scholarly journals Extracts ofCistanche deserticolaCan Antagonize Immunosenescence and Extend Life Span in Senescence-Accelerated Mouse Prone 8 (SAM-P8) Mice

2014 ◽  
Vol 2014 ◽  
pp. 1-14 ◽  
Author(s):  
Ke Zhang ◽  
Xu Ma ◽  
Wenjun He ◽  
Haixia Li ◽  
Shuyan Han ◽  
...  

The senescence accelerated mouse prone 8 substrain (SAM-P8), widely accepted as an animal model for studying aging and antiaging drugs, was used to examine the effects of dietary supplementation with extracts ofCistanche deserticola(ECD) which has been used extensively in traditional Chinese medicine because of its perceived ability to promote immune function in the elderly. Eight-month-old male SAM-P8 mice were treated with ECD by daily oral administrations for 4 weeks. The results showed that dietary supplementation of 150 mg/kg and 450 mg/kg of ECD could extend the life span measured by Kaplan-Meier survival analysis in dose-dependent manner. Dietary supplementation of SAM-P8 mice for 4 weeks with 100, 500, and 2500 mg/kg of ECD was shown to result in significant increases in both naive T and natural killer cells in blood and spleen cell populations. In contrast, peripheral memory T cells and proinflammatory cytokine, IL-6 in serum, were substantially decreased in the mice that ingested 100 and 500 mg/kg of ECD daily. Additionally, Sca-1 positive cells, the recognized progenitors of peripheral naive T cells, were restored in parallel. Our results provide clear experimental support for long standing clinical observational studies showing thatCistanche deserticolapossesses significant effects in extending life span and suggest this is achieved by antagonizing immunosenescence.

1992 ◽  
Vol 175 (1) ◽  
pp. 211-216 ◽  
Author(s):  
T G Yin ◽  
P Schendel ◽  
Y C Yang

The availability of large quantities of highly purified recombinant interleukin 11 (rhuIL-11) has allowed us to investigate the effects of rhuIL-11 on sheep red blood cell (SRBC)-specific antibody responses in the murine system. The results showed that rhuIL-11 was effective in enhancing the generation of mouse spleen SRBC-specific plaque-forming cells (PFC) in the in vitro cell culture system in a dose-dependent manner. These effects of rhuIL-11 were abrogated completely by the addition of anti-rhuIL-11 antibody, but not by the addition of preimmunized rabbit serum. Cell-depletion studies revealed that L3T4 (CD4)+ T cells, but not Lyt-2 (CD8)+ T cells, are required in the rhuIL-11-stimulated augmentation of SRBC-specific antibody responses. The effects of rhuIL-11 on the SRBC-specific antibody responses in vivo were also examined. RhuIL-11 administration to normal C3H/HeJ mice resulted in a dose-dependent increase in the number of spleen SRBC-specific PFC as well as serum SRBC-specific antibody titer in both the primary and secondary immune responses. In mice immunosuppressed by cyclophosphamide treatment, rhuIL-11 administration significantly augmented the number of spleen SRBC-specific PFC as well as serum SRBC-specific antibody titer when compared with the cyclophosphamide-treated mice without IL-11 treatment. These results demonstrated that IL-11 is a novel cytokine involved in modulating antigen-specific antibody responses in vitro as well as in vivo.


1995 ◽  
Vol 182 (6) ◽  
pp. 1785-1792 ◽  
Author(s):  
P Jeannin ◽  
Y Delneste ◽  
S Lecoanet-Henchoz ◽  
J F Gauchat ◽  
P Life ◽  
...  

