scholarly journals Direct synergistic effects of leukemia inhibitory factor on hematopoietic progenitor cell growth: comparison with other hematopoietins that use the gp130 receptor subunit

Blood ◽  
1996 ◽  
Vol 88 (3) ◽  
pp. 863-869 ◽  
Author(s):  
JR Keller ◽  
JM Gooya ◽  
FW Ruscetti

Because leukemia inhibitory factor (LIF) has little or no effect on murine hematopoietic progenitor cell growth yet enhances hematopoiesis in vivo, we sought to determine whether the effects of LIF were directly or indirectly mediated, or a combination of both. Although LIF alone or in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-3 (IL-3) has no effect on colony formation of unfractionated bone marrow cells (BMCs), it enhances M-CSF- induced colony formation. In comparison, LIF synergizes with IL-3, GM- CSF, M-CSF, and Steel Factor (SLF) to promote the colony formation of partially purified lineage-negative (Lin-) BM progenitors without altering their differentiation. These effects were directly mediated since identical results were observed in single-cell assays. Comparing the effect of LIF with other members of this subclass of hematopoietins (IL-6, oncostatin M [OSM], and ciliary neurotrophic factor [CNTF]), we found that while LIF and IL-6 equally synergize with M-CSF and SLF to promote the colony formation of Lin- BMCs, OSM, and CNTF have no effect. In agreement with OSMs ability to directly bind gp130, preincubation of BMCs with OSM inhibits progenitor cell growth stimulated by the combination of LIF or IL-6 plus SLF. LIF can also directly enhance the growth of further purified more primitive Lin- c- kit+ progenitor cells in the presence of IL-3, GM-CSF, or SLF. Thus, LIF can directly synergize with growth factors to promote the proliferation of purified hematopoietic progenitors, suggesting that the direct effects of LIF on hematopoietic cell growth can, in part, explain the observed hematopoietic effects in vivo. This is a US government work. There are no restrictions on its use.

Blood ◽  
1996 ◽  
Vol 88 (3) ◽  
pp. 863-869 ◽  
Author(s):  
JR Keller ◽  
JM Gooya ◽  
FW Ruscetti

Abstract Because leukemia inhibitory factor (LIF) has little or no effect on murine hematopoietic progenitor cell growth yet enhances hematopoiesis in vivo, we sought to determine whether the effects of LIF were directly or indirectly mediated, or a combination of both. Although LIF alone or in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-3 (IL-3) has no effect on colony formation of unfractionated bone marrow cells (BMCs), it enhances M-CSF- induced colony formation. In comparison, LIF synergizes with IL-3, GM- CSF, M-CSF, and Steel Factor (SLF) to promote the colony formation of partially purified lineage-negative (Lin-) BM progenitors without altering their differentiation. These effects were directly mediated since identical results were observed in single-cell assays. Comparing the effect of LIF with other members of this subclass of hematopoietins (IL-6, oncostatin M [OSM], and ciliary neurotrophic factor [CNTF]), we found that while LIF and IL-6 equally synergize with M-CSF and SLF to promote the colony formation of Lin- BMCs, OSM, and CNTF have no effect. In agreement with OSMs ability to directly bind gp130, preincubation of BMCs with OSM inhibits progenitor cell growth stimulated by the combination of LIF or IL-6 plus SLF. LIF can also directly enhance the growth of further purified more primitive Lin- c- kit+ progenitor cells in the presence of IL-3, GM-CSF, or SLF. Thus, LIF can directly synergize with growth factors to promote the proliferation of purified hematopoietic progenitors, suggesting that the direct effects of LIF on hematopoietic cell growth can, in part, explain the observed hematopoietic effects in vivo. This is a US government work. There are no restrictions on its use.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3602-3602
Author(s):  
Hal E. Broxmeyer ◽  
Jonathan Hoggatt ◽  
Scott Cooper ◽  
Giao Hangoc ◽  
Louis M. Pelus ◽  
...  

