scholarly journals Therapeutic targeting of NOTCH signaling ameliorates immune-mediated bone marrow failure of aplastic anemia

2013 ◽  
Vol 210 (7) ◽  
pp. 1311-1329 ◽  
Author(s):  
Justine E. Roderick ◽  
Gabriela Gonzalez-Perez ◽  
Christina Arieta Kuksin ◽  
Anushka Dongre ◽  
Emily R. Roberts ◽  
...  

Severe aplastic anemia (AA) is a bone marrow (BM) failure (BMF) disease frequently caused by aberrant immune destruction of blood progenitors. Although a Th1-mediated pathology is well described for AA, molecular mechanisms driving disease progression remain ill defined. The NOTCH signaling pathway mediates Th1 cell differentiation in the presence of polarizing cytokines, an action requiring enzymatic processing of NOTCH receptors by γ-secretase. Using a mouse model of AA, we demonstrate that expression of both intracellular NOTCH1IC and T-BET, a key transcription factor regulating Th1 cell differentiation, was increased in spleen and BM-infiltrating T cells during active disease. Conditionally deleting Notch1 or administering γ-secretase inhibitors (GSIs) in vivo attenuated disease and rescued mice from lethal BMF. In peripheral T cells from patients with untreated AA, NOTCH1IC was significantly elevated and bound to the TBX21 promoter, showing NOTCH1 directly regulates the gene encoding T-BET. Treating patient cells with GSIs in vitro lowered NOTCH1IC levels, decreased NOTCH1 detectable at the TBX21 promoter, and decreased T-BET expression, indicating that NOTCH1 signaling is responsive to GSIs during active disease. Collectively, these results identify NOTCH signaling as a primary driver of Th1-mediated pathogenesis in AA and may represent a novel target for therapeutic intervention.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 180-180
Author(s):  
Gloria T Shan ◽  
Ivy Tran ◽  
Ashley R Sandy ◽  
Ann Friedman ◽  
Yi Zhang ◽  
...  

Abstract Abstract 180 Aplastic anemia is a severe bone marrow disorder characterized by the loss of hematopoietic stem cells (HSC). HSC destruction is thought to be T cell-mediated in a majority of patients with aplastic anemia. Global immunosuppression and HSC transplantation can induce disease remission, but these treatments are not effective in all patients and can promote life-threatening complications. Thus, novel immunomodulatory approaches are needed in this disorder. Notch is a conserved cell-cell communication pathway that can regulate T cell differentiation and function with context-dependent effects. To study the role of Notch signaling in pathogenic T cells causing immune-mediated bone marrow failure, we inhibited canonical Notch signaling in mature T cells through conditional expression of the pan-Notch inhibitor DNMAML (ROSA-DNMAMLf × Cd4-Cre mice). We used two complementary mouse models of immune-mediated bone marrow failure that mimic features of aplastic anemia: administration of C57BL/6 (B6) T cells into sublethally irradiated (500 rads) minor histocompatibility antigen mismatched BALB/b recipients (Chen et al., J Immunol 2007; 178:4159), or infusion of B6 lymphocytes into unirradiated MHC-mismatched B6×DBA F1 recipients. In contrast to control B6 T cells which led to lethal bone marrow failure in virtually all recipients, DNMAML-expressing Notch-deprived T cells were profoundly deficient at inducing HSC loss in both disease models, leading to markedly improved long-term survival (>90%). Notch-deficient T cells showed a modest decrease in overall expansion within secondary lymphoid organs, but their accumulation in the target bone marrow was preserved. Upon restimulation with anti-CD3 and anti-CD28 antibodies, DNMAML T cells had decreased production of IL-2 and interferon gamma. Activated CD4+ and CD8+ DNMAML T cells had reduced interferon gamma, granzyme B, and perforin transcripts despite preserved induction of the master transcription factors Tb×21 (encoding T-bet) and Eomes. In vivo infusion of CFSE-labeled host-type target cells revealed a decreased cytotoxicity in DNMAML as compared to control B6 T cell recipients. These observations point to a novel spectrum and mechanism of Notch action in mature T cells. Since we have shown recently that canonical Notch signaling is dispensable for the maintenance of adult HSCs (Maillard et al., Cell Stem Cell 2008, 2:356), our findings suggest that Notch inhibition could represent a novel therapeutic modality to target the T cell response and reverse immune-mediated HSC destruction in aplastic anemia. Disclosures: Shan: American Society of Hematology: Research Funding. Zhang:University of Michigan Comprehensive Cancer Center: Research Funding; Damon Runyon Cancer Research Foundation: Research Funding. Maillard:Damon Runyon Cancer Research Foundation: Research Funding; American Society of Hematology: Research Funding; University of Michigan Comprehensive Cancer Center: Research Funding.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2467-2467
Author(s):  
Qing Tong ◽  
Shan He ◽  
Fang Xie ◽  
Amanda Wong ◽  
Kazuhiro Mochizuki ◽  
...  