N-Acetyl-L-cysteine (NAC) is an antioxidant precursor of intracellular glutathione (GSH), usually given in human as a mucolytic agent. In vitro, NAC and GSH have been shown to act on T cells by increasing interleukin (IL) 2 production, synthesis and turnover of IL-2 receptors, proliferation, cytotoxic properties, and resistance to apoptosis. We report here that NAC and GSH decrease in a dose-dependent manner human IL-4 production by stimulated peripheral blood T cells and by T helper (Th) 0- and Th2-like T cell clones. This effect was associated with a decrease in IL-4 messenger RNA transcription. In contrast, NAC and GSH had no effect on interferon gamma and increased IL-2 production and T cell proliferation. A functional consequence was the capacity of NAC and GSH to selectively decrease in a dose-dependent manner IL-4-induced immunoglobulin (Ig) E and IgG4 production by human peripheral blood mononuclear cells. Interestingly, NAC and GSH also acted directly on purified tonsillar B cells by decreasing the mature epsilon messenger RNA, hence decreasing IgE production. In contrast, IgA and IgM production were not affected. At the same time, B cell proliferation was increased in a dose-dependent manner. Not all antioxidants tested but only SH-bearing molecules mimicked these properties. Finally, when given orally to mice, NAC decreased both IgE and IgG1 antibody responses to ovalbumin. These results demonstrate that NAC, GSH, and other thiols may control the production of both the Th2-derived cytokine IL-4 and IL-4-induced Ig in vitro and in vivo.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2314-2314
Author(s):  
Jin Im ◽  
Amanda Cernosek ◽  
Anna Sergeeva ◽  
Jeffrey J. Molldrem ◽  
Lu Sijie

Abstract PR1 is a HLA-A2 restricted immune-dominant peptide derived from proteinase 3 and neutrophil elastase - leukemia associated self-antigens aberrantly over-expressed in myeloid leukemia. The persistent PR1/HLA-A2 specific cytotoxic T cell responses are associated with prolonged remission from myeloid leukemia, suggesting that PR1/HLA-A2 would be a potent target for T cell based immunotherapy. Previously, we have successfully developed a high affinity T cell receptor (TCR)-like antibody specific for the PR1/HLA-A2 complex (h8F4), and demonstrated that h8F4 mediated specific lyses of myeloid leukemia cells invitro via antibody dependent cell mediated cytotoxicity (ADCC), and significantly reduced leukemic blasts invivo in murine xenograft model of treatment refractory acute myeloid leukemia. We hypothesize that the therapeutic efficacy of h8F4 would be enhanced by combining T cell mediated cytotoxicity through a novel antibody engineering technology, called, Bi-Specific T cell Engager (BiTE) specific for T cells and tumor associated antigens. The BiTE molecules redirect effector function of polyclonal T cells against the tumor leading to lyses of tumor cells, and early clinical data show promising therapeutic efficacy of this approach. In the current study, we constructed Bi-Specific T cell Engaging antibody: h8F4-BiTE, a single chain Fragment of variable domain (scFv) of h8F4 conjugated with scFv of anti-CD3 antibody – OKT3, and produced h8F4-BiTE proteins using eukaryotic expression system. The h8F4-BiTE proteins retained the similar binding specificity to PR1/HLA-A2 as compared to the parent antibody (h8F4), demonstrated by flowcytometry based binding assays with a series of alanine substituted PR1 peptide/HLA-A2 complexes on T2 cells. The 8F4-BiTE bound to native and endogenous PR1/HLA-A2 complexes presented by HLA-A2 positive or transduced myeloid cell lines such as K562, THP-1, and HL-60. In addition, h8F4-BiTE showed comparable binding affinity to CD3 moiety of T cells. Lastly, h8F4-BiTE proteins activated T cells in a PR1/HLA-A2 specific and dose dependent manner, shown by increased expression of activation markers such as CD69 when co-cultured with PR1-pulsed T2 target cells in various concentrations of h8F4-BiTE proteins. Importantly, the PR1/HLA-A2 specific activation of T cells by h8F4-BiTE proteins was equally efficient among all subsets of T cells regardless of their phenotypes, e.g., CD4 vs CD8, and naïve vs memory, suggesting that h8F4-BiTE would be a highly potent inducer of cytotoxic T cells against PR1/HLA-A2 positive myeloid leukemia. Moreover, we generated multiple h8F4-BiTE variants with specific mutations in frameworks and/or complement determining regions (CDRs), and compared binding affinity and function to wildtype h8F4-BiTE. The h8F4-BiTE variants indeed improved specificity to PR1/HLA-A2 complex by complete elimination of residual non-specific binding to HLA-A2, and increased the binding affinity to CD3 by 10-folds resulting in dramatic 50 fold increase in potency to activate T cells. In conclusion, we have developed a novel immunetherapeutics: h8F4-BiTE targeting PR1/HLA-A2 myeloid leukemia antigen. We demonstrated high binding affinity and specificity of h8F4-BiTE to both PR1/HLA-A2 on myeloid leukemia cells and CD3 on T cells and in vitro activation of T cells in the presence of PR1/HLA-A2 expressing target cells in a dose dependent manner. Finally, we improved antigen specificity and functional activity of h8F4-BiTE via selective site-directed mutagenesis. Further preclinical evaluation of therapeutic efficacy is thus warranted using animal in vivo animal treatment model with NOD/SCID/IL2Rγ −/− mice xenografted with human myeloid leukemia cells. This unique TCR-like therapeutic agent adds an additional mechanism of action to PR1/HLA-A2 specific antibody (h8F4) and promises to enhance T cell function against myeloid leukemia. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4983-4983
Author(s):  
Hideaki Maeba ◽  
Rie Kuroda ◽  
Masaki Fukuda ◽  
Toshihiro Fujiki ◽  
Shintaro Mase ◽  
...  

