Quantitative Imaging of Femoral Bone Marrow Microenvironments Reveals a Heterogenous Distribution of Hematopoietic Stem and Progenitor Cells.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1455-1455
Author(s):  
Cesar Nombela-Arrieta ◽  
Brendan Harley ◽  
Gregory Pivarnik ◽  
John E Mahoney ◽  
Elena Levantini ◽  
...  

Abstract Abstract 1455 Poster Board I-478 Sustained production of all mature blood cell types relies on the continuous proliferation and differentiation of a rare population of self-renewing, multipotent hematopoietic stem cells (HSCs). HSC maintenance and lineage differentiation are thought to be regulated by spatially confined niches, defined by cellular components, soluble regulators, and the extracellular matrix immediately surrounding stem cells. Identification of these microenvironments in which endogenous and transferred HSCs reside within the BM is a major challenge in stem cell biology with relevant clinical implications. Yet the extreme rarity of HSCs, their dynamic nature, and the lack of specific markers to identify them, have precluded an accurate definition of HSC niches to date. Quantitative imaging technologies such as Laser Scanning Cytometry (LSC) are designed for the automated analysis of large cell numbers at a single cell level with high resolution while preserving the morphological information lost in flow cytometry, therefore providing data of statistical significance even for rare cell populations such as HSCs. We have employed LSC to analyze the localization of both adoptively transferred and endogenous hematopoietic stem and progenitor cell (HSPC) populations inside whole longitudinal sections of murine femoral BM cavities. Our results indicate that, as previously suggested, purified HSPC (Lin−c-kit+Sca-1+) significantly accumulate in endosteal regions (ER) of BM cavities (within 100μm of inner bone surface) upon transplantation. Nevertheless, analysis of sufficient numbers of more differentiated cell subsets (Lin−c-kit+Sca-1− progenitors, pro B cells and mature B cells) indicated that these areas serve as homing sites for most hematopoietic cells, highlighting the limitations of any conclusions drawn on HSC niche identity from studies performed with transferred HSPC populations. Immunofluorescent staining of endogenous cell populations revealed a gradient in distribution of early hematopoietic progenitors (c-kit+), which accumulated in but were not restricted to ER regions. Of note, a vast majority (>80%) of HSPC (Bmi-GFPhic-kit+, or Lin−c-kit+Sca-1+),were found inside ER, although not directly adjacent to endosteal surfaces. Our studies define endosteal areas as tissue regions where HSPC reside in close proximity, but not necessarily in direct contact with a dense vascular network, osteoblastic cells and other potential niche cell types and growth factors currently under investigation. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4231-4231
Author(s):  
Amos S. Gaikwad ◽  
Michael Cubbage ◽  
Tatiana Goltsova ◽  
Christopher Threeton ◽  
Maria Ty ◽  
...  

Abstract Abstract 4231 Cord blood (CB) is a rich source of hematopoietic stem cells (HSC) with long-term repopulating activity necessary for allogeneic stem cell transplantation. CD34+ stem cells are considered sufficient for transplantation, however recent progress in stem cell biology indicates that cells with other surface markers such as CD133 or cells expressing high aldehyde dehydrogenase activity with low side scatter (ALDHhigh/SSClow) or a rare side population (SP) of cells that exclude the Hoechst 33342 vital dye via multi drug transporters have been shown to possess stem cell properties. We characterized CD34+, CD133+, ALDH+ and SP in mononuclear cells (MNC) isolated from human CB. While the SP cell population is rare and detectable in few CB-MNC examined, we found abundant CD34+ and CD133+ cells (1.0+/-0.5 and 0.8+/-0.4 per 100 CD45+ MNC cells, respectively) following the ISHAGE protocol. A distinct ALDH+ cell population (median of 0.26%; range of 0.1 to 0.5%) was also present in all of the CB-MNC analyzed. Over 90% of the ALDH+ cells were also CD34+ and CD133+. The ability of CB-MNC to form colonies in methocult semi-solid media supplemented with cytokines yielded myeloid, lymphoid and erythroid colonies. The clonogenic potential of CB-MNC ranged from 16-48%. We are assessing the colony forming ability of purified stem cell fractions using flow cytometry. The clonogenic efficiency of these individual putative stem cells will be discussed. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 422-422