Abstract Abstract 3602 Poster Board III-539 CD26 is a dipeptidylpeptidase IV (DPPIV) that cleaves dipeptides from the N-terminus after a proline or alanine. An amino acid sequence search identified putative CD26/DPPIV truncation sites in a number of colony stimulating factors (CSFs), including human (hu) and mouse (mu) GM-CSF and G-CSF, hu IL-3, and hu and mu EPO. These truncation sites were not apparent in mu IL-3, hu and mu M-CSF, or in hu and mu stem cell factor (SCF) or Flt3-ligand (FL). We hypothesized that CD26/DPPIV served as a regulator of the potency of the selected CSFs that contain this putative truncation site, and that hematopoietic recovery after stress would be enhanced and accelerated in CD26 −/− mice. We first used Diprotin A (Ile-Pro-Ile), a known CD26/DPPIV inhibitor for mu and hu cells. Mu cytokines were assessed for activity on mu BM, and hu cytokines on hu cord blood (CB), all in dose-response fashion. Hu EPO was tested on mu and hu cells. One hour pre-treatment of mu BM cells with Diprotin A, or use of CD26 −/− mu BM cells, resulted in a two-fold or greater enhancement of CFU-GM-, CFU-G-, and BFU-E- colony formation of cells respectively stimulated by mu GM-CSF, mu G-CSF, and hu EPO. The CSF activities of mu M-CSF for CFU-M, and mu IL-3 for CFU-GM were not enhanced by inhibition/deletion of CD26/DPPIV. Also, pretreatment of hu CB cells with Diprotin A, enhanced colony formation of CFU-GM stimulated by hu GM-CSF or hu IL-3, and of BFU-E stimulated by hu EPO, but did not influence stimulation of CFU-G or CFU-M by hu M-CSF. Stimulation of cells with two CSFs results in additive to greater than additive hematopoietic progenitor cell (HPC) colony formation compared to that of each CSF alone. When both CSFs had putative CD26/DPPIV truncation sites, colony formation by HPC was even further increased by pretreatment of target cells with Diprotin A. Pretreatment of cells with Diprotin A did not enhance colony formation of mu BM or hu CB cells each respectively stimulated with SCF or FL alone, nor did it enhance the synergistic effects noted when SCF or FL was used in combination with CSFs. To assess truncation and activity directly, CSFs were pretreated with purified soluble DPPIV. Truncation was detected by Mass Spectrometry for CSFs with the putative truncation site, but not for those without this site. Truncated CSFs manifested greatly reduced activity against target cells, effects that were more apparent when the cells were pretreated with Diprotin A. Moreover, mixture of a truncated CSF with a non-truncated form of that CSF reduced colony formation to the level of the truncated CSF, suggesting that the truncated CSF interfered with or blocked activity of the full-length CSF. To evaluate effects of CD26/DPPIV on hematopoietic recovery, CD26 −/− and +/+ mice were treated with sublethal dosages of irradiation, or either cycle-specific or non-cycle specific drugs. In all cases, enhanced and accelerated recovery of hematopoietic progenitor cells was noted in the CD26 −/− mice, compared to the control CD26 +/+ mice. These results demonstrate that CD26/DPPIV regulates the activity of selected CSFs, and inhibition/deletion of CD26/DPPIV allows for enhanced in vitro potency of selected CSFs, and in vivo recovery of hematopoiesis in mice stressed with irradiation and cytotoxic drugs. These results may have practical relevance for understanding, and manipulating hematopoietic recovery after cytotoxic treatment or hematopoietic stem cell transplantation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1347-1347
Author(s):  
Hal E. Broxmeyer ◽  
Nico Ghilardi ◽  
Giao Hangoc ◽  
Scott Cooper ◽  
Wen Tao ◽  
...  