Abstract Intravenous immunoglobulin (IVIg) has been used after allogeneic stem cell transplants (HSCT) mainly to prevent infections. In addition, some studies have also demonstrated that IVIg reduced GVHD and GVHD-related mortality, but others failed to show these effects. To answer whether IVIg could reduce GVHD and/or has preventive effects, as a first step, we compared the mixed lymphocyte reactions (MLRs) between a variety of responder and stimulator combinations (Balb/c and C57BL/6) with/without immunoglobulin (Ig). We showed that the MLR was strongly inhibited when high concentration of Ig was added to the cocluture (Stimulator (S): bone marrow-derived dendritic cells (BM-DCs) obtained from C57BL/6 using GM-CSF, Responder (R): splenocytes obtained from Balb/c). Interestingly, dose dependent inhibition was not observed only when high concentration of Ig could suppresses allo-reaction. The next concern was whether Ig could suppresse MLR via modulation of DCs or T-cell directly, or both. To test this, splenocytes were stimulated with phytohemagglutinin (PHA) in the presence of various Ig concentrations. We observed that Ig suppressed PHA-induced T-cell proliferation in a dose dependent manner. To eliminate the possibility that Ig might have direct cytotoxicity against lymphocytes and consequently allo-immune reaction seemed to be inhibited, we performed dead cell analysis by flowcytometry using 7-AAD and annexin V on T-cells in the various concentrations of Ig. Even in high concentration of Ig, the frequency of apoptotic or dead cells were observed as same levels as in low concentration or absence of Ig, suggesting that Ig inhibited T-lymphocyte activation directly. Next concern was when Ig allowed tolerogenic function to T-cells and/or DCs. That is, only pretreatment of Ig before MLR was enough to suppress allo-reaction or persistent high concentration of Ig exposure was needed during MLR. To test this, splenocytes and BM-DCs were cultured with Ig for 24 hours before MLRs, and then washed twice and we compared the MLRs. Ig pretreatment to either splenocytes or BM-DCs or both did not show any suppression of MLRs as shown in the figure below, indicating that persistent high levels of Ig during interaction were needed to induce maximum inhibiting effects. In conclusion, we have clearly demonstrated that Ig has a strong capacity to suppress allo-immune reaction. Repetitive high dose IVIg treatment would be a promising approach to reduce or prevent sever acute GVHD, only if serum Ig concentration could be kept in high level. In addition, to apply this in clinical settings, disadvantage of high dose IVIg should be further investigated. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Cells ◽  
2021 ◽  
Vol 10 (10) ◽  
pp. 2718
Author(s):  
Mandy Mun Yee Kwong ◽  
Jee Whu Lee ◽  
Mohammed Razip Samian ◽  
Habibah A. Wahab ◽  
Nobumoto Watanabe ◽  
...  

Certain plant extracts (PEs) contain bioactive compounds that have antioxidant and lifespan-extending activities on organisms. These PEs play different roles in cellular processes, such as enhancing stress resistance and modulating longevity-defined signaling pathways that contribute to longevity. Here, we report the discovery of PEs that extended chronological life span (CLS) in budding yeast from a screen of 222 PEs. We identified two PEs, the leaf extracts of Manihot esculenta and Wodyetia bifurcata that extended CLS in a dose-dependent manner. The CLS-extending PEs also conferred oxidative stress tolerance, suggesting that these PEs might extend yeast CLS through the upregulation of stress response pathways.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2364-2364
Author(s):  
Anwar A. Sayed ◽  
Amna Malik ◽  
Grace Ayoola ◽  
Elisa Lucchini ◽  
Sasfia Candrianita ◽  
...  

Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by a skewed proinflammatory T cell profile. Thrombopoietin-receptor agonists (TPO-RA) have largely replaced immunosuppressants in the management of this disorder, with some patients achieving remission after a period of treatment with TPO-RA. The potential immune modulatory role of TPO-RA has not been fully investigated. The two current TPO-RA licensed for use in ITP; Eltrombopag (Elt) and Romiplostim (Romi) act on different parts of the TPO-R and have similar response rates. However, patients can respond to one agent but not the other. Elt has been described to have a strong iron chelating effect, and hence we propose that it may have an additive immunomodulatory effect on the T cells, absent in Romi. We determined the immunomodulatory effect of Elt by assessing the proliferation and functionality of T-cell lines and primary T-cells. T cell proliferation was assessed using both CFSE proliferation assay and MTT cell viability assay. T cell phenotype and functionality were assessed by multicolor surface and intracellular flow cytometric staining. Cells were co-cultured with Elt and Romi in vitro and ex vivo with both Jurkat and DG75 cells lines as well as primary cells, respectively. Deferoxamine (DFX) was used as a positive control for iron-chelation, and human TPO was used as a positive control for TPO-RA. All treatment doses were based on their calculated therapeutic serum levels. Mann Whitney U and Kruskal-Wallis H statistical tests were applied where applicable, and a P value of less than 0.05 were considered significant. Elt significantly decreased Jurkat T cells proliferation in a dose-dependent manner compared to no treatment and Romi. DFX, an iron chelator, also decreased Jurkat T cell proliferation to comparable levels of Elt. Interestingly, this anti-proliferative effect of Elt was only observed on Jurkat T cells, but not DG75 B cell line. Ex vivo CFSE proliferation assay was performed on primary CD4 and CD8 T cells assessing the antiproliferative effect of Elt. Elt significantly reduced proliferation compared to no treatment. DFX exhibited a similar antiproliferative effect on primary T cells, however, less potent compared to Elt. Neither Romi nor TPO affected the proliferation of Jurkat cells, DG75 cells or primary T cells. The functionality of CD4 and CD8 T cells was assessed based on the capacity of T cells to produce intracellular TNFα, IFNγ and Granzyme B. Elt significantly reduced the percentages of TNFα+/IFNγ+ CD4+ and CD8+ T cells in a dose-dependent manner. This reduction was also observed, albeit to a lesser extent, when T cells were treated with DFX. Furthermore, Granzyme B expression in CD8+ T cells was significantly reduced when cells were treated Elt, compared to no treatment. Romi did not affect the frequency of CD8+ TNFα+/IFNγ+ populations nor the expression of Granzyme B in CD8+ T cells. CD4+ and CD8+ T cells did not express TPO-R on their surface. To confirm the immunomodulatory role of Elt in vivo, the terminally-differentiated effector (CD45RA+CD62L-) CD8+ T cells were assessed in 13 Elt-treated patients and 11 Romi-treated patients. Patients on Elt had significantly reduced frequency of effector CD8 T cells compared to Romi-treated patients (44% vs 76.8%; p<0.01). Taken together, these novel findings suggest an off target immunomodulatory nature of Elt besides its thrombopoietic effect. This dose-dependent immunomodulatory effect is not TPO-R dependent and targets T cells primarily. This study is the first to display such property of Elt and could explain why there is a differential response to Elt and Romi. We hypothesise that Elt may be more effective in patients with T cell mediated disease, whilst patients with predominantly antibody mediated disease are more likely respond to Romi. These findings can also offer an explanation for Elt effectiveness in other T cell-mediated autoimmune conditions such as Aplastic Anemia. Disclosures Cooper: Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Rigel: Consultancy, Membership on an entity's Board of Directors or advisory committees; Principia: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees.


