-60%. We developed a clinical trial to decrease relapse after allo-SCT for these patients using romidepsin (rom), a histone deacetylase inhibitor approved for the treatment of relapsed T-cell lymphomas. Based on pre-clinical data demonstrating enhanced and synergistic cell killing with the addition of rom to busulfan (Bu) and fludarabine (Flu) in malignant T-cells, we created a novel transplant regimen (BuFluRom). We hypothesized this regimen, coupled with maintenance rom (m-rom), would enhance malignant T-cell killing, eradicate MRD at transplant, decrease relapse, and stimulate the GVL effect by stimulating NK-cells. Here we present results of this clinical trial, with correlative data evaluating NK-cytotoxicity. This is the first trial designed specifically to treat T-cell malignancies with allo-SCT. (NCT02512497) Methods: This is a phase I/II clinical trial. Eligible patients had: a diagnosis of T-cell leukemia (including T-acute lymphoblastic leukemia) or T-cell lymphoma (cutaneous or peripheral) in at least a partial remission requiring an allo-SCT, &lt;70 years of age, with a matched sibling/unrelated donor. The primary objective was to determine the recommended phase 2 dose (RP2D) of rom from 3 dose levels (1, 2, 3 mg/m2) when combined with BuFlu (AUC 20000 or 16000, Figure). Patients received standard tacrolimus/methotrexate GVHD prophylaxis with ATG for MUDs. Once RP2D was determined, an expansion cohort of up to 30 patients (total) was included. M-rom was initiated between day +28 and +100 for 1 year (2 years max). The effect of rom on NK-cell cytotoxicity was assessed on samples taken pre-transplant, and 1, 3, 6, 12 months post allo-SCT. NK cytotoxicity was assessed by isolating mononuclear cells from patient samples and targeting them against K562 and T-cell lymphoma targets using the calcein-AM assay. Fine-Gray models were used to estimate PFS, OS, and cumulative incidence, and compare survival curves across groups. Results: 21 patients have been enrolled (Table). One DLT was observed (VOD), at dose level 2, and the RP2D of rom in conditioning was determined to be 2 mg/m2. With a median follow-up time of 10.1 months, the median OS has not been reached (3.3-NR months), with a 1 and 3-year OS probability of 62.8% & 55.8%. The median PFS is 28.2 months (3.8-28.1), with 1 and 3 year PFS of 57% & 30.4%. Cumulative incidence (CI) of NRM at day 100 and 1 year were 14.8% and 20%. CI of grade II-IV aGHVD and extensive cGVHD were 47.6% and 18.5%. The CI of relapse (CIR) was 22.8% at 1 year (95% CI 6.6-44.9%). There was no difference between PFS among patients with MRD versus those without MRD prior to transplant (p=0.96), and no difference in 1-year CIR (p=0.9). PFS and CIR at 1 year was substantially better in the lymphoma than leukemia patients (85.7% vs 44%, p=0.049), and (0% vs 32.1%, p=0.05). No patients with PTCL relapsed, and 3/5 patients with T-PLL are alive, disease free. 13/21 (62%) of patients received m-rom with a median number of 10 cycles (range 1-41). (Table) 7 patients experienced grade 3/4 adverse events (AE), though no patients discontinued m-rom due to toxicity. NK-cytotoxicity was higher at each time point in patients who received m-rom compared to those who did not, though there were insufficient patients to reach statistical significance. When NK-cytotoxicity was assessed between the two groups after starting maintenance, NK-cytotoxicity in the m-rom group was significantly higher than in those without m-rom (p=0.05) (Figure). Conclusions: BuFluRom with m-rom is effective at decreasing relapse in patients with T-cell malignancies, with 1-year CI relapse below expected relapse rates for this set of diseases. Toxicities were similar to standard BuFlu alone and the RP2D of rom in conditioning was established at 2 m g/m2. Intriguingly, BuFluRom mitigated the poor outcomes of patients with MRD prior to transplant. Further, early data suggests m-rom enhances NK-cell cytotoxicity post allo-SCT, potentially augmenting the GVL effect and accounting for decreased relapse rates. Long-term follow-up is needed to evaluate these results, but these results suggest the BuFluRom regimen with m-rom could become a new option for patients receiving allo-SCT for T-cell malignancies to mitigate relapse. Figure 1 Figure 1. Disclosures Hosing: Nkarta Therapeutics: Membership on an entity's Board of Directors or advisory committees. Popat: Bayer: Research Funding; Abbvie: Research Funding; Novartis: Research Funding; Incyte: Research Funding. Vasu: Boehringer Ingelheim: Other: Travel support; Seattle Genetics: Other: travel support; Kiadis, Inc.: Research Funding; Omeros, Inc.: Membership on an entity's Board of Directors or advisory committees. de Lima: Miltenyi Biotec: Research Funding; Incyte: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees. William: Dova Pharmaceuticals: Research Funding; Incyte: Research Funding; Kyowa Kirin: Consultancy; Merck: Research Funding; Guidepoint Global: Consultancy. Lee: Kiadis Pharma: Divested equity in a private or publicly-traded company in the past 24 months, Honoraria, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding; Courier Therapeutics: Current holder of individual stocks in a privately-held company. Brammer: Kymera Therapeutics: Consultancy; Celgene: Research Funding; Seattle Genetics: Speakers Bureau.


