scholarly journals Acute Myeloid Leukemia Induced Remodelling of the Human Bone Marrow Niche Predicts Clinical Outcome

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3851-3851
Author(s):  
Yiyang Chen ◽  
Yasmin Zaun ◽  
Lucas Arnold ◽  
Kerstin Weissenfels ◽  
Kurt Werner Schmid ◽  
...  

Abstract The hematopoietic stem cell (HSC) niche consists of different cellular and non-cellular constituents which regulate HSC maintenance and retention in the bone marrow. It has been shown in a number of murine models of myeloid neoplasia how leukemia infiltration alters the HSC niche to reinforce malignancy. Acute myeloid leukemia (AML) is characterized in human by a high relapse rate indicating that leukemia initiating cells are protected by its niche. However, despite our knowledge in murine models little is known about the bone marrow architecture in human and the impact of the leukemic niche on clinical outcome. In this study, we combined immunohistochemical stainings (IHC) with protein and global gene expression analyses together with clinical data to dissect the human bone marrow architecture in AML and assess its clinical impact. Human bone marrow was collected from AML patients at first diagnosis and matching non-leukemic donors. To evaluate the bone marrow architecture CD271+ mesenchymal stem and progenitor cells (MSPCs) were automatically quantified on bone marrow sections. In fact, AML patients showed 1.5-fold increase in bone marrow MSPCs compared to non-leukemic donors (Median (IQR), AML: 5.5% (2.8-9.5), n=36; control: 3.7% (2.1-5.7), n=58; p < 0.01). MSPCs proved to produce reticular fibers, an extracellular matrix protein frequently associated with different malignancies. In AML bone marrow these fibers were also found to be more abundantly expressed (Median (IQR), AML: 3.4% (1.8-4.5), n=37; control: 1.6% (1.1-3.3), n=19; p < 0.05). Next, to globally assess the gene expression profile of MSPCs in AML bone marrow we performed microarray analyses (ClariomTM S Human Assay) of freshly isolated uncultured lineage- CD146+ CD271+ MSPCs. Strikingly, HSC-regulating genes in particular CXCL12, ANGPT1 and VCAM1 showed lower expression in AML MSPCs which correlated with the degree of hematopoietic failure in AML patients. Along with the increased number of MSPCs, geneset enrichment analysis (GSEA) revealed higher proliferation of MSPCs in AML. In murine models loss of quiescence of MSPCs was previously found to be due to bone marrow sympathetic neuropathy. We therefore measured catecholamines and neurotrophic factor in the bone marrow extracellular fluid of AML patients and non-leukemic donors at first diagnosis. In fact, noradrenalin and brain-derived neurotrophic factor (BDNF) showed a 2-fold (p=0.26) resp. 4-fold (p<0.0001) lower expression in AML bone marrow. Importantly, BDNF is proved to be essential for sympathetic neuron proliferation and differentiation. In order to get an overview of alterations of canonical pathways in bone marrow MSPCs upon AML infiltration, we applied QIAGEN's Ingenuity® Pathway Analysis software. Several of the major differently regulated pathways proved to involve differentiation and mineralization of MSPCs. We therefore assessed bone metabolism in AML patients at first diagnosis and quantified serum osteocalcin levels. Notably, AML patients showed 30% lower osteocalcin levels than non-leukemic donors (Median (IQR), AML, 12.15ng/ml (7.53-16.28) n=58; control, 17.2ng/ml (12.5-23.45) n=31; p < 0.05). To evaluate if the deficiency in osteoblast mineralization is specifically due to AML infiltration we performed in vitro co-culture assays. Both MSPCs and an osteoblast-like cell line (SaOS2) showed significant impaired mineralization in presence of certain AML cell lines as well as primary human AML cells, while healthy mononuclear cells did not affect mineralization. Strikingly, this AML-induced defect in osteoblast mineralization proved to be of clinical significance. Patients with low osteocalcin levels (<11ng/ml) showed inferior overall survival with 1-year survival rate of 38.7% while patients with high osteocalcin levels reached 66.8% (n=58; median duration of follow-up 9.7 months). In summary, we globally characterized the bone marrow architecture in AML patients in comparison to non-leukemic donors and assessed its clinical significance. This increasing understanding of the human AML bone marrow microenvironment might open the window for new niche-targeted therapies to eradicate leukemic stem cells and eventually decrease the high relapse rate in AML. Disclosures Duehrsen: Amgen: Research Funding; Roche: Honoraria, Research Funding; Gilead: Consultancy, Honoraria; Celgene: Honoraria, Research Funding; AbbVie: Consultancy, Honoraria; Janssen: Honoraria.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3850-3850
Author(s):  
Xingbing Wang ◽  
Qiansong Cheng ◽  
Jian Wang ◽  
Liang Xia ◽  
Xuhan Zhang ◽  
...  

