scholarly journals Flt3 ligand level reflects hematopoietic progenitor cell function in aplastic anemia and chemotherapy-induced bone marrow aplasia

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.

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 ◽  
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 ◽  
2009 ◽  
Vol 114 (6) ◽  
pp. 1186-1195 ◽  
Author(s):  
Hyung Chan Suh ◽  
Ming Ji ◽  
John Gooya ◽  
Michael Lee ◽  
Kimberly D. Klarmann ◽  
...  

Abstract Development of hematopoietic stem cells (HSCs) and their immediate progeny is maintained by the interaction with cells in the microenvironment. We found that hematopoiesis was dysregulated in Id1−/− mice. Although the frequency of HSCs in Id1−/− bone marrow was increased, their total numbers remained unchanged as the result of decreased bone marrow cellularity. In addition, the ability of Id1−/− HSCs to self-renew was normal, suggesting Id1 does not affect HSC function. Id1−/− progenitors showed increased cycling in vivo but not in vitro, suggesting cell nonautonomous mechanisms for the increased cycling. Id1−/− HSCs developed normally when transplanted into Id1+/+ mice, whereas the development of Id1+/+ HSCs was impaired in Id1−/− recipients undergoing transplantation and reproduced the hematologic features of Id1−/− mice, indicating that the Id1−/− microenvironment cannot support normal hematopoietic development. Id1−/− stromal cells showed altered production of cytokines in vitro, and cytokine levels were deregulated in vivo, which could account for the Id1−/− hematopoietic phenotypes. Thus, Id1 is required for regulating the hematopoietic progenitor cell niche but is dispensable for maintaining HSCs.


Blood ◽  
2004 ◽  
Vol 104 (8) ◽  
pp. 2299-2306 ◽  
Author(s):  
Halvard Bonig ◽  
Gregory V. Priestley ◽  
Lina M. Nilsson ◽  
Yi Jiang ◽  
Thalia Papayannopoulou

Abstract Several examples suggest a relationship between in vitro migratory capacity and bone marrow (BM) homing. Pertussis toxin (PTX) is a potent inhibitor of serpentine receptor–associated inhibitory trimeric guanidine nucleotide binding (Gi) protein signals. As such, it blocks hematopoietic progenitor cell migration in vitro, but contrary to expectation, no effects on BM homing were observed in previous studies. We therefore re-examined the effect of PTX on homing of murine BM and fetal liver (FL). We found that BM homing of PTX-incubated progenitor cells (colony-forming cells in culture [CFU-Cs]) from BM or FL in irradiated and nonirradiated recipients was reduced by more than 75%, with a concomitant increase in circulating CFU-Cs in peripheral blood. Additional studies confirmed the functional significance of this reduction in homing: PTX-treated cells did not provide radioprotection, and their short-term engraftment in BM and spleen was drastically reduced. Furthermore, several approaches show that cell-intrinsic rather than host-derived mechanisms are responsible for the PTX-induced homing defect. In summary, we show that Gi protein signals are required for BM homing and, as such, provide a new example of the association between BM homing and in vitro migration. Moreover, our data suggest that the behavior of hematopoietic progenitors in obeying Gi signaling does not diverge from that of mature leukocytes.


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.


Hematology ◽  
2008 ◽  
Vol 2008 (1) ◽  
pp. 104-110 ◽  
Author(s):  
Monica Bessler ◽  
Jeffrey Hiken