Author(s):  
Rong Lu ◽  
Lisa Nguyen

Abstract In most organ systems, regeneration is a coordinated effort that involves many stem cells, but little is known about how individual stem cells compensate for the functional deficiencies of other stem cells. Functional coordination between stem cells is critically important during disease progression and treatment when a subset of HSCs fail or become malignant. We hypothesize that individual HSCs heterogeneously compensate for specific deficiencies, as recent work from our group and others suggest that HSCs heterogeneously supply blood. To test this hypothesis, we tracked mouse HSCs in vivo using a single-cell tracking technology that we had previously developed. We found that individual HSCs heterogeneously compensate for the lymphopoiesis deficiencies of other HSCs by increasing individual clonal expansion and altering lineage bias. Clonal expansion refers to the increase in clonal progenies. Lineage bias refers to the preferential production of specific blood cell types. This compensation rescues the overall blood supply and influences blood cell types outside of the deficient lineages in distinct patterns. We identified the molecular regulators and signaling pathways associated with this form of HSC coordination using RNA sequencing. Specifically, the STAT3 pathway and NF-B signalingwere activated, and PTEN signaling was inhibited in HSCs during the compensation process. To investigate the dynamics of HSC coordination, we employed a genetically modified mouse model that expresses simian diphtheria toxin (DT) receptor under the control of the CD11b promoter. Monocytes derived from this mouse line can be ablated upon DT administration. We co-transplanted HSCs derived from normal and the genetically modified mice, then conditionally ablated the monocyte population repeatedly, and tracked the temporal responses of individual normal HSCs. Our time-course analysis revealed that a distinct subset of HSC clones produced rapid and persistent responses to the blood perturbations. These clones had not been highly active in the affected lineages prior to the perturbation. We identified several significant temporal profiles that indicate a remarkable heterogeneity in the responses of HSCs to blood system changes. Together, these data suggest that HSC differentiation is coordinated in a deterministic manner during compensation and is independent of the normal differentiation program. Our findings suggest that stem cells interact with each other and form a coordinated cellular network that is robust enough to withstand minor functional disruptions. Individual HSCs distinctly adapt their differentiation program to compensate for deficient HSCs and specifically overproduce undersupplied cell types. The heterogeneity in the compensation activities of individual HSC clones may be essential for maintaining robustness in blood regeneration and suggests that stem cell coordination is a complex process. A better understanding of the clonal level differences in individual HSCs is critically important for identifying the pathogenesis of blood diseases. Exploiting the innate compensation capacity of stem cell networks may improve the diagnosis and treatment of many diseases. For example, the identification of the molecular regulators and pathways involved in HSC compensation can help develop new therapeutic treatments that enhance the innate compensation capacity of stem cells. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4592-4592
Author(s):  
Anna Machalinska ◽  
Monika Modrzejewska ◽  
Maciej Kotowski ◽  
Magdalena Kucia ◽  
Milosz Kawa ◽  
...  

