Parathyroid Hormone-Induced Modulation of the Bone Marrow Microenvironment Reduces Leukemic Stem Cells in Murine Chronic Myelogenous-Leukemia-Like Disease Via a TGFbeta-Dependent Pathway

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1670-1670
Author(s):  
Daniela S. Krause ◽  
Keertik Fulzele ◽  
Andre Catic ◽  
Michael Hurley ◽  
Sanon Lezeau ◽  
...  

Abstract Abstract 1670 It is known that osteoblastic cells regulate the normal hematopoietic stem cell (HSC) niche and control its size. Parathyroid hormone (PTH) is an important regulator of osteoblasts and osteoclasts maintaining calcium homeostasis and, additionally, increasing HSC number in transplant recipients and protecting HSCs from repeated exposure to cytotoxic chemotherapy. We, therefore, hypothesized, that PTH-treatment may allow normal HSCs to outcompete leukemic stem cells (LSCs) in a murine model of chronic myelogenous leukemia. Mice with osteoblastic cell-specific constitutive activation of the receptor for PTH and PTH-related protein (Col1-caPPR mice) are characterized by activation of osteoblastic cells and increases in osteoclast and osteoblast number, trabecular bone, bone turnover and cortical porosity. Col1-caPPR mice have significantly prolonged survival and reduced leukemic mortality compared to wildtype (wt) littermates in a murine retroviral transduction/transplantation model of BCR-ABL1-induced CML-like disease (p=0.002) and B-cell acute lymphoblastic leukemia (B-ALL) (p=0.0004). However, a leukemogenic fusion transcription factor, MLL-AF9, known to cause acute myeloid leukemia in this model, led to more rapid death in the Col1-caPPR recipients compared with their wt counterparts (p<0.0001), indicating that the increased survival of Col1-caPPR recipients is specific for BCR-ABL1-induced leukemia. Continuous infusion of human PTH(1–34) into wt mice with BCR-ABL1-induced CML led to a statistically significant decrease in spleen weights and decreased bone marrow infiltration by BCR-ABL+ cells. Limiting dilution secondary transplantation of BM cells from saline- or PTH-treated primary animals with fully established CML into wt recipients revealed a 15-fold reduction of LSCs in a PTH-treated environment. Secondary mice who received BM from saline-treated donors had an overall survival that was 1/4 that of recipients of marrow from a PTH-treated BM microenvironment. Transforming growth factor beta-1 (TGFβ-1), whose largest and most concentrated tissue source is bone, was increased in the bones of Col1-caPPR mice. TGFβ-1 significantly decreased the in-vitro growth of the BCR-ABL+ cell line K562, but not the MLL-AF9+ cell line THP-1 suggesting that TGFβ-1, increased in the bone marrow microenvironment of Col1-caPPR mice, may be actively suppressing the growth of the BCR-ABL+ diseases, but not of MLL-AF9+ AML. Conversely, blockade of TGFβ-1, -2, and -3 by anti- TGFβ antibody treatment increased the incidence of CML in Col1-caPPR mice from 50% to 75%. Knockdown of TGF Receptor I in transplanted BCR-ABL+ BM in the CML model increased the percentage of BCR-ABL+ myeloid cells in peripheral blood in wt and, more strikingly, in Col1-caPPR recipient mice and increased the overall incidence of CML in Col1-caPPR mice. These results argue that reduction in TGFβ-1 signaling may rescue the CML phenotype in Col1-caPPR mice. In conclusion, these studies suggest that modulation of the bone marrow microenvironment by PTH reduces the frequency of LSCs in CML, possibly by suppression of LSCs via TGFβ-1. Consequently, a clinical trial on the combined use of imatinib and PTH in patients with CML has been initiated at our institution. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 191-191
Author(s):  
Amitava Sengupta ◽  
Jose Cancelas