Abstract Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired hemolytic anemia caused by the expansion of a hematopoietic progenitor cell that has acquired a mutation in the X-linked PIGA gene. PNH occurs on the background of bone marrow failure. Bone marrow failure and the presence of the abnormal cells account for the clinical phenotype of patients with PNH including hemolysis, cytopenia, and thrombophilia. PIGA is essential for the synthesis of glycosyl phosphatidylinositol (GPI) anchor molecules. PNH blood cells are therefore deficient in all proteins that use such an anchor molecule for attachment to the cell membrane. Two of these proteins regulate complement activation on the cell surface. Their deficiency therefore explains the exquisite sensitivity of PNH red blood cells to complement-mediated lysis. Complement-mediated lysis of red blood cells is intravascular, and intravascular hemolysis contributes significantly to the morbidity and mortality in patients with this condition. PNH is an outstanding example of how an increased understanding of pathophysiology may directly improve the diagnosis, care, and treatment of disease.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3483-3483
Author(s):  
Kazuya Sato ◽  
Xingmin Feng ◽  
Jichun Chen ◽  
Marie J. Desierto ◽  
Keyvan Keyvanfar ◽  
...  

Abstract Abstract 3483 In aplastic anemia (AA), the marrow is not “empty” but replaced by fat; the increase in adipocytes number and size is most obvious on three dimensional reconstructions of the marrow in human and murine (Takaku T, Blood. 2008). A reciprocal relationship exists between adipogenesis and osteogenesis, and osteoblasts constitute the hematopoietic niche and play an active role in the regulation of stem cells and progenitors. Fat in the marrow has been considered an epiphenomenon in AA. However, a recent report suggested that bone marrow (BM) adipocytes negatively regulated hematopoiesis in mouse models (Naveiras O, Nature. 2009). Peroxisome proliferator-activated receptor-g (PPAR-g) is a key transcription factor for adipogenesis, and blocking PPAR-g signaling inhibited adipogenesis in vitro (Wright HM, J Biol Chem. 2000). To examine the role of BM adipocytes, we investigated the effects of PPAR-g antagonists, bisphenol A diglycidyl ether (BADGE, 30 mg/kg/day) and GW9662 (1 mg/kg/day), on hematopoiesis in a mouse model of immune-mediated BM failure (Chen J, J Immunol. 2007). We induced BM failure by infusion of lymph node (LN) cells from C57BL/6 mice into sublethally irradiated C.B10-H2(b)/LilMcd (C.B10) recipient mice that were matched at major histocompatibility antigens but differed in multiple minor histocompatibility antigens. In adaptation of the “runt” disease model, mice uniformly develop progressive and fatal pancytopenia, closely resembling human BM failure, without other evidence of graft-versus-host disease. We treated recipient mice with BADGE, GW9662, or control vehicle from day -1 to day 14. On day 14, mice were sacrificed and evaluated by peripheral blood (PB) cell counting and BM cellularity, as well as morphology of marrow adipocytes. Mice in the BADGE- and GW9662-treated groups showed higher numbers of leukocytes, neutrophils, and platelets in PB and higher total nucleated cells and Lin- Sca1+ c-kit+ stem cells in BM than did animals in the control group. Both confocal microscopic imaging and hematoxylin and eosin staining of BM also showed significantly higher numbers of nucleated cells and many fewer and smaller adipocytes in the treated groups (Figure 1). We also investigated dose response of BADGE in the treatment of AA mice. Low dose of BADGE (15 mg/kg/day) had no effect while high dose of BADGE (60 mg/kg/day) seemed to have no extra benefit for the BM hematopoiesis compared with the medium dose (30 mg/kg/day). However, we also noted in PPAR-g antagonist-treated groups that there was significantly less CD8+ T cell infiltration of BM, as determined by flow cytometry. We speculated that PPAR- g antagonists might also negatively affect activation of cytotoxic T cells. By magnetic beads-based multiplex assay, we found the concentrations of inflammation-related cytokines in the plasma, including Interleukin-6, tumor necrosis factor alpha, monocyte chemotactic protein-1 were markedly decreased in PPAR- g antagonist-treated groups. When we performed PCR arrays focusing on adipogenesis and inflammasome pathways, we found that expression of adipogenesis genes was greatly decreased in the treated groups, including Agt (−149 folds), Cebpa (−4.7 folds), Acacb (−11.7 folds), Fabp4 (−3.2 folds), Adig (−14.2 folds), and Bmp2 (−12.9 folds). The expression of inflammation- or inflammasome-related genes including Nlrc4 (−11.3 folds), Mapk12 (−4.8 folds), Ptgs2 (−8.7 folds), and Rela (−5.9 folds) was also decreased while apoptosis inhibitor genes including Xiap (+17.5 folds), Mapk1 (+6.6 folds), and Bcl2l1 (+3.9 folds) were increased in the treated groups. In vitro, BADGE and GW9662 inhibited activation and proliferation of T cells stimulated with anti-CD3/CD28 or phorbol myristate acetate/ionomycin. These data suggested that BADGE and GW9662 inhibition was not specific for adipogenesis but affected T cell activation. Indeed, PPAR-g antagonists failed to ameliorate pancytopenia and BM hypoplasia in the mice exposed to either a lethal or sublethal dose of total body irradiation. PPAR-g antagonists may act to attenuate murine immune mediated marrow failure by mechanism of inhibition of T-cell activation. Figure 1. Histology of femurs from untreated bone marrow failure mice and PPAR-g antagonists treated mice. Both BADGE and GW9662 inhibited adipogenesis and increased cellularity in the bone marrow of AA mice. Figure 1. Histology of femurs from untreated bone marrow failure mice and PPAR-g antagonists treated mice. Both BADGE and GW9662 inhibited adipogenesis and increased cellularity in the bone marrow of AA mice. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 296-296 ◽  
Author(s):  
Carine Domenech ◽  
Alix Rousseau ◽  
Laurence Petit ◽  
Sandra Sanfilippo ◽  
Jean Soulier ◽  
...  