Abstract Interleukin (IL)-31 receptor (R), also called gp130-like monocyte-receptor (GLM-R; Ghilardi et al. J. Biol. Chem.277:16831, 2002) is related to gp130 (~25% homology), and G-CSF-R (~24%). Its signaling activates STAT3 and STAT5. IL-31 is a four helix bundle cytokine preferentially produced by T helper 2 cells. Nothing is known of the possible hematopoietic effects of IL-31R and IL-31. However, since: IL-31 signals through a receptor composed of IL-31RA and Oncostatin M R (Dillon et al. Nature Immunol. 5:752,2004), Oncostatin M is implicated in homeostasis of myeloid progenitor cells (Broxmeyer et al. Immunity.16:815, 2002), and STAT3 and 5 are implicated by a number of groups in cytokine regulation of hematopoiesis, we hypothesized that the IL-31/IL-31R axis would be involved in regulation of hematopoiesis. We first compared myeloid progenitor cell (MPC: CFU-GM, BFU-E, and CFU-GEMM) numbers and cycling status in marrow and spleen of IL-31R −/− vs. littermate control mice (+/+) using a combination of Epo, SCF and PWMSCM to stimulate in vitro the cells taken from these mice. IL-31R −/− mice had significantly decreased numbers of MPC per femur (~51%) and spleen (~36–45%) with significantly decreased MPC cycling status in marrow (% MPC in S-phase: 0–3% in IL-31R −/− vs. 41–53% in +/+ mice). MPC in spleen of IL-31 −/− and +/+ were both in a slow or non cycling state (0–3% in S-phase). In contrast to CFU-GM from +/+ mice, CFU-GM from IL-31R −/− mice demonstrated little or no synergistic response to combined stimulation in vitro with GM-CSF or IL-3 with either SCF or Flt3-ligand (FL). This translated to decreased absolute numbers per femur of GM-CSF+SCF-, GM-CSF+FL-, IL-3+SCF-, and IL-3+FL- responsive CFU-GM in IL-31R −/− mice. However, there were no significant differences between GM-CSF- or IL-3- responsive CFU-GM per femur between IL-31R −/− vs. +/+ mice suggesting effects on immature subsets of MPC. Recombinant IL-31 was assessed for effects in vitro. IL-31, at concentrations up to 100ng/ml, did not stimulate colony formation by marrow MPC, nor did it enhance or suppress colony formation stimulated by GM-CSF, Epo, Epo+SCF, or Epo+SCF+GM-CSF. However, IL-31 did enhance survival of MPC subjected to delayed growth factor addition in a manner similar to, but not as potent as, that of SDF-1/CXCL12. IL-31 manifested no chemotaxis activity for +/+ MPC, and IL-31R −/− and +/+ MPC were equally responsive to the chemotactic effects of SDF-1/CXCL12. These results suggest that the IL-31R in vivo acts to positively regulate numbers and cycling of immature subsets of MPC in the marrow, and that IL-31 has survival enhancing effects on MPC in vitro.


Blood ◽  
1992 ◽  
Vol 80 (6) ◽  
pp. 1443-1447
Author(s):  
WJ Murphy ◽  
G Tsarfaty ◽  
DL Longo

Recombinant human growth hormone (rhGH) was administered to mice to determine its effect on hematopoiesis. BALB/c mice and mice with severe combined immune deficiency (SCID), which lack T cells and B cells, were administered intraperitoneal injections of rhGH for 7 days. Upon analysis, both strains of mice exhibited an increase in splenic and bone marrow hematopoietic progenitor cell content and cellularity, indicating that rhGH can act as a hematopoietic growth factor. C57BL/6 mice were then placed on azidothymidine (AZT). AZT is a reverse transcriptase inhibitor currently used as a treatment for acquired immune deficiency syndrome (AIDS), but which also produces significant myelotoxic effects. Treatment of mice with rhGH partially counteracted the myelosuppressive properties of AZT. Bone marrow cellularity, hematocrit values, white blood cell counts, and splenic hematopoietic progenitor cell content were all significantly increased if rhGH (20 micrograms injected intraperitoneally every other day) was concurrently administered with AZT. Administration of ovine GH (ovGH), which, unlike rhGH, has no effect on murine prolactin receptors, also prevented the erythroid-suppressive effects of AZT in mice, but had no significant effect on granulocyte counts. Thus, the effects of GH are mediated at least in part through GH receptors in vivo. Additionally, when mice were initially myelosuppressed by several weeks of AZT treatment, the subsequent administration of ovGH resulted in an increase in splenic hematopoietic progenitor cells. No significant pathologic effects were observed in mice receiving either repeated rhGH or ovGH injections. Thus, GH exerts significant direct hematopoietic growth-promoting effects in vivo and may be of potential clinical use to promote hematopoiesis in the face of myelotoxic therapy.


Blood ◽  
1997 ◽  
Vol 90 (9) ◽  
pp. 3296-3303 ◽  
Author(s):  
Kai-Ling Fu ◽  
Jerome R. Lo Ten Foe ◽  
Hans Joenje ◽  
Kathleen W. Rao ◽  
Johnson M. Liu ◽  
...  