1982 ◽  
Vol 155 (6) ◽  
pp. 1610-1622 ◽  
Author(s):  
D Fradelizi ◽  
I Gresser

The effect of human interferon alpha on the differentiation of functional populations of lymphocytes during the human allogeneic response in vitro was studied. Interferon alpha inhibited the generation of allospecific suppressor T lymphocytes that normally develop from lymphocytes primed in vitro against allogeneic cells. This effect was not the result of the destruction by interferon of precursor suppressor cells but rather to inhibition of their differentiation into active suppressor T lymphocytes. This inhibition was reversible and could be overcome by repeated allogeneic stimulation even in the presence of interferon. Inhibition of the generation of allospecific suppressor lymphocytes by interferon might play an important role in the allogeneic response. Interferon inhibited the proliferation of lymphocytes after allogeneic stimulation in a primary mixed lymphocyte reaction but enhanced their cytotoxicity. Despite the inhibitory effect in the primary mixed lymphocyte reaction, the specific secondary proliferative response of lymphocytes primed against a single HLA-DR antigen was only slightly affected by interferon. On the other hand, the nonspecific secondary proliferative response of lymphocytes primed in the presence of interferon was significantly reduced, indicating that interferon might decrease the recruitment of nonspecific "irrelevant" clones of responding cells during the sensitization period.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 207-207
Author(s):  
Steven Scoville ◽  
Bethany Mundy-Bosse ◽  
Michael Zhang ◽  
Li Chen ◽  
Ryan Sanderson ◽  
...  