Abstract Abstract 4592 Retinopathy of prematurity (ROP) is a result of increased pathological neoangiogenesis of retina in preterm infants. The source of cells that are responsible for pathogenesis of ROP is still controversial, and some evidence indicates that they may be bone marrow (BM) derived. BM similarly as other tissues contains a subset of stem cells (SCs) which give rise to non-hematopoietic lineages. These non-hematopoietic stem cells appear heterogeneous and can be mobilized from the BM and other non-hematopoietic niches in response to tissue injury into peripheral blood (PB). Based on this we become interested in SCs populations that circulate in peripheral blood in preterm infants, to see whether their level and lineage commitment plays a role and has a prognostic value for development of ROP. Totally eighty-eight subjects were involved in this study: 29 preterm infants with ROP, 29 preterm infants without ROP, and 30 healthy control full-term infants. We investigated the association between i) different populations of SCs circulating in PB, ii) level of selected growth factors/chemokines regulating SCs migration/mobilization, and iii) the incidence of ROP. PB samples were collected 10 weeks after delivery and analyzed by employing flow cytometry (FACS), epimmunofluorescence staining, real time-PCR (RQ-PCR) and ELISA. We analyzed various cell populations enriched in: i) pluripotent very small embryonic-like stem cells, VSELs (lin-CXCR4+CD45-), ii) hematopoietic stem cells, HSCs (lin-CXCR4+CD45+), and iii) early endothelial progenitor cells, EPCs (CD34+CD133+CD144+). In parallel we measured serum concentration of VEGF, bFGF, HGF, and SDF-1, factors that regulate trafficking of SCs. We found that the number of circulating EPCs and VSELs were significantly increased in PB of the preterm infants (almost 5- and 2-times, respectively). Moreover, the number of EPCs in the PB was significantly higher in the preterm infants with ROP as compared to preterm infants without retinopathy. At the same time we observed in preterm infants with ROP an increase in the serum concentrations of VEGF and HGF. The increased EPCs levels along with elevated levels of VEGF and HGF in preterm infants with ROP suggest that circulating EPCs and pro-angiopoietic factors may play a role in the development and progression of ROP. We conclude that EPCs mobilized into PB contribute to development of ROP in preterm children, and increase in number of these cells correlates with development of this serious complication. In addition, the observed by us increase in number of circulating VSELs in preterm infants suggest that the growth and development of immature tissues and organs may require mobilization and recruitment of pluripotent stem cells that are important in regeneration processes to reestablish proper organ function. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3000-3000 ◽  
Author(s):  
Aleksandr Lazaryan ◽  
Hien Duong ◽  
Lisa Rybicki ◽  
Frederic J. Reu ◽  
Robert Dean ◽  
...  

Abstract Abstract 3000 Autologous stem cell transplantation (ASCT) remains the standard upfront therapy of younger patients with multiple myeloma (MM). Identifying a prognostic threshold amount of mobilized CD34+ hematopoietic stem cells (SC) may become an important modifiable parameter in the era of novel stem cell mobilization strategies. While poor mobilization of CD34+ cells has been shown to cause delays in hematopoietic recovery, the data on long-term clinical outcomes of patients with inferior CD34+ SC collection (‘under mobilizers’) are limited. We analyzed prospectively collected data on 239 adult patients with MM who underwent ASCT at our institution from 01/1996 to 12/2009. Fifty-one patients (21.3%) who collected less than 4 × 106/kg CD34+ SC were classified as ‘under mobilizers’ and were compared to the rest of the study population (n=188) who collected ≥ 4 × 106/kg CD34+ SC. Even though under mobilizers were slightly older (median 59 vs. 54 years, p =0.01), had longer time from diagnosis to ASCT (11 vs. 8 months, p =0.05), and required more days of leukapheresis (5 vs. 3 days, p <0.001), they did not differ from the other group according to the number of prior treatment regimens, mobilization method (only 2 patients received plerixafor), performance status, or disease remission status at transplant (all p >0.2). Median time-to-recovery for both neutrophils (11 vs. 10 days, p <0.001) and platelets (13 vs. 12 days, p <0.001) was delayed among under mobilizers. Under mobilizers had worse relapse-free survival (RFS) (hazard ratio [HR]=1.49, 95% CI, 1.03–2.16, p =0.03) and non-relapse mortality (NRM) (HR=3.59, 95% CI, 1.51–8.56, p <0.01) in multivariable Cox proportional hazards analysis. Even though the association between poor CD34+ SC collection and inferior overall survival did not reach statistical significance (HR=1.42, 95% CI, 0.94–2.16, p =0.1), under mobilizers were found to have significantly higher rates of 100-day post-transplant mortality (p =0.02). We conclude that in the context of ASCT for MM, failure to collect ≥ 4 × 106/kg CD34+ SC is independently associated with worse RFS and NRM. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4755-4755
Author(s):  
John Astle ◽  
Yangfei Xiang ◽  
Anthony Rongvaux ◽  
Carla Weibel ◽  
Henchey Elizabeth ◽  
...  