Abstract Chronic myelogenous leukemia (CML) is a stem cell malignancy induced by p210 BCR-ABL and characterized by myeloproliferation in BM and egression of leukemic stem cells and progenitors (LSC/P) to extramedullary sites. Persistence of BCR-ABL+ HSC in patients under Imatinib suggests inhibition of ABL-kinase alone is not sufficient to eliminate the LSC/P. One of the major hallmarks of CML induced by signaling downstream BCR-ABL is the loss of control of the hematopoietic microenvironment on LSC/P. Expression of p210 BCR-ABL has been associated with loss of adhesion to the bone marrow, impaired migration in response to CXCL12 and decreased retention in the BM. In order to study the putative LSC/P niches in steady-state chronic-phase leukemia, we have analyzed the ability of LSC/P to proliferate and get retained in the bone marrow (BM) in an inducible model of CML. Binary transgenic SCL-tTA/TRE-BCR-ABL mice (Koschmieder S et al., Blood 2005) express p210 BCR-ABL in LSC/P upon doxycycline withdrawal (CML mice). Induced myeloproliferation was associated with activation of the downstream signaling effectors CrkL and p38-MAPK and expansion of circulating (Table 1) and splenic LSC/P but not in BM, suggesting massive LSC/P egression from the marrow (Table 2). Proliferation analysis showed that myeloid expansion in the spleen was secondary to increased cycling of Lin−Sca1+c-Kit+ (LSK) cells (3.1-fold increase in S-phase cells, P&lt;0.05), but not in Lin−/c-Kit+ (LK) population, compared with the control spleens. In agreement with the LSC/P BM content data, the frequency of BM-derived LSK and LK cells incorporating BrdU in CML and in control mice remained similar, suggesting a specific egression of LSC/P from the BM to extramedullary sites. To test whether this model truly represented a model of BM LSC/P egression, we compared the splenic and BM LSC/P compared with their controls regarding their adhesion molecule expression, interaction with the hematopoietic microenvironment (HM) and homing to the overall marrow cavity and endosteal space. Splenic, but not BM-derived, LSK and LSK CD34+ ST-HSCs had increased cell surface expression of CD44 compared to controls (1.35 fold, P=0.006 and 1.23 fold, P&lt;0.05 respectively) and decreased expression of L-selectin (8.7 fold, P&lt;0.05) while expression of CXCR4, α4β1 and α5β1 integrins remain similar in bone marrow and splenocytes from CML and control mice. CML BM progenitors also showed 18-fold reduced adhesion to fibronectin and 1.4-fold increased migration towards CXCL12 compared to control BM progenitors. Myeloproliferative disease was transplantable into non-transgenic littermates and homing of CML BM progenitors was increased (4.3 fold, P&lt;0.005) in myeloablated littermate recipient BM. However, lineage-negative leukemic BM-derived cells which had increased homing in BM of recipient mice had an impaired ability to migrate to the BM endosteal space compared with their littermate controls (control: 31 ± 18% vs CML mice: 17.6 ± 17%), suggesting an specific impairment to lodge in specialized anatomically-defined hematopoietic “niches”. Altogether, this murine model may represent an adequate in vivo system to analyze the ability of p210 BCR-ABL-expressing LSC/P to interact with BM niches and study the control of the hematopoietic microenvironment on LSC/P survival, proliferation and retention. Table 1 Increase in circulating LSC/P in the CML mice after withdrawal of doxycyclin Peripheral Blood LSK (×103)Cells/mL Blood P&lt;0.05 LT-HSC(×103)Cells/mL Blood P&lt;0.05 CFU-GM+BFU-E/mL Blood P&lt;0.05 Control 1.56 ± 0.25 0.459 ± 0.29 60.86 ± 51.09 CML mice 3.56 ± 1.52 2.159 ± 2.03 869.6 ± 628.4 Table 2. Immunophenotypic analysis of BM and splenocytes in control and CML mice Population BM (Cells ×104) (Control) BM (Cells x104) (CML) SP (Cells ×104) (Control) SP (Cells x104) (CML) C-Kit + Sca1 + 24.3 ± 9.9 21.3 ± 11 6.8 ± 4.5 30.1 ± 12.3 (P&lt;0.05) Mac1 + Gr1 + 1779 ± 307 1583 ± 265 78.4 ± 32 608 ± 377 (P&lt;0.05) CFU-C/10 5 Cells 342 ± 66 334 ± 99 63.3 ± 7.09 79 ± 6.54 (P&lt;0.05)