Abstract Fanconi anemia (FA) is an autosomal recessive genetic disorder characterized by a variety of physical anomalies, bone marrow failure, and an increased risk for malignancy. FA cells exhibit chromosomal instability and are hypersensitive to DNA cross-linking agents such as mitomycin C (MMC). FA is a clinically heterogeneous disorder and can be functionally divided into at least five different complementation groups (A-E). We previously described the use of a retroviral vector expressing the FAC cDNA in the complementation of mutant hematopoietic cells from FA-C patients. This vector is currently being tested in a clinical trial of ex vivo hematopoietic progenitor cell transduction. The FA-A group accounts for over 65% of all FA cases, and the FAA cDNA was recently identified by both expression and positional cloning techniques. We report here the transduction and phenotypic correction of lymphoblastoid cell lines from four unrelated FA-A patients, using two amphotropic FAA retroviral vectors. Expression of the FAA transgene was adequate to normalize cell growth, cell-cycle kinetics, and chromosomal breakage in the presence of MMC. We then analyzed the effect of retroviral vector transduction on hematopoietic progenitor cell growth. After FAA transduction of mutant progenitor cells, either colony number or colony size increased in the presence of MMC. In addition, FAA but not FAC retroviral transduction markedly improved colony growth of progenitor cells derived from an unclassified FA patient. FAA retroviral vectors should be useful for both complementation studies and clinical trials of gene transduction.


2000 ◽  
Vol 18 (9) ◽  
pp. 1824-1830 ◽  
Author(s):  
Omer N. Koç ◽  
Stanton L. Gerson ◽  
Brenda W. Cooper ◽  
Mary Laughlin ◽  
Howard Meyerson ◽  
...  

PURPOSE: Patient response to hematopoietic progenitor-cell mobilizing regimens seems to vary considerably, making comparison between regimens difficult. To eliminate this inter-patient variability, we designed a cross-over trial and prospectively compared the number of progenitors mobilized into blood after granulocyte-macrophage colony-stimulating factor (GM-CSF) days 1 to 12 plus granulocyte colony-stimulating factor (G-CSF) days 7 to 12 (regimen G) with the number of progenitors after cyclophosphamide plus G-CSF days 3 to 14 (regimen C) in the same patient. PATIENTS AND METHODS: Twenty-nine patients were randomized to receive either regimen G or C first (G1 and C1, respectively) and underwent two leukaphereses. After a washout period, patients were then crossed over to the alternate regimen (C2 and G2, respectively) and underwent two additional leukaphereses. The hematopoietic progenitor-cell content of each collection was determined. In addition, toxicity and charges were tracked. RESULTS: Regimen C (n = 50) resulted in mobilization of more CD34+ cells (2.7-fold/kg/apheresis), erythroid burst-forming units (1.8-fold/kg/apheresis), and colony-forming units–granulocyte-macrophage (2.2-fold/kg/apheresis) compared with regimen G given to the same patients (n = 46; paired t test, P < .01 for all comparisons). Compared with regimen G, regimen C resulted in better mobilization, whether it was given first (P = .025) or second (P = .02). The ability to achieve a target collection of ≥ 2 × 106 CD34+ cells/kg using two leukaphereses was 50% after G1 and 90% after C1. Three of the seven patients in whom mobilization was poor after G1 had ≥ 2 × 106 CD34+ cells/kg with two leukaphereses after C2. In contrast, when regimen G was given second (G2), seven out of 10 patients failed to achieve the target CD34+ cell dose despite adequate collections after C1. Thirty percent of the patients (nine of 29) given regimen C were admitted to the hospital because of neutropenic fever for a median duration of 4 days (range, 2 to 10 days). The higher cost of regimen C was balanced by higher CD34+ cell yield, resulting in equivalent charges based on cost per CD34+ cell collected. CONCLUSION: We report the first clinical trial that used a cross-over design showing that high-dose cyclophosphamide plus G-CSF results in mobilization of more progenitors then GM-CSF plus G-CSF when tested in the same patient regardless of sequence of administration, although the regimen is associated with greater morbidity. Patients who fail to achieve adequate mobilization after regimen G can be treated with regimen C as an effective salvage regimen, whereas patients who fail regimen C are unlikely to benefit from subsequent treatment with regimen G. The cross-over design allowed detection of significant differences between regimens in a small cohort of patients and should be considered in design of future comparisons of mobilization regimens.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1206-1206 ◽  
Author(s):  
Heather A. O'Leary ◽  
Charlie Mantel ◽  
Xianyin Lai ◽  
Scott Cooper ◽  
Giao Hangoc ◽  
...  