Abstract Severe acquired aplastic anemia (AA) is a fatal disorder characterized by immune-mediated destruction of hematopoietic stem and progenitor cells. Evidence in most AA patients indicates that IFN-g-producing T helper (Th)1 effector CD4+ T cells are important for mediating bone marrow (BM) failure in AA. However, the efficacy of standard therapies that typically include antithymocyte globulin and cyclosporine A is limited, and novel approaches are urgently needed. Ezh2 is a histone methyltransferase that specifically catalyzes trimethylation of histone H3 at lysine 27 (H3K27me3) and acts primarily as a gene silencer. We investigated whether Ezh2 regulatory control governs Th1 immune-mediated cytopenias in AA. We tested this hypothesis in a mouse model of AA using genetic approaches of Ezh2 inhibition. In naïve CD4+ T cells, high levels of H3K27me3 are correlated with repressed expression of IFNG and TBX21, the gene that encodes T-bet, which is essential for inducing IFN-g expression. Upon Th1 cell differentiation, the regulatory regions of both IFNG and TBX21 gene loci show a marked reduction of H3K27me3. We found that Ezh2 is required to induce Th1 cell differentiation and T cell-mediated AA in mice. Conditionally deleting Ezh2 in mature T cells had the effect of dramatically reducing the production of Th1 cells secreting high levels of IFN-g in vivo, decreasing BM-infiltrating Th1 cells during active disease, and rescuing mice from BM failure. In vitro culture assays confirmed that Ezh2-deficient T cells showed significantly reduced production of IFN-g under Th1-skewing conditions compared to wild-type (WT) T cells. This effect of Ezh2 deficiency on Th1 cell differentiation was accompanied by a marked decrease in the expression of both IFNG and TBX21 genes. These results stand in sharp contrast to the conventional view that Ezh2 and its catalyzed H3K27me3 may repress gene expression, and the corollary that loss of Ezh2 may result in increased production of IFN-g and T-bet. Using chromatin immunoprecipitation assay, we found that upon Th1 cell differentiation in vitro, naïve WT CD4+ T cells showed a significant reduction of H3K27me3 at the regulatory region of both IFNG and TBX21 gene loci, in agreement with previous reports. In contrast, high levels of Ezh2 were detected at the regulatory region of the TBX21 gene in activated WT CD4+ T cells, suggesting that Ezh2 may be required to promote TBX21 transcription during Th1 cell development. To test this possibility, we infected Ezh2-deficient CD4+ T cells with a retrovirus construct encoding T-bet. Ectopic expression of T-bet rescued Th1 cell differentiation of Ezh2-deficient T cells in vitro. Collectively, our findings identify a critical role for Ezh2 in regulating Th1 responses and AA. Given the availability of Ezh2-specific inhibitors newly developed for cancer therapy in clinical trials, we propose that targeting Ezh2 should be investigated as a new strategy for treating AA in patients. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Yi Yu ◽  
Alejandra Vargas Valderrama ◽  
Zhongchao Han ◽  
Georges Uzan ◽  
Sina Naserian ◽  
...  