Abstract De novo generation of HSCs has been described as a "holy grail" of stem cell biology, however the factors required for converting human pluripotent stem cells (PSCs) to true hematopoietic stem cells (HSCs) capable of robust long-term engraftment have yet to be fully characterized. Two groups have shown that injection of PSCs into immunodeficient mice leads to teratomas containing niches producing hematopoietic progenitors capable of long-term engraftment. Once these hematopoietic progenitors and their microenvironments are better characterized, this system could be used as a model to help direct in vitro differentiation of PSCs to HSCs. Toward this end, we have injected human PSCs into immunodeficient mice expressing human rather than mouse M-CSF, IL-3, GM-CSF, and thrombopoietin, as well as both human and mouse versions of the "don't eat me signal" Sirpa (collectively termed MISTRG mice). These cytokines are known to support different aspects of hematopoiesis, and thrombopoietin in particular has been shown to support HSC maintenance, suggesting these mice may provide a better environment for human PSC-derived HSCs than the more traditional mice used for human HSC engraftment. The majority of teratomas developed so far in MISTRG contain human hematopoietic cells, and the CD34+ population isolated from over half of the teratomas contained hematopoietic colony forming cells by colony forming assay. These findings further corroborate this approach as a viable method for studying human PSC to HSC differentiation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1591-1591
Author(s):  
Adam D Wolfe ◽  
Karen Downs

Abstract Abstract 1591 MixL1, a paired-type homeodomain transcription factor, is implicated in pre-hematopoietic commitment of stem cell populations. In poorly-differentiated human lymphoma and leukemia lines, MixL1 is inappropriately over-expressed (Drakos et al., Human Pathol 2007:38:500). When constitutively expressed in mice, MixL1 is sufficient to induce Acute Myeloid Leukemia (Glaser et al., PNAS 2006:103:16460). On the basis of these observations, we hypothesize that MixL1 plays an important role in gating the cellular decision to remain in a poorly differentiated and proliferative phase rather than proceeding to definitive hematopoietic stem cell (HSC) identity. Several years ago, the placenta was shown to be a major site of hematopoiesis (Gekas et al., Dev Cell 2005:8:365; Ottersbach and Dzierzak, Dev Cell 2005:8:377). The placenta is composed of the chorionic disc and the allantois, the latter of which matures into the umbilical component of the placenta. The allantois exhibits definitive hematopoietic potential (Ziegler et al., Development 2006:133:4183; Corbel et al., Dev Biol 2007:301:478), and has recently been demonstrated to contain a core of stem cells referred to as the Allantoic Core Domain, or ACD, where potential placental hematopoietic activity may originate (Downs et al., Dev Dyn 2009:238:532). Our immediate goal is to evaluate whether MixL1 is expressed in the allantois, and to establish its precise spatiotemporal whereabouts with respect to early markers of hematopoietic cells, such as Runx1 (Chen et al., Nature 2009:457:887). Using immunohistochemistry in conjunction with the LacZ/Runx1 reporter mouse (North et al., Development 1999:126:2563), we have demonstrated that MixL1 is strongly expressed in the blood islands of the yolk sac, as well as in a broad, contiguous posterior domain of the embryo that extends to include the ACD stem cell core of the allantois. This domain does not include Runx1, and MixL1 expression temporally precedes that of Runx1 in the allantois. As development proceeds, the MixL1 signal becomes most prominent in putative nascent blood cells budding off from the poorly-described blood vessel common to the allantois, yolk sac and dorsal aortae, which we have called the “Vessel of Confluence” (VOC). Here, fetal blood is shuttled into the umbilical cord to the chorion for exchange with the mother. Shortly thereafter, Runx1 begins to appear within VOC, and is co-expressed with MixL1. These findings provide preliminary evidence that MixL1 is expressed within the allantois, and within nascent blood cells derived from a specific arterial site common to the allantois, yolk sac, and fetus. Moreover, MixL1 expression appears to precede that of Runx1. Thus, MixL1 may identify one of the earliest hematopoietic precursor cell populations thus far known in mammals. Further, these data provide additional evidence that the allantois is a promising model system for the study of definitive hematopoiesis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1173-1173 ◽  