Abstract Fanconi anemia (FA) is a genetic disorder due to mutations in one of the sixteen FANC genes involved in DNA repair. Many FA patients develop bone marrow failure (BMF) during childhood, and FA strongly predisposes to myelodysplasia syndrome and/or acute myeloid leukaemia. The pathogenesis of the BMF remains uncompletely understood. Low hematopoietic progenitor cell (HPCs) counts observed early in life and preceeding the onset of blood cytopenia in patients led we, and other, to hypothesize that the hematopoietic development might be abnormal in the FA embryo. Indeed, unlike adult hematopoietic stem cells (HSCs) which are quiescent in the BM niche, during embryonic life HSCs are in active proliferation in sites of expansion such as fetal liver and placenta, where they get amplified and acquire properties of adult HSC .We hypothesized that in FA, the FA defect in response to the replicative stress could impair the expension of the HSC pool.In order to investigate this hypothesis, we carried out studies in Fancg-/- knock out mice and in human FA fetuses obtained with informed consent from medical abortion. In Fancg-/- mice, FACS analysis revealed a 1,5- to 3-fold deficiency in hematopoietic stem and progenitor cells (HSPC) very early during embryonic development (i.e 11.5 days of gestation - E11.5) in fetal liver (FL) and placenta (Pl) (p <0.001). In both organs, this defect persists during the whole period of amplification (until E14.5 for FL and E12.5 for Pl). In vitro clonogenic assays also demonstrated a 2- fold defect in granulocyte, erythrocyte and macrophage (GEM) progenitors both in Fancg-/- FL or Pl compared to WT (p <0.001), and 4 to 5- fold defect in more immature mixed GEM progenitors in FL (p <0.001). LTC-IC frequency of the HSC-enriched Lineage- Sca1+ AA4.1+ population (LSA) of E14.5 Fancg-/- FL comforted this later result, since it was 5-fold lower than for WT. In vivo long-term hematopoietic reconstitution (LTR) assays confirmed a deficit of the HSC enriched LSA population of E14.5 Fancg-/- FL. Indeed, although the percentage of mice reconstituted was as good as that obtained with the same number of WT LSA, the CD45 Ly5.2 chimerism was reduced (49±20% vs 84±4% for 1000 LSA injected, and 56±12% vs 87±2% for 5000 LSA). Interestingly, bone marrow analysis of mice reconstituted with Fancg-/- LSA 22 weeks after injection showed a level of CD45 Ly5.2 chimerism 3-fold lower than that found in blood, spleen and thymus, as well as a very low chimerism for myeloid GEM lineages, contrasting with a high chimerism for B and T lymphoid lineages. Moreover, we were able to demonstrate that this deficit is already present at E12.5, both in Fancg-/- FL and Pl. Indeed, no mice reconstituted with 3.105 total Fancg-/- fetal liver cells, while 100% injected with the same number of WT FL cells got reconstituted with a chimerism of 59,5±5%. For Pl, when 500 000 cells were injected, reconstitution was observed in only 1 out of 3 mice for Fancg-/- (29% chimerism), and in 3 out of 3 mice for WT (88±4% chimerism). In human FA FL of 14 weeks of gestation, we also observed a 4-fold defect of HSPC with a total lack of in vitro amplification compared to control, in agreement with the mice data. Taken together, these data demonstrate that a profound deficit of HSCs and progenitors cells is present since the earlier stages of embryonic development in FA. In addition, using organotypic cultures of E11 aortas, we could show that this defect of amplification is already present in HSCs emerging from Fancg-/- aorta, which showed a 2-fold lower rate of amplification compared to WT. More importantly, our results show for the first time exhaustion in myeloid lineage of FA, in agreement with what is observed in children with FA disease. Altogether, our work suggests a role of the FA pathway during the development of the hematopoietic system leading to a deficit of amplification of HSC. Comparison of FA HSC transcriptome with that of control HSC in FL and Pl is in progress. It should allow to identify the key pathways involved in the embryonic HSC amplification that are deregulated in FA, and hopefully getting more insights in the pathogenesis of the BMF and leukemogenesis in FA patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 87 (9) ◽  
pp. 3563-3570 ◽  
Author(s):  
AJ Shah ◽  
EM Smogorzewska ◽  
C Hannum ◽  
GM Crooks