2022 ◽  
Vol 12 (3) ◽  
pp. 625-629
Author(s):  
Chunpei Ou ◽  
Pengfei Chen ◽  
Jinqi Song ◽  
Xuefeng Deng ◽  
Feiqiang Chen ◽  
...  

Osteoarthritis (OA) is a degenerative disease of joints commonly occurring in the elderly and middleaged people. This study aimed to investigate the effect of recombinant human endostatin (rhEndo) on OA and the levels of MMP-13, IL-1 and IL-6 in the synovial fluid in osteoarthritis rats. OA models were made by injecting 4% papain into the knee joint cavity of rats once every three days for three times. The models were then injected subcutaneously with rhEndo and examined six weeks later for the Mankin scores and levels of MMP-13, IL-1 and IL-6 using ELISA. Compared with control, the Mankin score as well as the levels of IL-1, IL-6 and MMP-13 were significantly increased in the models (0.30 vs. 5.80, 1.12 vs. 12.84 pg/ mL, 12.22 vs. 43.82 pg/ mL and 0.23 vs. 26.31 ng/ mL). Following treatment with 4 mg/kg rhEndo, the Mankin score in model decreased to 0.90, meanwhile, the levels of IL-1, IL-6 and MMP-13 decreased significantly to 0.79 pg/ mL, 2.89 pg/mL and 1.17 ng/mL, respectively, in a dose dependent manner. Therefore, rhEndo can alleviate osteoarthritis by reducing MMP-13, IL-1 and IL-6 expression in rats.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3204-3204
Author(s):  
Nainong Li ◽  
Chunyan Zhang ◽  
Chia-Lei Lin ◽  
Bill McCulloch ◽  
John Wright ◽  
...  

Abstract Hematopoietic cell transplantation (HCT) is a curative therapy for hematological malignancies as well as refractory autoimmune diseases. However, graft versus host disease (GVHD) remains a major obstacle in classical HCT, where recipients are usually conditioned with total body irradiation or high dose chemotherapy. We recently reported that donor CD8+ T cells facilitated engraftment and mediated graft versus leukemia (GVL) without causing graft versus host disease (GVHD) in young (6–8 weeks old) MHC-mismatched mouse recipients conditioned with anti-CD3 mAb (Blood 2005). Thereafter, we observed that anti-CD3 conditioning alone was not sufficient for induction of chimerism in old (>12 weeks) recipients, due to the higher percentage of residual host CD8+ T cells in the old recipients. Romidepsin (Desipeptide), a histone deacetylase inhibitor, was reported to induce apoptosis of human T cell lymphoma lines. We hypothesize that depsipeptide will induce the apoptosis of anti-CD3 activated proliferating T cells, and that conditioning with a combination of anti-CD3 and depsipeptide will markedly reduce the residual host T cells and allow donor stem cell engraftment. To test our hypothesis, we first added Romidepsin (1.25–10 ng/ml) to cultures of T cells with or without anti-CD3 stimulation. We found that, although Romidepsin showed no effect on un-stimulated T cells, it augmented apoptosis of anti-CD3 activated T cells in a dose dependent manner, and the maximum augmentation was 5 fold. In addition, when Romidepsin (1.25–10 ng/ml) was added to a culture of mixed lymphocyte reaction (MLR), we found that it suppressed MLR in a dose dependent manner also, and the maximum suppression was greater than 98%. Second, old (> 12 weeks) BALB/c recipients were conditioned with one I.V. injection of anti-CD3 (20μg/g) and three I.P. injections (every other day) of Romidepsin at a dose of 0.4 μg/g. 7 days after anti-CD3 injection, recipients were injected with donor bone marrow cells (100×106) and CD4+- T depleted spleen (CD4−-SPL) cells (100×106). CD4−-SPL cells were injected again 7 days later. We found that, 4 weeks after HCT, 7/8 of the recipients conditioned with a combination of anti-CD3 and Romidepsin but only 1/8 of the recipients conditioned with anti-CD3 alone became chimeric. The recipients showed healthy appearance without signs of GVHD. The results are combined from two replicate experiments. This radiation free and GVHD preventive conditioning regimen may provide a novel approach for clinical HCT.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1804-1804
Author(s):  
David J. Chung ◽  
Marco Rossi ◽  
Jennifer Pressley ◽  
David H. Munn ◽  
James W. Young