Author(s):  
Lei Sun

Abstract The production of mammalian blood cells is sustained throughout life by the self-renewal and differentiation of hematopoietic stem cells (HSCs). Dysregulation in this system leads to different pathologies including anemia, bone marrow failure and hematopoietic malignancies. The Helix-Loop-Helix transcriptional regulator Id2 plays essential roles in regulating proliferation and cell fate of hematopoietic progenitors; however, its role in regulating HSC development remains largely unknown. To assess the function of Id2 in HSCs, we developed two mouse models, including an Id2 conditional knockout model and an Id2-EYFP model, in which EYFP expression is driven by endogenous Id2 promoter. When we examined HSC function by serial transplantation, we found that mice transplanted with Id2F/F Mx1-Cre+ conditionally deleted bone marrow cells became moribund more rapidly after primary and secondary transplantation, compared to those transplanted with Id2+/F Mx1-Cre+ bone marrow, suggesting that HSC self-renewal is impaired when Id2 is deleted. To further determine if self-renewal and maintenance of HSCs depends on the expression level of Id2, we purified HSCs with different levels of Id2 expression using Id2-EYFP mice to specifically address the role of Id2 in HSCs. First, we confirmed Id2 is highly expressed in HSCs in this model. Second, when HSCs with either low or high levels of Id2-EYFP were transplanted into irradiated mice, cells with high levels of Id2 reconstituted transplanted recipients faster than those with low levels of Id2 at 3 weeks and longer, suggesting that Id2 expression is associated with repopulation advantage. Furthermore, Ki-67 staining showed that HSCs with high levels of Id2 have 15-fold more cells in G2/M phase, and fewer cells in G0. BrdU staining also suggested that there are 5-fold more BrdU+ cells in HSCs with high levels of Id2, indicating that Id2 expression correlates with cell cycle progression in HSCs. In addition, p57 has been reported to be required for quiescence of HSCs. Our preliminary data showed that p57 is downregulated in HSCs with high levels of Id2, and p57 is correspondingly upregulated in Id2-null HSCs. Altogether, our data demonstrate that Id2 is required for the self-renewal and proliferation of HSCs, and suggest a link between Id2 and the transcriptional regulatory networks that regulate the functional hematopoietic system. Since Id2 is also expressed in other adult stem cells including muscle and neuronal stem cells, as well as cancer cells, we believe our results can improve our understanding of stem cell biology and cancer development, and contribute to the identification of novel molecules that may be targeted to eliminate cancer stem cells. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2144-2144
Author(s):  
Simon Raffel ◽  
Jenny Hansson ◽  
Carl Herrmann ◽  
Christoph Lutz ◽  
Eike C. Buss ◽  
...  

Abstract Acute Myeloid Leukemia (AML) is a heterogeneous group of myeloid malignancies. The classification of AML subtypes is based on recurrent genetic abnormalities and typical histopathological features, which impact on the patient’s prognosis. AML is also the model disease for the cancer stem cell model with leukemia stem cells (LSCs) residing at the top of a hierarchical organization. LSCs have self-renewal activity and generate leukemic progeny, which make up the majority of leukemic cells. Because LSCs can be quiescent and reside in specific niches in the bone marrow, rendering them resistant to conventional chemotherapy approaches, they are considered the source of relapse. Hence, further strategies to eradicate LSCs are pivotal to improve patient outcomes of this dismal disease. LSCs present within cell populations can be detected by their capacity to re-initiate the leukemia after xenotransplantation into immuno-compromised mice. However, using current methods, it is neither possible to prospectively isolate pure functional LSCs nor to distinguish them reliably from normal hematopoietic stem cells (HSCs). To define functional LSCs we FACS-sorted primary patient samples of different AML subtypes according to surface-expression of CD34 and CD38 and transplanted each of the resulting four cell populations into conditioned NSG recipients. Several AML samples showed human leukemic engraftment in at least one of the subsets, dissecting LSC-containing and LSC-free