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 937-937
Author(s):  
Daniela S. Krause ◽  
Sanon Lezeau ◽  
Michael Hurley ◽  
Ernestina Schipani ◽  
David T. Scadden

Abstract Abstract 937 The role of the bone marrow microenvironment, and in particular of osteoblastic cells, for normal hematopoiesis has recently been described. However, the role of elements of the bone marrow microenvironment on the initiation, maintenance or progression of leukemia is less clear. To test the influence of activated osteoblasts on the progression of chronic myelogenous leukemia (CML) we used the well-described murine retroviral transduction/transplantation model of BCR-ABL1-induced CML-like disease and mice with osteoblastic cell-specific constitutive activation of the parathyroid-hormone (PTH) receptor (PPR mice) as recipients. Compared to wildtype (wt) littermate control mice PPR mice had significantly prolonged survival (p=0.002) and reduced leukemic mortality with splenic leukemopoiesis contributing to leukemic fatality. Analysis of distinct proviral integration sites in splenic tissue by Southern blotting showed no difference in engraftment of viral clones in wt versus PPR mice. Survival of PPR recipients in the BCR-ABL1-induced model of B-cell acute lymphoblastic leukemia using non-5-fluorouracil-treated donor bone marrow was also significantly prolonged compared to wt mice (p=0.0004). However, a leukemogenic allele known to result in acute myeloid leukemia (AML), MLL-AF9, led to more rapid death in the PPR recipients compared with their wt counterparts arguing that the prolongation of survival in the BCR-ABL1-induced diseases was oncogene-specific. In-vitro assays including the Whitlock-Witte assay to test the role of PPR stroma or a cobblestone colony formation assay in osteogenic medium revealed no difference in the growth of plated BCR-ABL1+ or BCR-ABL1- B-lymphoid progenitors or lin- c-kit+ Sca-1+ cells on wt versus PPR stroma, respectively. In vivo, treatment of mice with the phosphodiesterase inhibitor forskolin, which increases intracellular cyclic adenosine monophosphate (cAMP) levels, similar to increased signaling from the PTH receptor, did not lead to prolonged survival in the murine model of CML-like disease. Prior splenectomy of wt, as well as PPR recipients of BCR-ABL1-induced CML-like disease did not significantly prolong survival, but drastically reduced the efficiency of induction of CML-like disease with the great majority of wt and PPR animals succumbing to non-CML causes. Secondary transplantation of CML-like disease from bone marrow or spleen from a wt or PPR microenvironment revealed, firstly, less efficient induction of secondary disease in wt recipients of PPR bone marrow compared to wt bone marrow and, secondly, superiority of CML-induction in secondary recipients of spleen compared to bone marrow from a PPR microenvironment. In order to test for an osteoblast-extrinsic cause of the prolonged survival of PPR recipients in the CML-model and to test for the role of bone remodeling, wt and PPR recipients of BCR-ABL1-transduced bone marrow were treated with saline or osteoprotegerin, an inhibitor of osteoclast differentiation and proliferation. Surprisingly, 100% of PPR and wt control mice succumbed to CML-like disease sooner than the control mice treated with saline. Additionally, continuous infusion of human PTH(1-34) into wt mice with BCR-ABL1-induced CML-like disease by minipump led to prolonged survival compared to saline-treated animals arguing that PTH may be an intervention beneficial in human CML. This, to our knowledge, represents first evidence that modulation of the bone marrow microenvironment may improve the outcome in leukemia. Disclosures: Scadden: Fate Therapeutics: Consultancy, Equity Ownership, Patents & Royalties.