Abstract Effective immunotherapy must overcome tolerance toward tumor antigens and avoid subsequent inhibition of stimulated antitumor immunity. The specific contribution of immune regulatory mechanisms intrinsic to dendritic cells (DCs), especially with regard to regulatory T cells (T regs), is of emerging importance. We have found that all conventional, immunogenic human DCs express the immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO) and that IDO protein expression and activity are markedly increased in mature compared with immature DCs. Priming of resting T cells with mature IDO+ DCs in an autologous mixed leukocyte reaction (MLR) increases the proportion of CD4+CD25+ T cells capable of suppressing allogeneic T cell proliferation in secondary MLRs as much as 10-fold above baseline (Figure 1A). Conversely, 1-methyl-tryptophan, a competitive inhibitor of IDO, dampens the inhibitory activity (Figure 1A). Further characterization of the suppressor T cells was performed after cytofluorographic sorting into CD4+CD25hi, CD4+CD25int, and CD4+CD25low/− subpopulations. Post-sort analysis revealed that the majority (&gt;60%) of the CD4+CD25hi cells coexpressed Foxp3, which was absent in the CD4+CD25low/− cells. Separate studies showed that these Foxp3+ cells express little or no CD127 (IL-7R-alpha). Candidate CD4+CD25hi T regs inhibited DC-stimulated allogeneic T cell proliferation in a dose dependent manner, with &gt;90% inhibition at a suppressor to responder T cell ratio of 1:1 and ∼50% inhibition at a ratio as low as 1:25 (Figure 1B). CD4+CD25low/− cells were not inhibitory, and CD4+CD25int cells exerted intermediate suppression depending on dose (Figure 1B). CD4+CD25hi T regs exert similar inhibition of autologous T cell responses to stimulation de novo by DCs. Both the priming and effector phases of T reg suppression were contact dependent. Moreover, depletion of the trace population of CD4+CD25hi T cells at the outset of autologous priming largely abolished the relative expansion of this population. These results clearly demonstrate that mature conventional human DCs support relative but significant expansion of autologous, constitutive CD4+CD25hi T cells, which coexpress Foxp3, express little or no CD127, and exert significant suppression of both allogeneic and autologous T cells stimulated de novo by DCs. Although contrary to the anticipated enrichment of IDO in immature DCs because of their expected tolerogenicity, these findings underscore the importance of regulatory mechanisms exerted by immunogenic cells like mature conventional DCs. While this may provide a physiologic means of turning off an otherwise unchecked immune response, this IDO-mediated pathway in DCs provides a rational target for optimizing host immune responses against tumor antigens. This should result in more sustained benefit from active immunotherapy with DC-based vaccines. FIGURE 1: A) T cell mediated suppression of secondary allogeneic MLR. Autologous T eels primed in the presence of the IDO inhibitor. 1-metnyl-D-tryptophan (1-MT), do not develop as much suppressor activity as T cells primed in the absence of 1-MT, resulting in loss inhibition of the secondary MLR Data presented are representative of 6 experiments. B) CD4−CD25***** T cells art potent inhibitors of T cell proliferation. FACS-sorted CD4−CD25******, CD4−CD25*****, and CD4−CD25***** T cells (‘suppressor T cells’) were added to allogeneic MLRs composed of autologous DCs + allogeneic T cells. DC to T cell ratio was 1:30. Suppressor T cell to responder T cell ratios were 1:25, 1:5, and 1:1. After 4–5 days in culture, responder T cell proliferation was assessed by measuring 3HTdR incorporation and comparing with controls containing no suppressor cells (black bar). CD4−CD25***** cells inhibited T cell proliferation in a dose-dependent manner. CD4−CD25***** cells inhibited proliferation to a lesser degree, and CD4-CD25***** cells showed no inhibition. While the CD4−CD25***** cells and to a lesser extent the CD4−CD25***** cells suppressed the proliferative response of alloreactive T cells in the MLRs, they were themselves anergic to the allogeneic DCs in proportion to CD25 and FOXP3 expression (data not shown). Values are pooled from 3 independent experiments (N=4 for each condition), and error bars indicate standard deviation of the mean. FIGURE 1:. A) T cell mediated suppression of secondary allogeneic MLR. Autologous T eels primed in the presence of the IDO inhibitor. 1-metnyl-D-tryptophan (1-MT), do not develop as much suppressor activity as T cells primed in the absence of 1-MT, resulting in loss inhibition of the secondary MLR Data presented are representative of 6 experiments. B) CD4−CD25***** T cells art potent inhibitors of T cell proliferation. FACS-sorted CD4−CD25******, CD4−CD25*****, and CD4−CD25***** T cells (‘suppressor T cells’) were added to allogeneic MLRs composed of autologous DCs + allogeneic T cells. DC to T cell ratio was 1:30. Suppressor T cell to responder T cell ratios were 1:25, 1:5, and 1:1. After 4–5 days in culture, responder T cell proliferation was assessed by measuring 3HTdR incorporation and comparing with controls containing no suppressor cells (black bar). CD4−CD25***** cells inhibited T cell proliferation in a dose-dependent manner. CD4−CD25***** cells inhibited proliferation to a lesser degree, and CD4-CD25***** cells showed no inhibition. While the CD4−CD25***** cells and to a lesser extent the CD4−CD25***** cells suppressed the proliferative response of alloreactive T cells in the MLRs, they were themselves anergic to the allogeneic DCs in proportion to CD25 and FOXP3 expression (data not shown). Values are pooled from 3 independent experiments (N=4 for each condition), and error bars indicate standard deviation of the mean.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3932-3932
Author(s):  
Mary Faris ◽  
Uriel M Malyankar ◽  
Qingping Zeng ◽  
Gary A Flynn ◽  
Gerold Feuer ◽  
...  

