scholarly journals Multiple Myeloma Cell-Derived Interleukin-32gamma Increases the Immunosuppressive Function of Macrophages By Promoting Indoleamine 2,3-Dioxygenase (IDO) Expression

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3179-3179
Author(s):  
Haimeng Yan ◽  
Donghua He ◽  
Xi Huang ◽  
Zhang En Fan ◽  
He Huang ◽  
...  

Abstract Background: The interaction of multiple myeloma (MM) cells with macrophages (MΦs) in the bone marrow microenvironment contributes to the pathophysiology of MM. In addition to promoting angiogenesis through vasculogenic mimicry, MM-associated MΦs (mMΦs) protect MM cells from spontaneous and chemotherapy-induced apoptosis. mMΦs therefore represent a potential target for myeloma treatment and it is essential to explore the mechanisms underlying normal MΦ polarization to mMΦs. We previously showed that IL-32 is overexpressed in MM patients and is mainly derived from MM cells. The present study was designed to explore the clinical significance of IL-32 in MM and to further elucidate the molecular mechanisms underlying the IL-32-mediated immune function of MΦs. Methods: We examined the expression of IL-32 in bone marrow biopsy samples using immunohistochemistry. Quantitative real-time PCR, western blot analysis and immunofluorescence were applied to measure the expression of IL-32, IDO and proteinase 3 (PR3). We obtained the global transcriptional profile of the IL-32γ-treated MΦs by RNA sequencing (RNA-Seq). Immunoprecipitation (IP) and GST pulldown experiments was applied to confirm the binding affinity of PR3 for IL-32. We created IL-32-knockdown MM cells by transfection of IL-32 shRNA and silenced PR3 expression in MΦs using siRNA targeting PR3. CD4+ T cell proliferation and IL-2, IFN-γ and TNF-α production were measured by flow cytometry. Results: We found that high IL-32 expression in MM patients was associated with advanced clinical stage and high serum β2-microglobulin levels. Several isoforms of IL-32 were detected in MM cells and IL-32γ was the most active subtype. RNA sequencing revealed that IL-32γ significantly induced the production of the immunosuppressive molecule indoleamine 2,3-dioxygenase (IDO) in MΦs and this effect was verified at the protein level. Furthermore, IL-32-knockdown MM cells showed less ability than control MM cells to promote IDO expression. As a binding protein for IL-32, PR3 was universally expressed on the surface of MΦs and knockdown of PR3 or inhibition of the STAT3 and nuclear factor κB (NF-κB) pathways hindered the IL-32γ-mediated stimulation of IDO expression. Finally, IDO-positive IL-32γ-educated MΦs inhibited CD4+ T cell proliferation and IL-2, IFN-γ and TNF-α production in response to activation. Conclusion: Our study showed that MM cell-derived IL-32γ induced IDO production in MΦs through PR3 and the downstream STAT3 and NF-κB pathways, resulting in the suppression of the proliferation and effector function of CD4+ T cells. High IL-32 expression in MM may contribute to an immunosuppressive microenvironment by upregulating IDO production in MΦs and promote MM progression. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
1997 ◽  
Vol 90 (11) ◽  
pp. 4513-4521 ◽  
Author(s):  
Dieter Körholz ◽  
Ursula Banning ◽  
Halvard Bönig ◽  
Markus Grewe ◽  
Marion Schneider ◽  
...  

Abstract Interleukin-15 (IL-15) is a potent T-cell stimulating factor, which has recently been used for pre-clinical in vivo immunotherapy. Here, the IL-15 effect on CD3-stimulated peripheral human T cells was investigated. IL-15 induced a significant T-cell proliferation and upregulated CD25 expression. IL-15 significantly enhanced T-cell production of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and IL-10. Between 10- and 100-fold greater concentrations of IL-15 were necessary to reach a biological effect equivalent to that of IL-2. Blockade of IL-2 binding to the high-affinity IL-2 receptor did not affect the IL-15 effects, suggesting that IL-15 did not act by inducing endogenous IL-2. Exogenously administered IL-10 significantly reduced the IL-15 and IL-2–mediated IFN-γ and TNF-α production, whereas T-cell proliferation and CD25 expression were not affected. The inhibitory effects of exogenously administered IL-10 on T-cell cytokine production appeared indirect, and are likely secondary to decreased IL-12 production by accessory cells. Inhibition of endogenous IL-10 binding to the IL-10 receptor significantly increased IFN-γ and TNF-α release from T cells. These data suggest that endogenous IL-10 can regulate activated T-cell production of IFN-γ and TNF-α via a paracrine negative feedback loop. The observations of this study could be of relevance for the therapeutic use of IL-15 in vivo.