Blood ◽  
1992 ◽  
Vol 79 (4) ◽  
pp. 997-1002 ◽  
Author(s):  
D Claxton ◽  
A Deisseroth ◽  
M Talpaz ◽  
C Reading ◽  
H Kantarjian ◽  
...  

Interferon (IFN) therapy of early chronic myelogenous leukemia (CML) frequently produces partial or complete cytogenetic remission of the disease. Patients with complete cytogenetic remission often continue on therapy for several years with bone marrow showing only diploid (normal) metaphases. We studied hematopoiesis in five female patients with major cytogenetic remissions from CML during IFN therapy. Clonality analysis using the BstXI PGK gene polymorphism showed that granulocytes were nonclonal in all patients during cytogenetic remission. BCR region studies showed rearrangement only in the one patient whose remission was incomplete at the time of sampling. Granulopoiesis is nonclonal in IFN-induced remissions of CML and may be derived from normal hematopoietic stem cells.


Haematologica ◽  
2020 ◽  
Vol 106 (1) ◽  
pp. 111-122 ◽  
Author(s):  
Sandrine Jeanpierre ◽  
Kawtar Arizkane ◽  
Supat Thongjuea ◽  
Elodie Grockowiak ◽  
Kevin Geistlich ◽  
...  

Chronic myelogenous leukemia arises from the transformation of hematopoietic stem cells by the BCR-ABL oncogene. Though transformed cells are predominantly BCR-ABL-dependent and sensitive to tyrosine kinase inhibitor treatment, some BMPR1B+ leukemic stem cells are treatment-insensitive and rely, among others, on the bone morphogenetic protein (BMP) pathway for their survival via a BMP4 autocrine loop. Here, we further studied the involvement of BMP signaling in favoring residual leukemic stem cell persistence in the bone marrow of patients having achieved remission under treatment. We demonstrate by single-cell RNA-Seq analysis that a sub-fraction of surviving BMPR1B+ leukemic stem cells are co-enriched in BMP signaling, quiescence and stem cell signatures, without modulation of the canonical BMP target genes, but enrichment in actors of the Jak2/Stat3 signaling pathway. Indeed, based on a new model of persisting CD34+CD38- leukemic stem cells, we show that BMPR1B+ cells display co-activated Smad1/5/8 and Stat3 pathways. Interestingly, we reveal that only the BMPR1B+ cells adhering to stromal cells display a quiescent status. Surprisingly, this quiescence is induced by treatment, while non-adherent BMPR1B+ cells treated with tyrosine kinase inhibitors continued to proliferate. The subsequent targeting of BMPR1B and Jak2 pathways decreased quiescent leukemic stem cells by promoting their cell cycle re-entry and differentiation. Moreover, while Jak2-inhibitors alone increased BMP4 production by mesenchymal cells, the addition of the newly described BMPR1B inhibitor (E6201) impaired BMP4-mediated production by stromal cells. Altogether, our data demonstrate that targeting both BMPR1B and Jak2/Stat3 efficiently impacts persisting and dormant leukemic stem cells hidden in their bone marrow microenvironment.


2014 ◽  
Vol 42 (8) ◽  
pp. S68
Author(s):  
Maiko Matsushita ◽  
Miki Nakamura ◽  
Daiju Ichikawa ◽  
Nobuo Tsukamoto ◽  
Yutaka Kawakami ◽  
...  

Blood ◽  
1992 ◽  
Vol 79 (4) ◽  
pp. 997-1002 ◽  
Author(s):  
D Claxton ◽  
A Deisseroth ◽  
M Talpaz ◽  
C Reading ◽  
H Kantarjian ◽  
...  