Abstract Abstract 3850 Human bone marrow-derived mesenchymal stem cells (BM-MSCs) are multipotent nonhematopoietic progenitor cells, which can differentiate into osteoblasts, adipocytes, chondrocytes and other tissues. The most important function of BM-MSCs is to support hematopoiesis. Toll-like receptors (TLRs) are a conserved family of receptors that can be activated by both pathogen components and mammalian endogenous molecules such as heat-shock proteins and extracellular matrix breakdown products. In the past a few year, several studies reported that TLRs are expressed in hematopoietic and non-hematopoietic to modulate their biological functions. We hypothesized that MSCs are equipped with TLRs that enable them to dynamically change hematopoiesis-related cytokines expression pattern and level by sensing correspondent agonists, thus efficiently supporting hematopoiesis. In this study, BM-MSCs were analyzed for mRNA expression of TLR 1–9 by reverse transcription-polymerase chain reaction. TLR 1–6, but not TLR 7–9 were expressed by MSCs. The expression of TLR2 and TLR4 was also confirmed by flow cytometic assay. We further explored the role of TLR2 and TLR4 in mediating the capacity of MSC to support the proliferation and differentiation of CD34+ cells. The pre-stimulation with TLR2 agonists (Pam3Cys) or TLR4 agonists (LPS) enable MSCs to enhance CD34+ cells proliferation and promote CD34+ cells differentiation towards the myeloid lineage (CD33+, CD11b+), as well as granulocyte colony formation by those cells. The production of interleukin 8 (IL-8), IL-11, stem cell factor (SCF), granulocyte colony-stimulating factor (CSF), macrophage CSF and granulocyte-macrophage CSF were also increased by stimulated MSCs. Interestingly, although Pam3Cys and LPS displayed different inductive magnitudes, they have no synergistic effect on MSCs. We hypothesized there may be some antagonistic effect between TLR2 and TLR4 intracellular signal conductive pathway, or they can downregulate the expressive level of the TLRs on MSCs. Together, our findings suggest that TLR2 and TLR4 signalings may indirectly regulate hematopoiesis by modulating MSCs' functions. The increased haemopoietic proliferation and myeloid lineage differentiation could be mediated, at least in part, by augmented hematopoiesis-related cytokine production. Disclosures: Wang: National Natural Science Foundation (30700329): Research Funding; Anhui Provincial Outstanding Young Investigator Program (08040106810): Research Funding; Fund of Anhui Provincial “115” Industrial Innovation Program: Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2402-2402
Author(s):  
Willy A Noort ◽  
Regina de Jong-Korlaar ◽  
Linda Lubbers-Aalders ◽  
Huipin Yuan ◽  
Joost D de Bruijn ◽  
...  

Abstract Previously, we have reported that our human bone marrow (BM)-like scaffold xenograft model allows the engraftment and outgrowth of normal and malignant hematopoiesis (e.g. multiple myeloma (MM), acute myelocytic/lymphocytic leukemia (AML/ALL) and MDS (Groen et al. Blood 2012; Gutierrez et al. JCI 2014 and data not shown). Whereas the presence of osteoblasts and bone of human origin mimics a human BM-like niche more closely than the murine BM in standard xenotransplant models (e.g. NOD-SCID/NSG mice), still some essential components of the human BM niche, i.e. human blood vessels, are missing. To this end, in addition to human mesenchymal stromal cells we now incorporated cord blood-derived endothelial progenitor cells (CB-EPCs) in the hybrid scaffold production process, to create a multi-tissue compartment that "maximally humanizes" the BM-like niche of our scaffolds. Towards successful implementation of a human vascular system we compared: i) scaffold material composition (biphasic calcium phosphate (BCP) vs. tricalcium phosphate (TCP)); ii) scaffold shape (particles vs. tubes); iii) different types of matrigel for CB-EPC embedding. Histological analysis of the humanized scaffolds, eight weeks after implantation in mice, showed a large number of functional human blood vessels, as indicated by hCD31+ staining and the presence of erythrocytes within. Comparison of the composition and the shapes of the scaffolds indicated superiority of TCP and tube-shaped scaffolds in supporting the formation of vessels. Further analysis of scaffolds for CD44, CD146, LEPR and nestin-positive cells, revealed the presence of other stromal niche cells besides human osteoblasts and endothelial cells. Irradiation of mice carrying these humanized implants did not have a significant deleterious effect on the established human vessels, allowing their further functional evaluation in xenotransplantation. Additionally, mice carrying tubes with and without human CB-EPC derived vessels (on either flank) were subsequently inoculated with adult BM-derived CD34-positive cells by intracardiac injection. Upon analysis 12 weeks later, all tubes showed multi-lineage hematopoietic outgrowth. Interestingly, CB-EPC embedment resulted in increased numbers of CD45+ (2-fold), CD13+ (4-fold) and CD7+ (2-fold), while CD19+ cell numbers were equal. In contrast, in mouse BM almost only CD19+ cells could be detected. Moreover, we observed that the use of CB-EPCs in our scaffolds provides faster kinetics of in vivo engraftment and growth of both patient-derived MM or AML cells. With the addition of both human CB-EPCs and human BM stromal cells, our scaffold systems now simulate both human endosteal and vascular niches of the BM, thereby more closely recapitulating the human hematopoietic niche. Disclosures Yuan: Xpand Biotechnology BV: Employment. de Bruijn:Xpand Biotechnology BV: Employment. Mitsiades:TEVA: Research Funding; Janssen/Johnson & Johnson: Research Funding; Novartis: Research Funding. Martens:Johnson & Johnson: Research Funding. Groen:Johnson & Johnson: Research Funding.


Blood ◽  
1973 ◽  
Vol 42 (3) ◽  
pp. 331-339 ◽  
Author(s):  
Uri Mintz ◽  
Leo Sachs

Abstract Normal serum and serums from patients with acute and chronic leukemia were assayed for granulocyte colony-inducing activity with human bone marrow cells. Serum from untreated acute leukemia, but not from the other patients, showed about normal inducing activity at low serum concentration and lower than normal activity at high concentration. This suggests that serum from patients with acute leukemia contained an inhibitor for colony formation. Serums from chronic myeloid leukemia were in about the same range as normal, whereas serums from chronic lymphocytic leukemia showed the highest colony-inducing activity.


2017 ◽  
Author(s):  
D Pal ◽  
H Blair ◽  
S Boyd ◽  
P Bakelis ◽  
A Elder ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document