Abstract Natural killer (NK) cells are innate effector cells that can spontaneously recognize and kill cancer cells. While important in inducing long-term disease free survival (DFS) in the setting of killer immunoglobulin-like receptor (KIR) mismatch for acute myeloid leukemia (AML), harnessing NK cells to kill autologous or self AML blasts to extend DFS has had no success. In this study, we uncover one potential mechanism by which AML blasts can evade NK cell cytotoxicity. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor that has now been shown to be expressed in immature human NK cells. Furthermore, activation of AHR in these immature cells suppresses human NK cell maturation and function. We and others have shown that human AML blasts secrete ligands that can activate AHR, leading us to hypothesize that AML may evade autologous NK cell cytotoxicity in part by inhibiting NK cell maturation. Expression of microRNA (miR)-29b has previously been shown to suppress the expression of transcription factors Tbx21 (TBET) and Eomesodermin (EOMES), both of which are critical for terminal NK cell differentiation and function. Here we show that AHR is able to directly regulate the expression of miR-29b and thus may serve as the link between AML and immune evasion of NK cells. We first identified putative AHR binding sites within the proximal promoter of miR-29b, suggesting that AHR may directly regulate miR-29b expression. To test this, we transfected the AHR responsive HepG2 human cell line with a miR-29b promoter driven luciferase reporter and measured luciferase activity after treatment with the known AHR agonist, FICZ, compared to vehicle control. We discovered that cells treated with 6-formylindolol[3,2-b]carbazole (FICZ) had increased luciferase activity compared to cells treated with vehicle control (P<0.05). This effect was subsequently shown to be directly mediated by AHR, as mutation of the putative AHR binding site as well as siRNA targeting of AHR mRNA both significantly diminished the induced luciferase activity (P<0.01). We then established the importance of miR-29b in human NK cell development by transducing immature NK cells, characterized as Lin(-)CD117(+)CD94(-), with a miR-29b knockdown virus and testing for the ability of these cells to become mature [i.e., Lin(-)CD117(-)CD94(+)] NK cells, after two weeks in IL-15 and FICZ. Indeed, in early experiments, knockdown of miR-29b resulted in increased percentages of mature NK cells compared to cells transduced with control virus (16.8% compared to 3.6%, respectively), despite being cultured with an AHR agonist. Finally, utilizing a translational in vivo murine AML model developed by our laboratory that recapitulates human AML, we have found that AML blasts harvested from these leukemic mice release an AHR agonist (P < 0.01), similar to previous reports that have described AHR ligands being produced by human AML blasts. In addition,NK cells isolated from these mice have increased levels of miR-29b when leukemic, compared to NK cells from wild type control littermates, consistent with our hypothesis that AHR regulates miR-29b expression. Thus, we propose that AHR, when activated by AML-derived ligands, upregulates miR-29b in NK cells to ultimately suppress the primary regulators of NK cell maturation and function, resulting in immune evasion (See Figure). Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3464-3464
Author(s):  
Jooeun Bae ◽  
Constantine Mitsiades ◽  
Rao Prabhala Prabhala ◽  
Tai Yu-Tzu ◽  
Jeff Martinson ◽  
...  

Abstract Hsp90 inhibitor has shown promising anti-tumor activity through the destabilization and eventual degradation of Hsp90 client proteins critical for cell survival. In this study, we examined the in vitro effects of Hsp90 inhibitor on the phenotype and function of human T lymphocytes and NK cells. We observed no significant effects of Hsp90 inhibitor treatment on cell survivals. However, Hsp90 inhibitor treatment for 24 hours led to irreversible down-regulation of expression of critical T-cell surface antigens including CD3, CD4, CD8, CD28, CD154 (CD40L) and TCRab. Among the antigens evaluated, expression of CD4 antigen was most significantly downregulated (untrt vs. trt = 326 vs. 88 in Mean Fluorescence Intensity) following Hsp90 inhibitor treatment. Decreased CD3+ T lymphocytes proliferation (untrt vs. trt = 222839 cpm vs. 111102 cpm, 3[H]-thymidine incorporation) and reduced IFN-g secretion (untrt vs. trt = 77 vs. 48 pg/ml) was observed upon stimulation with allogeneic dendritic cells following 24 hrs treatments of T cells with Hsp90 inhibitor. Furthermore, CD3+ T-cell proliferation in response to mitogen stimulation, as measured by flow cytometry using CFSE was decreased following Hsp90 inhibitor treatment (untrt vs. trt = 41% vs. 3%, CFSE). Specifically, the CD4+CD28+ (untrt vs. trt = 32% vs. 1%) and CD8+CD28+ (untrt vs. trt = 27% vs. 17%) activated T-cell subpopulations displayed a significant decrease in proliferation in response to mitogen. Similarly, NK cells displayed decreased activation receptor expression including CD2, CD11a, CD94, NKp30, NKp44, NKp46, and KARp50.3 and reduced cytotoxic activity against multiple myeloma cells (untrt vs. trt = 49% vs. 11% against MM1S cells, 65% vs. 8% against ARP cells) following Hsp90 inhibitor treatment. These studies demonstrate that Hsp90 inhibitor treatment significantly affects phenotype and function of human T-lymphocytes as well as NK cells, and suggest the need to monitor immune functions in patients being treated with Hsp90 inhibitor in our future studies.


Sign in / Sign up

Export Citation Format

Share Document