Abstract Interferon (IFN) therapy of early chronic myelogenous leukemia (CML) frequently produces partial or complete cytogenetic remission of the disease. Patients with complete cytogenetic remission often continue on therapy for several years with bone marrow showing only diploid (normal) metaphases. We studied hematopoiesis in five female patients with major cytogenetic remissions from CML during IFN therapy. Clonality analysis using the BstXI PGK gene polymorphism showed that granulocytes were nonclonal in all patients during cytogenetic remission. BCR region studies showed rearrangement only in the one patient whose remission was incomplete at the time of sampling. Granulopoiesis is nonclonal in IFN-induced remissions of CML and may be derived from normal hematopoietic stem cells.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4328-4328
Author(s):  
Alieta Ciocea ◽  
Natalya V. Narizhneva ◽  
Evrim Erdogan ◽  
Tatiana V. Byzova ◽  
Michael Kalafatis

Abstract Megakaryocytes are polyploid cells, originating from hematopoietic stem cells in the bone marrow. Megakaryocytopoiesis is the process of production of anucleated cells, the platelets, from megakaryocytes. Megakaryocytes undergo endomitosis and maturation to the stage of proplatelets bearing megakaryocytes, which fragment to give rise to platelets. Platelets are vital for both maintaining normal hemostasis and for the response of the human body to trauma. Apoptosis is a genetically programmed and evolutionary conserved mechanism through which the normal development and tissue homeostasis are maintained. The platelets production is the result of constitutive apoptosis of megakaryocytes. Casein kinase II (CKII) is a pleiotropic, ubiquitous ectokinase that phosphorylates Serine, Threonine and Tyrosine amino acid residues. CKII has two catalytic subunits, α and α’ and two regulatory β subunits. The upregulation and hyperactivity of CKII has a general anti-apoptotic, pro-survival function in different overproliferative and carcinogenic processes. In chronic myelogenous leukemia (CML), specifically, CKII upregulation and hyperactivity induces stem cells increased proliferation. This results in bone marrow and circulating high blast levels and abnormal blood cells counts (trombocytosis, leukocytosis). We used the megakaryoblastic CML cell line, MEG-01, to study the effect of the inhibition of CKII, by CKII α subunit specific inhibitors, on platelet formation and cell proliferation. MEG-01 cells, which are defined as cytokine independent, are unresponsive to thrombopoietin and interferon. Incubation of MEG-01 cells with two different CKII inhibitors results in proliferation arrest, megakaryopoiesis, apoptosis and megakaryocytopoiesis. Platelet release from MEG-01 cells strongly depends on inhibitors concentration and treatment length. The platelets obtained from MEG-01 cells, following incubation with CKII inhibitors, get activated when stimulated by agonists, like phorbol-12-mystrate-13-acetate (PMA) and the peptide TRAP. These agonist-activated platelets undergo shape change and expose P-Selectin as a consequence of their activation. The platelet population stains positive for GpIIb/IIIa complex. Small size and negative staining for propidium iodide clearly distinguish platelets from megakaryocytes. In conclusion, we found a novel activator of platelet release from megakaryocytes. PMA, used as a megakaryocytopoiesis inducer, is known to stimulate release of functional platelets from MEG-01 cells, with normal blood platelet morphology. However, PMA, a very potent tumor promoter, induces cutaneous squamous cell carcinoma in mice. Our findings are important and demonstrate that platelets release from MEG-01 cells is a form of apoptosis that can be initiated following specific inhibition of CKII α subunit. Therefore, the processes of megakaryocytopoiesis and megakaryopoiesis are controllable through the fine-tuning of apoptosis. These results are original and of high physiological relevance since the high blast cell percentage can be decreased by proliferation arrest and differentiation. So the malignant processes in this type of leukemia can be stopped by controlling the stem cell number and their proliferation rate following inhibition of CKII α subunit. Thus, regulation of CKII α catalytic subunit may play a very important role in the future treatment of patients with CML that are resistant to the currently available treatments.


Sign in / Sign up

Export Citation Format

Share Document