Abstract Background Mesenchymal stem cells (MSCs) exhibit active abilities to suppress or modulate deleterious immune responses by various molecular mechanisms. These cells are the subject of major translational efforts as cellular therapies for immune-related diseases and transplantations. Plenty of preclinical studies and clinical trials employing MSCs have shown promising safety and efficacy outcomes and also shed light on the modifications in the frequency and function of regulatory T cells (T regs). Nevertheless, the mechanisms underlying these observations are not well known. Direct cell contact, soluble factor production, and turning antigen-presenting cells into tolerogenic phenotypes, have been proposed to be among possible mechanisms by which MSCs produce an immunomodulatory environment for T reg expansion and activity. We and others demonstrated that adult bone marrow (BM)-MSCs suppress adaptive immune responses directly by inhibiting the proliferation of CD4+ helper and CD8+ cytotoxic T cells but also indirectly through the induction of T regs. In parallel, we demonstrated that fetal liver (FL)-MSCs demonstrates much longer-lasting immunomodulatory properties compared to BM-MSCs, by inhibiting directly the proliferation and activation of CD4+ and CD8+ T cells. Therefore, we investigated if FL-MSCs exert their strong immunosuppressive effect also indirectly through induction of T regs. Methods MSCs were obtained from FL and adult BM and characterized according to their surface antigen expression, their multilineage differentiation, and their proliferation potential. Using different in vitro combinations, we performed co-cultures of FL- or BM-MSCs and murine CD3+CD25−T cells to investigate immunosuppressive effects of MSCs on T cells and to quantify their capacity to induce functional T regs. Results We demonstrated that although both types of MSC display similar cell surface phenotypic profile and differentiation capacity, FL-MSCs have significantly higher proliferative capacity and ability to suppress both CD4+ and CD8+ murine T cell proliferation and to modulate them towards less active phenotypes than adult BM-MSCs. Moreover, their substantial suppressive effect was associated with an outstanding increase of functional CD4+CD25+Foxp3+ T regs compared to BM-MSCs. Conclusions These results highlight the immunosuppressive activity of FL-MSCs on T cells and show for the first time that one of the main immunoregulatory mechanisms of FL-MSCs passes through active and functional T reg induction.


Blood ◽  
1985 ◽  
Vol 66 (5) ◽  
pp. 1043-1046
Author(s):  
GD Goss ◽  
MA Wittwer ◽  
WR Bezwoda ◽  
J Herman ◽  
A Rabson ◽  
...  

Bone marrow transplantation for severe idiopathic aplastic anemia was undertaken in a patient, using his monozygotic twin brother as the donor. In spite of the use of syngeneic bone marrow, failure of engraftment occurred on two occasions. In vitro studies demonstrated that natural killer (NK) cells from the recipient markedly inhibited the growth of donor bone marrow granulocyte progenitor cells. On a third attempt, successful bone marrow engraftment was achieved following high-dose cyclophosphamide, which has previously been shown to be inhibitory to NK cells. We conclude that NK cell activity may play an important role in bone marrow failure as well as being responsible for at least some cases of aplastic anemia.


Blood ◽  
2013 ◽  
Vol 122 (14) ◽  
pp. 2453-2459 ◽  
Author(s):  
Andres Jerez ◽  
Michael J. Clemente ◽  
Hideki Makishima ◽  
Hanna Rajala ◽  
Ines Gómez-Seguí ◽  
...  

Key PointsSTAT3+ T cells are found not only in detected concomitant LGL-BMFs, but in cases in which an LGL expansion was not suspected. Transformation via acquisition of a somatic mutation in T cells may be a mechanism of immune, mainly hypoplastic, bone marrow failure.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1141-1141
Author(s):  
Elena E. Solomou ◽  
Valeria Visconte ◽  
Federica Gibellini ◽  
Neal S. Young