Abstract Abstract 3932 ITK (Interluekin-2 Inducible Tyrosine Kinase) is a member of the TEC family of intracellular protein tyrosine kinases. ITK is highly expressed in T cells and NK cells, with expression detected in mast cells. ITK plays a key role in several aspects of T cell biology, including T cell development, differentiation, migration, proliferation and activation. The function of ITK in immunity and allergy is well documented. T cells from ITK knock out mice show several developmental and functional defects, including defective signal transduction, altered CD4+ to CD8+ T cells ratios, reduced Th2 lineage differentiation, diminished IL4 and IL2 production and reduced T cell proliferation. Importantly ITK deficient mice fail to mount an immune response to infection and show reduced allergic asthma reactions. In contrast to its well described role in immune function, ITK's function in cancer biology is still emerging. Recent studies had reported enhanced ITK expression and activation of the ITK pathway in several types of leukemias and lymphomas. In addition, the dependence of T cell malignancies on an ITK-regulated pathway, namely the IL2/IL2R (CD25) pathway, has also been observed. Taken together, this information indicates that ITK is a therapeutic target, with applicability in leukemias and lymphomas. MannKind scientists have developed a series of selective small molecule ITK inhibitors, including the orally available tool compound described within, and evaluated their activity in enzyme, cell-based and in vivo studies. In cellular assays, the compounds showed significant inhibition of the T cell-receptor mediated activation of the ITK pathways and related downstream cytokine production. In addition to inhibiting the phosphorylation of ITK and its downstream mediator, PLCg, our tool compounds inhibited the production of IL2 and expression of CD25 in a dose dependent manner. Importantly, our compound regulated the in vitro growth of tumor T cells but not that of unrelated control cells. In vivo studies revealed that the tool compounds inhibited the growth and progression of patient derived ATL tumors in a xenograft pre-clinical model, and prolonged the survival of treated mice in a dose dependent manner, in addition to regulating cytokine production in vivo. In summary, our team has identified ITK selective compounds with demonstrated on-target and anti-tumor activity in vitro and preclinical T cell tumor models, and validated this pathway relative to T cell malignancies. This effort provides a platform for further compound optimization and evaluation for hematologic malignancies. Disclosures: Faris: MannKind Corp: Employment. Malyankar:MannKind Corp: Employment. Zeng:MannKind Corp: Employment. Kertesz:Mannkind Corporation: Employment, Equity Ownership. Vuga:MannKind Corp.: Employment. Rosario:MannKind Corp: Employment. Bot:MannKind Corp: Employment.


Sign in / Sign up

Export Citation Format

Share Document