scholarly journals Screening of FDA-Approved Drug Library Identifies Adefovir Dipivoxil as Highly Potent Inhibitor of T Cell Proliferation

2021 ◽  
Vol 11 ◽  
Author(s):  
Linda Voss ◽  
Karina Guttek ◽  
Annika Reddig ◽  
Annegret Reinhold ◽  
Martin Voss ◽  
...  

Repositioning of approved drugs for identifying new therapeutic purposes is an alternative, time and cost saving strategy to classical drug development. Here, we screened a library of 786 FDA-approved drugs to find compounds, which can potentially be repurposed for treatment of T cell-mediated autoimmune diseases. Investigating the effect of these diverse substances on mitogen-stimulated proliferation of both, freshly stimulated and pre-activated (48 h) peripheral blood mononuclear cells (PBMCs), we discovered Adefovir Dipivoxil (ADV) as very potent compound, which inhibits T cell proliferation in a nanomolar range. We further analyzed the influence of ADV on proliferation, activation, cytokine production, viability and apoptosis of freshly stimulated as well as pre-activated human T cells stimulated with anti-CD3/CD28 antibodies. We observed that ADV was capable of suppressing the proliferation in both T cell stimulation systems in a dose-dependent manner (50% inhibition [IC50]: 63.12 and 364.8 nM for freshly stimulated T cells and pre-activated T cells, respectively). Moreover, the drug impaired T cell activation and inhibited Th1 (IFN-γ), Th2 (IL-5), and Th17 (IL-17) cytokine production dose-dependently. Furthermore, ADV treatment induced DNA double-strand breaks (γH2AX foci expression), which led to an increase of p53-phospho-Ser15 expression. In response to DNA damage p21 and PUMA are transactivated by p53. Subsequently, this caused cell cycle arrest at G0/G1 phase and activation of the intrinsic apoptosis pathway. Our results indicate that ADV could be a new potential candidate for treatment of T cell-mediated autoimmune diseases. Prospective studies should be performed to verify this possible therapeutic application of ADV for such disorders.

Blood ◽  
2010 ◽  
Vol 116 (22) ◽  
pp. 4501-4511 ◽  
Author(s):  
Willemijn Hobo ◽  
Frans Maas ◽  
Niken Adisty ◽  
Theo de Witte ◽  
Nicolaas Schaap ◽  
...  

Tumor relapse after human leukocyte antigen–matched allogeneic stem cell transplantation (SCT) remains a serious problem, despite the long-term presence of minor histocompatibility antigen (MiHA)–specific memory T cells. Dendritic cell (DC)–based vaccination boosting MiHA-specific T-cell immunity is an appealing strategy to prevent or counteract tumor recurrence, but improvement is necessary to increase the clinical benefit. Here, we investigated whether knockdown of programmed death ligand 1 (PD-L1) and PD-L2 on monocyte-derived DCs results in improved T-cell activation. Electroporation of single siRNA sequences into immature DCs resulted in efficient, specific, and long-lasting knockdown of PD-L1 and PD-L2 expression. PD-L knockdown DCs strongly augmented interferon-γ and interleukin-2 production by stimulated T cells in an allogeneic mixed lymphocyte reaction, whereas no effect was observed on T-cell proliferation. Moreover, we demonstrated that PD-L gene silencing, especially combined PD-L1 and PD-L2 knockdown, resulted in improved proliferation and cytokine production of keyhole limpet hemocyanin–specific CD4+ T cells. Most importantly, PD-L knockdown DCs showed superior potential to expand MiHA-specific CD8+ effector and memory T cells from leukemia patients early after donor lymphocyte infusion and later during relapse. These data demonstrate that PD-L siRNA electroporated DCs are highly effective in enhancing T-cell proliferation and cytokine production, and are therefore attractive cells for improving the efficacy of DC vaccines in cancer patients.


2007 ◽  
Vol 75 (4) ◽  
pp. 1730-1737 ◽  
Author(s):  
Ilaria Peluso ◽  
Daniele Fina ◽  
Roberta Caruso ◽  
Carmine Stolfi ◽  
Flavio Caprioli ◽  
...  