Blood ◽  
2006 ◽  
Vol 109 (8) ◽  
pp. 3351-3359 ◽  
Author(s):  
Adriano Boasso ◽  
Jean-Philippe Herbeuval ◽  
Andrew W. Hardy ◽  
Stephanie A. Anderson ◽  
Matthew J. Dolan ◽  
...  

AbstractInfection with the human immunodeficiency virus type-1 (HIV) results in acute and progressive numeric loss of CD4+ T-helper cells and functional impairment of T-cell responses. The mechanistic basis of the functional impairment of the surviving cells is not clear. Indoleamine 2,3-dioxygenase (IDO) is an immunosuppressive enzyme that inhibits T-cell proliferation by catabolizing the essential amino acid tryptophan (Trp) into the kynurenine (kyn) pathway. Here, we show that IDO mRNA expression is elevated in peripheral blood mononuclear cells (PBMCs) from HIV+ patients compared with uninfected healthy controls (HCs), and that in vitro inhibition of IDO with the competitive blocker 1-methyl tryptophan (1-mT) results in increased CD4+ T-cell proliferative response in PBMCs from HIV-infected patients. We developed an in vitro model in which exposure of PBMCs from HCs to either infectious or noninfectious, R5- or X4-tropic HIV induced IDO in plasmacytoid dendritic cells (pDCs). HIV-induced IDO was not inhibited by blocking antibodies against interferon type I or type II, which, however, induced IDO in pDCs when added to PBMC cultures. Blockade of gp120/CD4 interactions with anti-CD4 Ab inhibited HIV-mediated IDO induction. Thus, induction of IDO in pDCs by HIV may contribute to the T-cell functional impairment observed in HIV/AIDS by a non–interferon-dependent mechanism.


Blood ◽  
2009 ◽  
Vol 113 (3) ◽  
pp. 612-621 ◽  
Author(s):  
Mirko Paiardini ◽  
Barbara Cervasi ◽  
Jessica C. Engram ◽  
Shari N. Gordon ◽  
Nichole R. Klatt ◽  
...  

AbstractBone marrow (BM) is the key hematopoietic organ in mammals and is involved in the homeostatic proliferation of memory CD8+ T cells. Here we expanded on our previous observation that BM is a preferential site for T-cell proliferation in simian immunodeficiency virus (SIV)–infected sooty mangabeys (SMs) that do not progress to AIDS despite high viremia. We found high levels of mature T-cell proliferation, involving both naive and memory cells, in healthy SMs and rhesus macaques (RMs). In addition, we observed in both species that lineage-specific, BM-based T-cell proliferation follows antibody-mediated in vivo CD4+ or CD8+ T-cell depletion, thus indicating a role for the BM in maintaining T-cell homeostasis under depleting circumstances. We also observed that, in SIV-infected SMs, but not RMs, the level of proliferation of BM-based CD4+ T cells is higher than that of circulating CD4+ T cells. Interestingly, limited BM-based CD4+ T-cell proliferation was found in SIV-infected SMs with low CD4+ T-cell counts, suggesting a regenerative failure in these animals. Collectively, these results indicate that BM is involved in maintaining T-cell homeostasis in primates and suggest a role for BM-based CD4+ T-cell proliferation in determining the benign nature of natural SIV infection of SMs.


2016 ◽  
Vol 2016 ◽  
pp. 1-17 ◽  
Author(s):  
Sebastien Hagmann ◽  
Claudia Rimmele ◽  
Florin Bucur ◽  
Thomas Dreher ◽  
Felix Zeifang ◽  
...  

Introduction. The participation of an inflammatory joint milieu has been described in osteoarthritis (OA) pathogenesis. Mesenchymal stromal cells (MSCs) play an important role in modulating inflammatory processes. Based on previous studies in an allogeneic T-cell coculture model, we aimed at further determining the role of synovial MSCs in OA pathogenesis.Methods. Bone-marrow (BM) and synovial membrane (SM) MSCs from hip joints of late stage OA patients and CD4+ T-cells from healthy donors were analysed regarding surface marker expression before and after coculture. Proliferation upon CD3/CD28 stimulation and cytokine analyses were compared between MSCs.Results. SM-MSCs differed from BM-MSCs in several surface markers and their osteogenic differentiation potential. Cocultures of both MSCs with CD4+ T-cells resulted in recruitment of CD45RA+ FoxP3+ regulatory T-cells. Upon stimulation, only SM-MSCs suppressed CD4+ T-cell proliferation, while both SM-MSCs and BM-MSCs modified cytokine profiles through suppressing IL-2 and TNF-αas well as increasing IL-6 secretion.Conclusions. Synovial MSCs from OA joints are a unique fraction that can be distinguished from their bone-marrow derived counterparts. Their unique ability to suppress CD3/CD28 induced CD4+ T-cell proliferation makes them a potential target for future therapeutic approaches.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4109-4109
Author(s):  
Nathalie van Leeuwen-Kerkhoff ◽  
Theresia M. Westers ◽  
Hetty J Bontkes ◽  
Shahram Kordasti ◽  
Ghulam J. Mufti ◽  
...  