Abstract Ligation of the signaling lymphocyte activation molecule (SLAM), a member of the immunoglobulin superfamily expressed in T and B cells, results in T cell activation and Th1 cytokine production. SAP is a small cytoplasmic protein expressed in T- and NK cells that controls the activation signals mediated by SLAM. On T cell activation, SAP binds to Fyn kinase; Fyn is activated and phosphorylates tyrosine residues on SLAM; phosphorylation results in the formation of a complex that selectively down-regulates co-stimulatory signals in activated T cells, resulting in inhibition of IFN-γ production. Thus SAP acts as a natural suppressor of SLAM-mediated T cell activation, and, in the absence of SAP, T cells are constitutively activated and overproduce IFN-γ. Mutations in the SAP gene lead to abnormal T cell activation and enhanced Th1 cytokine production in mouse models and in humans: about half of patients with X-linked lympoproliferative disease (XLP) have functionally disabling SAP mutations. Acquired aplastic anemia (AA) is a bone marrow failure syndrome in which hematopoietic cell destruction is effected by cytotoxic T cells and type 1 cytokines. We have recently shown that T cells from patients with AA have increased protein levels of T-bet, resulting in IFN-γ overproduction (Solomou EE et al, Blood2006; 107:3983). IFN-γ inhibits hematopoietic stem cell proliferation and induces Fas-mediated apoptosis; stem cell depletion results in marrow hypoplasia and peripheral blood pancytopenia. We examined SAP expression as an explanation for aberrant T cell activation and extreme Th1 polarization. SAP protein expression on immunoblot was very low to absent in unstimulated T cells from 16 of 20 AA patients examined, as compared to normal levels of expression in equivalent numbers of healthy donors (p<0.001). No significant differences were detected in Fyn and SLAM protein levels between AA and controls. SAP mRNA levels were also significantly decreased in T cells from those AA patients with low SAP protein levels, as determined by RT-PCR. Peripheral blood DNA samples from 18 patients with AA were analyzed for SAP mutations: three novel intronic mutations, not present in controls, were identified among 7 unrelated patients: one mutation was in the promoter region of SAP (position 106, C to T; 3 patients), and two mutations in the intron-exon junction between exons 1 and 2 (position 38975, C toT; 3 patients) and 3 and 4 (position 62771, C to A; 1 patient). IFN-γ, as measured by ELISA, in three patients with undetectable SAP protein levels was significantly increased compared to healthy controls (n=5, p<0.001). Increased IFN-γ levels and Th1 polarization in AA can in part be explained by functional SAP deficiency. SAP-deficient T cells in AA would be unable to block co-stimulatory signals, leading to an activated T cell phenotype and ultimately hematopoietic cell destruction and bone marrow failure. The SAP-deficient phenotype in T cells from patients with aplastic anemia may be secondary to subtle genetic alteration in the gene’s regulation (abnormal promoter binding sites or epigenetic modulation due to mutations in introns) or as yet unidentified aberrant upstream pathways (Ets-1 and Ets-2, the transcription factors that regulate SAP expression).


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4218-4218
Author(s):  
Jiayu Chen ◽  
Weiwei Liu ◽  
Lingzhen Chen ◽  
Xiaohuai Wang ◽  
Weimin Zhang ◽  
...  

Abstract We have developed a novel cell-based immunotherapy for treatment of some hematopoietic and blood deficient diseases such as aplastic anemia, chemotherapy-induced severe myelosuppression, idiopathic thrombocytopenia purpura and autoimmunity-induced cytopenia. Autologous and/or allogeneic peripheral blood mononuclear cells were cultured in vitro with a combination of cytokines and a calcium mobilizing agent for 2 days before given to patients via intravenous infusion. The immunotherapy has been shown to have potent activities in stimulating multi-lineage hematopoiesis and blood production including platelet production, which remains a major clinical problem to be solved. The immunotherapy is more effective for treatment of chronic and severe bone marrow failure and inefficient blood production than currently available growth factors of G-CSF, GM-CSF, Erythropoietin and IL-11. The mechanism of the immunotherapy is yet completely clear to us, however, some evidence suggests that in vitro activated immune cells produce and secrete multiple cytokines, working in concert, these cytokines released by the infused cells in organs important for hematopoiesis and blood production such as bone marrow, liver and spleen have remarkable effects on target cells, resulting in improved hematopoiesis, blood cell differentiation and maturation. In the preliminary clinical studies, more than 100 patients with aplastic anemia, severe chemotherapy-induced myelosuppression, systemic lupus erythematosus-associated cytopenia and idiopathic thrombocytopenia refractory to conventional therapies have been treated with the immunotherapy and the results have been encouraging. In severe idiopathic and benzene-induced aplastic anemia, 90% patients have complete or partial remission after the immunotherapy and one and half year survival is 90%. We have used 2–5x108in vitro activated allogeneic immune cells per infusion per day for 5 consecutive days, followed by small numbers of autologous infusions (1 to 10 million from approximately 50 ml of peripheral blood, depending on the severity of the disease, once a week for 4 weeks). This cycle of therapy is repeated till patient’s absolute neutrophil count is more than 0.5x109/L. The duration of the immunotherapy required for patients with AA to significantly improve ranges from two months to two years depending on the severity of the disease. Idiopathic thrombocytopenia is as difficult as aplastic anemia to treat for the immunotherapy and also requires relatively long time (several months to a year) for patients to respond to the therapy. Approximately 50% adult patients treated with the immunotherapy have complete or partial remission. In severe myelosuppression induced by chemotherapy in leukemia patients, the immunotherapy is highly effective and capable of reducing infection, bleeding and blood transfusion. The recovery of severe myelosuppression (from a few days to a month depending on the severity) after the immunotherapy is much quicker than that of aplastic anemia and idiopathic thrombocytopenia purpura. In conclusion, animal and preliminary human clinical studies suggest that the immunotherapy is highly effective for some bone marrow failure and blood deficient disorders, which are usually difficult to treat with the conventional therapies. The immunotherapy described here merits further investigation.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3239-3239
Author(s):  
Wei Du ◽  
Ozlem Erden ◽  
Andrew Wilson ◽  
Jared Sipple ◽  
Jonathan Schick ◽  
...  