ABSTRACT Recent studies have shown that probiotics are beneficial in T-cell-mediated inflammatory diseases. The molecular mechanism by which probiotics work remains elusive, but accumulating evidence indicates that probiotics can modulate immune cell responses. Since T cells express receptors for bacterial products or components, we examined whether different strains of lactobacilli directly regulate the functions of human T cells. CD4+ T cells were isolated from blood and intestinal lamina propria (LP) of normal individuals and patients with inflammatory bowel disease (IBD). Mononuclear cells were also isolated from Peyer's patches. Cells were activated with anti-CD3/CD2/CD28 in the presence or absence of Lactobacillus paracasei subsp. paracasei B21060, L. paracasei subsp. paracasei F19, or L. casei subsp. casei DG. Cell proliferation and death, Foxp3, intracellular pH, and cytokine production were evaluated by flow cytometry. We showed that L. paracasei subsp. paracasei B21060 but neither L. paracasei subsp. paracasei F19 nor L. casei subsp. casei DG inhibited blood CD4+ T-cell growth. This effect was associated with no change in cell survival, expression of Foxp3, or production of gamma interferon, interleukin-4 (IL-4), IL-5, and IL-10. L. paracasei subsp. paracasei B21060-mediated blockade of CD4+ T-cell proliferation required a viable bacterium and was associated with decreased MCT-1 expression and low intracellular pH. L. paracasei subsp. paracasei B21060 also inhibited the growth of Peyer's patch mononuclear cells, normal lymphocytes, and IBD CD4+ LP lymphocytes without affecting cytokine production. The data show that L. paracasei subsp. paracasei B21060 blocks T-cell growth, thus suggesting a mechanism by which these probiotics could interfere with T-cell-driven immune responses.


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 (>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 >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.


1985 ◽  
Vol 161 (4) ◽  
pp. 641-656 ◽  
Author(s):  
T Hara ◽  
S M Fu

Three monoclonal antibodies (mAb), of IgG1, IgG2a, and IgM isotypes, raised against the T3 complex, were used to probe the activation of human T cells. The IgM antibody 235 was not mitogenic for peripheral blood mononuclear cells (PMC). It efficiently blocked the proliferation of PMC induced by T cell mitogens, alloantigens, and soluble antigens. The other two antibodies were mitogenic, and behaved similarly to Leu 4 and OKT3, respectively. In T cell preparations with less than 0.1% monocytes (as assayed by nonspecific esterase staining), all three mAb were not mitogenic. They failed to induce either interleukin 2 (IL-2) receptor expression or IL-2 secretion. Addition of IL-1 failed to collaborate with anti-T3 mAb to induce these T cells to proliferate, but IL-2 enhanced T cell proliferation slightly. Monocyte-depleted T cells, however, proliferated in response to all three anti-T3 mAb, when TPA was added, in a dose-dependent manner. TPA induced a low level of IL-2 receptor expression in monocyte-depleted T cells, without inducing IL-2 secretion. Anti-T3 plus TPA induced a marked enhancement in both quantity and intensity of IL-2 receptor expression. IL-2 secretion was also detected. These results indicate that anti-T3 IgM can deliver an inductive signal despite its blockage of T cell proliferation, and that two signals are necessary and perhaps sufficient to induce human T cell activation and proliferation.


2021 ◽  
Vol 22 (1) ◽  
Author(s):  
Haiting Wang ◽  
Xiangyu Teng ◽  
Georges Abboud ◽  
Wei Li ◽  
Shuang Ye ◽  
...  

Abstract Background Systemic lupus erythematosus is an autoimmune disease characterized by an overproduction of autoantibodies resulting from dysregulation in multiple immune cell types. D-mannose is a C− 2 epimer of glucose that exhibits immunoregulatory effects in models of autoimmune diseases, such as type 1 diabetes, induced rheumatoid arthritis, and airway inflammation. This study was conducted to evaluate the efficacy of D-mannose treatment in mouse models of lupus. Results Firstly, the effect of D-Mannose was evaluated by flow cytometry on the in vitro activation of non-autoimmune C57BL/6 (B6) bone marrow-derived dendritic cells (BMDCs) and their ability to induce antigen-specific CD4+ T cell proliferation and activation. D-mannose inhibited the maturation of BMDCs and their induction of antigen-specific T cell proliferation and activation. In vivo, D-mannose increased the frequency of Foxp3+ regulatory T cells in unmanipulated B6 mice. To assess the effect of D-mannose in mouse models of lupus, we used the graft-versus-host disease (cGVHD) induced model and the B6.lpr spontaneous model. In the cGVHD model, D-mannose treatment decreased autoantibody production, with a concomitant reduction of the frequency of effector memory and follicular helper T cells as well as germinal center B cells and plasma cells. These results were partially validated in the B6.lpr model of spontaneous lupus. Conclusion Overall, our results suggest that D-mannose ameliorates autoimmune activation in models of lupus, at least partially due to its expansion of Treg cells, the induction of immature conventional dendritic cells and the downregulation of effector T cells activation. D-Mannose showed however a weaker immunomodulatory effect in lupus than in other autoimmune diseases.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3557-3557
Author(s):  
Yukio Kondo ◽  
Luis Espinoza ◽  
Takamasa Katagiri ◽  
Zhirong Qi ◽  
Shinji Nakao