Abstract Introduction Immune response plays an important role in the pathogenesis and progression of myelodysplastic syndromes (MDS). It has been shown that a consistent immunological signature in MDS comprises an increase in Th17 cells in low risk MDS and an expansion of Tregs in high risk subtypes. The presence of high Treg numbers in lower risk stages is an independent prognostic value and is associated with poor survival. Whereas the immune response in low risk MDS is pro-inflammatory, high risk MDS is characterized by immunosuppression which could contribute into the expansion of the dysplastic clone and malignant transformation. The exact role of dendritic cells (DC) in inducing this immune dysfunction is yet to be understood. The aim of this comparative study was therefore to investigate the frequency and function of different DC subsets in the bone marrow (BM) and peripheral blood (PB) of MDS patients compared to healthy donors (HD). Patients and methods The enumeration of DC subsets was performed in 110 MDS and in 19 HD bone marrow samples. In peripheral blood 34 MDS and 18 HD samples were investigated. DC were identified by the expression of HLA-DR and the lack of the lineage markers CD14 and CD19. They were further subdivided into CD303+ plasmacytoid DC (pDC), and the CD11c+ myeloid subsets CD1c+ myeloid DC 1 (mDC1), CD141hi myeloid DC 2 (mDC2) and CD16+ 6-Sulfo LacNac (Slan) DC. Furthermore, their ability to upregulate co-stimulatory molecules was assessed by flow cytometry and the secretion of cytokines in response to TLR ligands was analyzed using Cytometric Bead Array (CBA). Their capacity to induce allogeneic T cell proliferation was evaluated in a mixed leukocyte reaction (MLR). Results The frequencies of pDC, mDC1 and mDC2 but not of SlanDC were significantly lower in patients' BM compared to HDs' BM (pDC 0.33% vs 0.72%, p=0.01; mDC1 0.30% vs 0.73%, p<0.001; mDC2 0.01% vs 0.05%, p<0.0001; SlanDC 0.10% vs 0.13%, p=0.20). In PB all DC subsets were significantly lower in patients' samples compared to HDs' samples (pDC 0.15% vs 0.28%, p=0.02; mDC1 0.12% vs 0.61%, p<0.0001; mDC2 0.003% vs 0.045%, p<0.0001; SlanDC 0.19% vs 0.46%, p=0.04). A positive correlation was found between the frequencies of DC subsets in a paired assessment of PB and BM of 30 MDS patients (pDC r=0.77, mDC1 r=0.57, mDC2 r=0.34, SlanDC r=0.60). Of note, in a more advanced disease state DC subsets showed a progressive decline in frequency. To investigate DC function they were isolated or FACS sorted from HD BM (n=3) or patient BM (n=4). Based on their TLR expression profile a combination of LPS and R848 has been used to stimulate DC overnight. In HD upregulation of the co-stimulatory molecules CD80 and CD86 was observed after this stimulation. However, MDS DC stimulated with the same combination of TLR ligands showed a reduced upregulation of CD80 and CD86 (median fluorescence intensity (MFI) CD80: SlanDC 2323 vs 6254, MFI CD86: mDC1 2785 vs 4608; SlanDC 2344 vs 5087). They also produced lower levels of pro-inflammatory cytokines compared to HD DC as measured in culture supernatants (mDC1 TNF [pg/ml] 997 vs 2980, IL-6 [pg/ml] 843 vs 2806, IL-8 [pg/ml] 966 vs 5549 and IL-12p70 [pg/ml] 105 vs 1115). Additionally, after 5 days co-culture of CFSE labeled T cells and DC, mDC1 and SlanDC derived from MDS BM displayed a defective induction of allogeneic CD4+ and CD8+ T cell proliferation compared to HD (mDC1 3.08% vs 10.49% CD4+ T cell proliferation and 4.54% vs 15.07% CD8+ T cell proliferation; SlanDC 1.28% vs 5.23% CD4+ T cell proliferation and 1.50% vs 8.67% CD8+ T cell proliferation). Conclusion Our data clearly point to both a numeric and functional impairment of myeloid DC subsets in BM as well as in PB of patients with MDS. A further decline of DC frequencies in high risk MDS may support the therapeutic targeting of DC in the BM microenvironment of these patients to redress immune dysfunction and possibly prevent further progression to acute myeloid leukemia. Extensive research is needed to further delineate the differences in DC function and immune composition in MDS risk categories. Disclosures Mufti: Celgene Corporation: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.