Abstract DPP4 (CD26) is a dipeptidyl peptidase that functions by enzymatically cleaving the penultimate proline, alanine or select other amino acids such as serine of proteins, resulting in functional alterations of the protein. We recently published that many cytokines, chemokines and growth factors have putative DPP4 truncations sites and that DPP4 specifically was able to truncate some colony stimulating factors such as GM-CSF and IL-3 with resultant blunting of their activity. However, the mechanism of action of the truncated factors is still unknown and requires further investigation. The expression, and activity, of DPP4 is relevant in normal and malignant hematopoiesis as we have data showing that CD34+ umbilical cord blood cells (UCB) as well as Acute Myelogenous Leukemia (AML) patient samples express active DPP4. Further, specific inhibition of DPP4 increases homing and engraftment of both human UCB and mouse bone marrow cells after transplantation in mice indicating the therapeutic potential of DPP4 activity altering compounds. Due to its potential importance in disease states, and their subsequent treatment, it is relevant to study how the activity of DPP4 alters the functions of the molecules it cleaves, and subsequently their interactions with each other. DPP4 can cleave the penultimate proline of GM-CSF and IL-3 resulting in truncated forms which have blunted colony stimulating factor activity for hematopoietic progenitor cells (HPC). Since GM-CSF and IL-3 receptors share a common receptor beta chain, we investigated if DPP4 truncation of GM-CSF (TGM) or IL-3 (T3) could inhibit the receptor binding and functional activity of the full length (FL) alternate compound (i.e TGM inhibition of FL3 activity or T3 inhibition of FLGM activity) in the factor dependent TF-1 cell line, UCB cells and in in vivo mouse studies. We determined using TF-1 and UCB that both T3 and TGM bound to the receptors with higher affinity than their FL forms and could blunt the receptor binding of the FLGM and FL3. Additionally, TGM and T3 decreased colony formation induced by either FLGM or FL3 in both TF-1, UCB, and primary AML patient cell samples. Strikingly, this inhibition of colony formation did not require a 1:1 ratio of the full length to truncated forms of these cytokines. Rather, approximately 4-10 fold less truncated molecules could be used to efficiently inhibit the colony formation activity of the full length form, even across molecules. In vivo injection of FL, T, or a mixture of FL/T or T/T factors into DPP4 activity knockout mice followed by colony assays showed the TGM and T3 suppresed the effect of FLGM or FL3 on progenitor cell numbers per femur and diminished cycling of hematopoietic progenitor cells as detected by high specificity tritiated thymidine kill assay. Proteomic analysis of the effects of full length and truncated factors (FLGM, FL3, TGM, T3) were performed with TF-1 cells where we detected differential protein regulation by the full length vs truncated factors. After 24 hour treatment with 10ng/ml of FLGM or TGM, TF-1 cells displayed statistically significant (p < .05) differences in 26 proteins of which 17 were higher in the FL vs the T, and 9 higher in the T vs FL treated groups. These proteins included, but were not limited to, cell cycle proteins such as CDK6, HDAC6, as well as signal transduction proteins and redox control proteins such as STAM1 and Glutaredoxin. Additionally, alterations in protein phosphphorylation were detected for TF-1 cells treated for 15 or 30 min with the full length vs truncated IL-3 and GM-CSF proteins. Interestingly, the protein expression or phosphorylation levels were not always decreased by the truncated protein compared to the full length. In some cases, the truncated molecules induced an increase in the protein expression or phosphorylation. These data suggest interesting roles for full length and truncated GM-CSF and IL-3 in both normal and malignant hematopoiesis. Further investigation into the regulation of DPP4, and the roles that full length and truncated factors play during normal and malignant hematopoiesis, are important and will allow for a better understanding of the signficance of DPP4 activity during steady state, stressed, and disease hematopoiesis. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document