Abstract Abstract 3557 Poster Board III-494 Immunization of allogeneic stem cell transplant (SCT) recipients with leukemia-associated antigens (LAAs) is an attractive approach to the augmentation of graft-versus-leukemia (GVL) effect. However, the induction of CTLs specific to LAAs is hampered by various inhibitory molecules expressed on leukemic cells that restrain the T cell function in connection with their receptors on T cells. Even if the cellular immunity is rebuilt after SCT by T cells of the donor origin, overcoming such an escape mechanism is required to effectively induce the CTLs specific to TAAs by vaccination after allogeneic SCT. Glucocorticoid-induced TNFR-related protein (GITR) belongs to the TNF receptor superfamily and is expressed on NK cells, CD25+ regulatory T cells and activated T cells. The binding of GITR ligand (GITRL) on leukemic cells to GITR on NK cells restrains NK cell activity but the influence on T cells of the GITR/GITRL binding has not been clarified. Myeloid dendritic cells derived from myeloid leukemic cells express GITRL which inhibits induction of LAA-specific CTLs (Blood 2008; 112:817a). The mechanisms of the negative effect on the induction of LAA-specific T cells through the GITR/GITRL interaction was investigated to improve the efficiency of the CTL induction. The expression of GITRL was observed on leukemic cells from 9 of 16 patients with myeloid leukemia and a monocytic leukemia cell line THP-1, and soluble GITRL (sGITRL) was detectable in the serum from 3 of 5 patients as well as in the culture supernatant of THP-1 cells. CFSE-labeled pan T cell, CD4+ T cell and CD8+ T cell proliferation in response to microbeads coated with anti-CD3 and anti-CD28 monoclonal antibodies (CD3/CD28 microbeads) was suppressed to 55.0%, 63.6%, 65.8% of the controls in a culture supernatant of THP-1 cells, and was restored to 86.9%, 65.1% and 76.8% respectively by addition of sGITR to block the binding of sGITRL in the supernatant and GITR on T cells. Flow cytometry detected GITRL in exosomes, which express HLA class II, purified from the culture supernatant of THP-1 with anti-HLA class II antibody-coated microbeads, and CFSE-labeled pan T cell, CD4+ T cell and CD8+ T cell proliferation was restrained as well by the addition of GITRL+ exosomes in a dose dependent manner (27.6%, 54.1%, 27.9% reduction of proliferation with 10 μl exosome, respectively). Indoleamine 2, 3-dioxygenase (IDO) activity in plasmacytoid DC (pDC) is negatively correlated with the activity of CD4+ T cells induced by their interaction with the pDC through the GITR/GITRL interaction in a mouse model. Kynurenine (Kyn), a metabolite of tryptophan in leukemic cells that is broken down by IDO, suppressed CFSE-labeled pan T cell, CD4+ T cell and CD8+ T cell proliferation in response to CD3/CD28 microbeads in a dose dependent manner (24.5%, 12.3%, 18.3% reduction in the proliferation at 100 μM, respectively). Significantly higher concentrations of Kyn were detected in the supernatant of THP-1 cells after incubation in the presence of sGITR than a control, and the production of Kyn was suppressed by the addition of an IDO inhibitor, 1-Methyl Tryptophan (1MT) (Fig). Moreover, the addition of sGITR to leukemic cells from five patients with AML induced Kyn (Fig). These findings indicate that GITRL on leukemic cells and sGITRL secreted by leukemic cells as an exosome protein suppress the induction of LAA-specific CTLs by directly binding GITR on LAA-specific CTLs, increasing the IDO activity in leukemic cells and inducing Kyn secretion from leukemic cells. The administration of anti-IDO agents or anti-GITRL blocking Abs combined with LAA vaccination may therefore effectively induce LAA-specific T cells in SCT recipients. Fig GITR/GITRL binding induces kyn secretion from THP-1 cell and primary AML cells. Fig. GITR/GITRL binding induces kyn secretion from THP-1 cell and primary AML cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3905-3905
Author(s):  
Bernd Jahrsdoerfer ◽  
Karen Dahlke ◽  
Magdalena Hagn ◽  
Kai Sontheimer ◽  
Thamara Beyer ◽  
...  