Abstract Fanconi anemia (FA) is a genetic disorder associated with bone marrow failure and leukemia. Recent studies demonstrate fundamental immune defects in FA. However, the mechanisms that are critical for FA immunodeficiency are not known. Here we report that deletion of Fanca or Fancd2 dysregulates the suppressive activity of regulatory T cells (Tregs) and exacerbates graft-versus-host disease (GVHD) in mice. Recipient mice of Fanca-/- or Fancd2-/- bone marrow chimeras exhibited severe acute GVHD after allogeneic bone marrow transplantation (BMT). Further study showed that T cells from Fanca-/- or Fancd2-/- mice induced higher GVHD lethality than those from WT littermates. Mechanistically, FA Tregs possessed lower proliferative suppression potential compared to WT Tregs, as demonstrated by in vitro proliferation assay and BMT. Analysis of CD25+Foxp3+ Tregs indicated that loss of Fanca or Fancd2 dysregulated Foxp3 transcriptional activity. Additionally, CD25+Foxp3+ Tregs of Fanca-/- or Fancd2-/- mice were less efficient in suppressing the production of GVHD-associated inflammatory cytokines. Consistently, incremental NF-kB transcriptional activity was observed in infiltrated T cells from FA GVHD mice. Conditional deletion of p65 in FA Tregs desreased GVHD mortality. Our study uncovers an essential role for the FA proteins in maintaining Treg homeostasis and suggests that targeted blocking NF-kB signaling within T cells represents an attractive therapeutic strategy to ameliorate GVHD in FA. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 88 (12) ◽  
pp. 4493-4499 ◽  
Author(s):  
A Wodnar-Filipowicz ◽  
SD Lyman ◽  
A Gratwohl ◽  
A Tichelli ◽  
B Speck ◽  
...  

Flt3 ligand (flt3L) is a member of a small family of cytokines acting as tyrosine kinase receptor ligands that stimulate the proliferation of primitive hematopoietic progenitors in vitro. To gain insight into the physiological role of flt3L in early hematopoiesis, levels of flt3L were determined in serum of patients with multilineage bone marrow failure and related to the severity of stem cell depletion. In patients with aplastic anemia (AA) and in cancer patients with chemotherapy-induced transient suppression of hematopoiesis, flt3L fluctuated in an inverse relationship to the degree of bone marrow failure. In severe AA at diagnosis, levels of circulating soluble flt3L were highly elevated (2,653 +/- 353 pg/mL) as compared with normal blood serum values of 14 +/- 39 pg/mL. Flt3L returned to near normal levels within the first 3 months following successful bone marrow transplantation and in autologous remission induced by immunosuppressive therapy with antilymphocyte globulin (ALG; 100 +/- 31 and 183 +/- 14 pg/mL, respectively). In contrast, rejection of the graft or relapse of the disease after ALG was accompanied by an increase to high pretreatment concentrations of the circulating cytokine (3,770 +/- 2,485 and 1,788 +/- 233 pg/mL, respectively). Flt3L in serum inversely correlated with the colony-forming ability of AA bone marrow precursors in vitro (R = - .86), indicating that the concentration of the ligand reflects hematopoiesis at the progenitor cell level. Flt3L increased to 2,500 pg/mL in the serum of leukemia patients during chemoradiotherapy- induced bone marrow suppression and returned to normal values along with hematopoietic recovery. Expression of the membrane-bound form of flt3L was significantly elevated in mononuclear bone marrow and peripheral blood cells from patients with severe pancytopenia, suggesting de novo synthesis of the factor in response to bone marrow failure. The data provide a strong argument for the involvement of flt3L in the regulation of early hematopoiesis in vivo.


Sign in / Sign up

Export Citation Format

Share Document