subpopulations within the same patient. AML engraftment was mainly observed within the CD34+CD38- fraction, but several cases showed LSC activity also in the CD34+CD38+ fraction or even in the CD34- subsets. As age-matched healthy controls, we collected bone marrow hematopoietic stem and progenitor cells (HSPCs, Lineage -CD34+CD38-) from individuals older than 60 years, undergoing hip replacement surgery. In-depth quantitative multiplex proteomic analysis was performed by employing tandem mass tag (TMT) labeling and high-resolution mass spectrometry. Using this approach, more than 7,000 proteins were quantified from 10 LSC-containing and 8 LSC-free fractions from six individual AML samples of different subtypes. Importantly, our data include many low abundance proteins or others that tend to be difficult to detect by mass spectrometry, such as transcription factors and membrane proteins. We identified up to 1500 differentially expressed proteins between LSC-containing and LSC-free fractions. Most interestingly, differentially expressed proteins also clustered according to AML subtype, indicating subtype-specific proteome differences at the level of AML LSCs. Along the same lines, Gene Ontology and Gene Set Enrichment Analyses showed distinct (in some instances even opposing) differences between AML subtypes. For example, interferon- and integrin-signaling were enriched in LSC-fractions of FLT3-ITD, NPM1-mutated AMLs while reduced in LSC-fractions of FLT3-wt, NPM1-wt AML samples. In summary, our data indicate that also at the level of functionally validated LSC populations, subtype-specific differences in protein expression are remarkably evident. This heterogeneity should be taken into account with respect to the development of targeting strategies for LSCs aiming to improve the clinical outcome of AML patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4279-4279
Author(s):  
Paolo Catarsi ◽  
Francesca Cordero ◽  
Giulio Ferrero ◽  
Marco Beccuti ◽  
Valentina Poletto ◽  
...  

Abstract An important issue in myeloproliferative diseases research is to test the hypothesis that in the neo-angiogenesis, which is observed in the spleen and bone marrow of patients, involves endothelial cells derived from the neoplastic clone. In this study we analyzed the gene expression profile of 138 genes in CD34+ hematopoietic cells from the spleen and peripheral blood of patients with myelofibrosis (MF) and healthy individuals (HI). These genes have been selected among those that characterize the neoangiogenic gene signature or belonging to deregulated expression pathways in MF, and are listed in Table1. By using quantitative reverse transcription-PCR (RT-qPCR), we measured the expression levels of 141 targets and 5 reference genes in CD34+ cells isolated by immunomagnetic method from the spleen and peripheral blood of 4 patients and 3 HI. For calculation of the statistical significance of the differences we used the t-test for unpaired samples. A p<0.05 was considered to be statistically significant. All data analyses were performed with GenEx (version 6.1, MultiD). To characterize the differentially expressed genes obtained in each comparison, the lists of genes were analyzed through several tools, i.e. String, Enrichr and GSEA analysis. The comparison between spleen CD34+ cells and circulating CD34+ cells reveal a group of common genes in both patients and heathy donors. Those genes are significantly overexpressed in spleen. Some of the common genes are involved in the regulation of cell migration and in the development of blood vessels were significantly overexpressed in splenic CD34+ cells compared to circulating CD34+ cells. However, we observed that many of the genes implicated in the development of the vessels were under-expressed in CD34+ cells isolated from the spleen of patients compared to those of HI. The most significant upregulated genes in CD34+ splenic cells from patients included transcripts (GATA1, HBB, TAL1, GATA2, PTGS1) belonging to the molecular signature of CD34+ cells isolated from the bone marrow (BM) of patients with chronic myeloid leukemia (Diaz-Blanco E. et al., Leukemia 2007). Another group of upregulated genes (GATA1, TAL1, ITGB3, GATA2, PF4) are also identified in a study designed to characterize the genes essential to the development of megakaryocytes (Tenedini et al. Blood 2004). These two groups of genes are part of an expression pattern characteristic for immature stem cells as well as megakaryocyte-erythrocyte progenitor cells. Another group of genes overexpressed