2005 ◽  
Vol 175 (3) ◽  
pp. 1586-1592 ◽  
Author(s):  
Tomohide Yamazaki ◽  
Hisaya Akiba ◽  
Akemi Koyanagi ◽  
Miyuki Azuma ◽  
Hideo Yagita ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1211-1211
Author(s):  
Ying Qu ◽  
Zhengxu Sun ◽  
Yan Yuan ◽  
Fen Wang ◽  
Kunpeng Wu ◽  
...  

Aplastic anemia (AA) is a hematopoietic disorder resulted from immune-related hypocellular hematopoiesis in bone marrow (BM). It has been clearly addressed that the activated T cells contribute to the exhaustion of hematopoietic progenitors and hypo-hematopoiesis. The adipogenic BM is one of the characteristics to make AA diagnosis. However, little is known about the relationship of intra-BM immune imbalance and hematopoietic microenvironment abnormity in this disease entity. Functional hematopoiesis relies on not only abundant hematopoietic stem cells (HSCs) but also the balanced supportive hematopoietic niche. Intra-BM immune balance, at either cellular or cytokine level, is one of the key footstones to maintain hematopoietic microenvironment. Various intra-BM immune cellular components play both sides of one coin. Among them, myeloid-derived suppressive cells (MDSCs) are heterogeneous myeloid progenitor cells characterized by the negative immune response in cancers and other inflammatory diseases. In BM aspiration and biopsy samples from the patients who were diagnosed as AA in our study, massive activated lymphocytes infiltration and adipocytes accumulation were observed. Interestingly, the absolute numbers of immune modulatory MDSCs either in AA patients' PB or in BM of immune-related AA mice were reduced, indicating a potential link between polarized BM adipo-osteogenic microenvironment and immune disorder under AA circumstance. We thus adopted AA mice model to look into the embedded details both in vivo and in vitro. We clarified that BM components were more vulnerable to the attack of CD8+ T cells than that of CD4+ T cells. Taking into the fact that BM adipocytes are more abundant either in AA patients or in AA mice models, we differentiated mesenchymal stromal cells (MSCs), the major BM stroma cells, into osteoblastic or adipogenic lineages to mimic the osteo-adipogenic differentiation in BM microenvironment. Interestingly, CD8+ T cells and interferon-γ(IFN-γ) exerted dramatically adipocytic stimulation on BM-MSCs either in vitro or in vivo, by determination of increasing expression of adipogenetic genes including Ap2, Perilipin, Pparg and Cebpα, as well as staining of Oil Red O and perilipin. To dissect intra-BM cellular immune balance, MDSCs were isolated as representative immune regulating population to investigate their function on osteo-adipogenic balance. Interestingly, not CD11b+Ly6G+Ly6C-granulocytic-MDSCs (gMDSCs) but CD11b+Ly6G-Ly6C+monocytic-MDSCs (mMDSCs) inhibited both T cell proliferation and IFN-γ production. Addition of L-NMMA, the antagonist of iNOS pathway in mMDSCs-containing system restored T cell proliferative curve and cell numbers, whereas Nor-NOHA, the antagonist of Arg-1 pathway didn't abrogate mMDSCs' immune-regulation properties, indicating that mMDSCs inhibited T cell proliferation via iNOS pathway. We then performed single dose or multi-dose injection of mMDSCs in AA mice to see whether mMDSCs are able to reconstitute the impacted hematopoiesis. Single injection of mMDSCs was able to prevent from CTL infiltration in a very short term. However, multi-injection of mMDSCs showed significant benefit in overall survival rate compared to AA mice. We further detected the function of mMDSCs on polarized BM-MSCs adipo-osteogenic differentiation potential. To detect sequential BM adipogenetic progression in AA microenvironment, we performed in vivo fluorescent microscopy on AP2 (Fabp4)-Cre×mT/mG reporting mice at different transfusion time points of T cells and mMDSCs. GFP-expressing AP2+ adipocytes accumulated adjacently to perivascular niches whose boarders were labelled by Dextran-CY5 in a time-dependent manner after T cell infusion. Monocytic MDSCs transfused AA mice showed decreased GFP+ adipocytes which was coincident with our in vitro findings. In conclusion, intra-BM immune balance is one of the environmental factors seesawing by activating and suppressive ends to support functional hematopoiesis. Adoptive transfusion of mMDSCs, the immune-suppressive population might be a novel immune-regulating strategy to treat AA, relying on not only restoring the intra-BM immune balance but also improving stroma's multi-differentiating microenvironment. Figure Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document