Flt3 is a class III tyrosine kinase receptor expressed on primitive human and murine hematopoietic progenitor cells (HPC). In previous studies using stroma-free short term assays, Flt3 ligand (FL) has been shown to induce proliferation of HPC at proportions similar to or less than c-kit ligand (steel factor, SF). Using long term stromal cocultivation assays, we studied the effects of FL on proliferation and differentiation of a highly primitive and cytokine nonresponsive subpopulation of human HPC, CD34+cd38- cells. Cell Proliferation was significantly greater with FL than with SF, when used individually or in combinations with interleukin-3 (IL-3) and/or IL-6. The effect of FL was greater on bone marrow (BM) CD34+CD38- cells than the more cytokine responsive cord blood CD34+CD38- cells. Little or no effect was seen with FL on more mature CD34+CD38+ cells from either BM or cord blood. The frequency of colony-forming units (CFU) and high proliferative potential-colony forming cells (HPP-CFC) during early culture ( < or = 30 days) was increased by both SF and FL to similar levels. However, in the LTC-IC period (35 to 60 days) and extended long-term culture initiating cell (ELTC-IC) period ( > 60 days), the frequency of CFU and HPP-CFC was significantly greater in cultures containing FL than those without FL (P < .0025). Fluorescence-activated cell sorter analysis of cultures after 21 days showed a significantly higher percentage of cells remained CD34+ in the combination of FL, IL-3, IL-6, and SF (F/3/6/S) than in 3/6/S (0.78% +/- 0.52% v 0.21% +/- 0.29% respectively, mean +/- SD). Cloning efficiency of BM CD34+CD38- cells was significantly increased by the addition of FL to the combination of 3/6/S (mean 11.7% v 0.5%, P < .0001). These data show that FL is able to induce proliferation of CD34+CD38-cells that are nonresponsive to other early acting cytokines and to improve the maintenance of progenitors in vitro.


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

Abstract 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.


Sign in / Sign up

Export Citation Format

Share Document