in patients spleens (CD34, ANGPT1, PF4, GATA2, PTGS1), which includes some of the above mentioned genes, has been observed in a comparison between circulating CD34+ cells from patients with myelofibrosis, and CD34 + cells isolated from the bone marrow of HI (Guglielmelli P. et al. Stem Cells 2007). Among the underexpressed genes in the spleen of patients we mention CXCR4, in keeping with previous observations. Interestingly, among the aforementioned genes, GATA1 and GATA2 genes are overexpressed in granulocytes of patients with MPNs regardless of the JAK2/CALR mutational status whereas PTGS1 is overexpressed in the JAK2-V617F homozygous and CALR-mutated granulocytes (Rampal et al. Blood 2014). We focused our attention on PTGS1 because its well-known activity in regulating angiogenesis, which can be inhibited in vitro by treatment with aspirin (Tsujii, M. et al. Cell 1998). To explain the mechanism of action of low-doses aspirin in this context, it has been proposed a model that involves permanent inactivation of PTGS1 in platelets. Indeed, PTGS1 is the only cyclooxygenase isoenzyme present in platelets. Moreover, Dixon and colleagues (Dixon, D.A. et al. JCI 2006), demonstrated that activated platelets induced the expression of PTGS2 in monocytes. It is believed that the synthesis of prostaglandin E2 by PTGS2 in tissues, is linked to an increment of angiogenesis and cell proliferation, and to a reduction of apoptosis. According to this concept, our data point toward a model in which, in the spleen of patients with MF, an altered hematopoietic stem cell differentiation could induce an inflammation-mediated angiogenesis through the overexpression of PTGS1 and a consequent induction of PTGS2 in cells of the splenic microenvironment. Table Table. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 57-57
Author(s):  
Vincenzo Giambra ◽  
Catherine E Jenkins ◽  
Sonya H Lam ◽  
Catherine Hoofd ◽  
Miriam Belmonte ◽  
...  

Abstract Prior work has shown that NOTCH1 is a prominent oncogene in T-cell acute lymphoblastic leukemia (T-ALL) with activating NOTCH1 mutations occurring in over 50% of cases (Weng et al, Science 2004) and loss-of-function mutations in its negative regulator FBXW7 occurring in 8-15% of cases (O’Neil et al, J Exp Med 2007; Thompson et al, J Exp Med 2007). Subsequent work has shown that continued Notch signaling is required for maintenance of T-ALL leukemia stem cells (Armstrong et al, Blood 2009; Tatarek et al, Blood 2011; Giambra et al, Nat Med 2012). Several lines of evidence have substantiated genetic interactions between the Notch and Wnt signaling pathways in various contexts, and Wnt signaling has been shown to play important roles in hematopoietic stem cell biology and also in hematopoietic cancers such as acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Luis et al, Leukemia 2012). To address what role if any Wnt signaling may play in T-ALL, we generated primary murine leukemias by viral transduction of bone marrow progenitors with activated NOTCH1, then delivered a fluorescent Wnt reporter construct (7TGP; Fuerer & Nusse, PLoS ONE 2010) by lentiviral transduction, and retransplanted the leukemias to interrogate Wnt signaling activity in vivo. We report here that active Wnt signaling is restricted to minor subpopulations within bulk T-ALL tumors, and that these Wnt-active subsets are highly enriched for leukemia-initiating cell (LIC) activity. Moreover, using Ctnnb1loxP/loxP animals we show that inducible Cre-mediated deletion of β-catenin or enforced expression of a dominant-negative TCF construct severely compromises LIC activity. We also show that β-catenin levels are upregulated by hypoxia through Hif1a stabilization, and that deletion of Hif1a also severely compromises LIC activity. Interestingly, Wnt-active subsets are distributed diffusely throughout the marrow interstitial space suggesting that tumor infiltration induces formation of local hypoxic niches as opposed to taking up residence in pre-existing anatomic compartments with low oxygen tensions. Taken together, these results suggest a model in which hypoxic niches in vivo facilitate Hif1a-dependent accumulation of β-catenin which drives Wnt signaling and self-renewal of leukemia stem cells. Finally, we show using patient-derived xenografts that antagonism of Hif1a or Wnt signaling also compromises human LIC activity, suggesting that pharmacologic targeting of these pathways could have therapeutic application in patients with T-ALL. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document