Abstract Abstract 3905 Immune regulation is central for the development of an efficient cellular immune response. Both Treg cells and plasmacytoid DC can suppress T cell proliferation in a granzyme B (GzmB)-dependent and perforin-independent manner. In the present study we found that, depending on stimulation with interleukin (IL-) 21, B cells (BC) can also express GzmB and effectively suppress T cell proliferation. GzmB expression in BC is enhanced by BC receptor engagement, and is suppressed by CD40 ligation. Since CD4+ T cells are a main source of IL-21, we tested whether they can induce GzmB in BC. We found that incompletely activated CD4+ T cells, but not fully activated T cells induce GzmB in co-cultured BC. Using confocal microscopy, we showed that BC-derived GzmB is enzymatically active and that GzmB+ BC transfer GzmB to CD4+ T cells. Furthermore, GzmB+ BC decreased CD4+ T cell expression of the TCR-zeta chain, a GzmB target, which is required for T cell proliferation. Our results suggest BC may regulate cellular adaptive immune responses by Treg cell-like mechanisms. Inhibition of BC-derived GzmB may represent a novel strategy to induce more effective and comprehensive cellular immune responses. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Jan-Hendrik Riedel ◽  
Jan-Eric Turner ◽  
Ulf Panzer

AbstractCD4+ T cells are key drivers of autoimmune diseases, including crescentic GN. Many effector mechanisms employed by T cells to mediate renal damage and repair, such as local cytokine production, depend on their presence at the site of inflammation. Therefore, the mechanisms regulating the renal CD4+ T cell infiltrate are of central importance. From a conceptual point of view, there are four distinct factors that can regulate the abundance of T cells in the kidney: (1) T cell infiltration, (2) T cell proliferation, (3) T cell death and (4) T cell retention/egress. While a substantial amount of data on the recruitment of T cells to the kidneys in crescentic GN have accumulated over the last decade, the roles of T cell proliferation and death in the kidney in crescentic GN is less well characterized. However, the findings from the data available so far do not indicate a major role of these processes. More importantly, the molecular mechanisms underlying both egress and retention of T cells from/in peripheral tissues, such as the kidney, are unknown. Here, we review the current knowledge of mechanisms and functions of T cell migration in renal autoimmune diseases with a special focus on chemokines and their receptors.


2005 ◽  
Vol 25 (16) ◽  
pp. 6869-6878 ◽  
Author(s):  
Jin Han ◽  
Jr-Wen Shui ◽  
Xuejun Zhang ◽  
Biao Zheng ◽  
Shuhua Han ◽  
...  

ABSTRACT Engagement of the T-cell receptor (TCR) triggers a series of signaling events that lead to the activation of T cells. HIP-55 (SH3P7 or mAbp1), an actin-binding adaptor protein, interacts with and is tyrosine phosphorylated by ZAP-70, which is a crucial proximal protein tyrosine kinase for TCR signaling. HIP-55 is important for JNK and HPK1 activation induced by TCR signaling. In this study, we report the generation and characterization of HIP-55 knockout mice. We found that HIP-55 knockout mice were viable and fertile but showed decreased body weight and increased occurrence of death within the first 4 weeks after birth. The lymphoid organs in HIP-55 knockout mice showed cellularity and T-cell development comparable to that of the wild-type mice. HIP-55 knockout T cells displayed defective T-cell proliferation, decreased cytokine production, and decreased up-regulation of the activation markers induced by TCR stimulation. TCR internalization was slightly increased in HIP-55 knockout T cells. These phenotypes were accompanied by reduced immune responses, including antigen-specific antibody production and T-cell proliferation in HIP-55 knockout mice. The TCR-induced signaling events, including LAT/phospholipase Cγ1 phosphorylation and HPK1/JNK activation, were partially defective in HIP-55 knockout T cells. These results demonstrate the importance of HIP-55 as an adaptor protein in the TCR signaling and immune system.


Sign in / Sign up

Export Citation Format

Share Document