scholarly journals Inflammatory signals from fatty bone marrow supports the early stages of DNMT3a driven clonal hematopoiesis

2022 ◽  
Author(s):  
Naama Zioni ◽  
Akihad Bercovich ◽  
Noa Chapal-Ilani ◽  
Aryeh Solomon ◽  
Ekaterina Petrovich ◽  
...  

Age related cancer is not only due to the random accumulation of mutations, but also how phenotypes are selected by the aging environment. While fatty bone marrow (FBM), is one of the hallmarks of bone marrow ageing, it is unknown whether FBM can modify the evolution of the early stages of leukemia and clonal hematopoiesis (CH). To address this question, we established FBM mice models and transplanted both human and mice preleukemic hematopoietic stem cells (PreL-HSCs) carrying DNMT3A mutations. We demonstrate that castration which models age related andropenia result in FBM. A significant increase in self-renewal was found when DNMT3AMut-preL-HSPCs were exposed to FBM. To better understand the mechanisms of the FBM-preL-HSPCs interaction, we performed single cell RNA-sequencing on HSPCs three days after FBM exposure. A 20-50 fold increase in DNMT3AMut-preL-HSCs was observed under FBM conditions in comparison to other conditions. PreL-HSPCs exposed to FBM exhibited an activated inflammatory signaling (IL-6 and INFγ). Cytokine analysis of BM fluid demonstrated increased IL-6 levels under FBM conditions. Anti-IL-6 neutralizing antibodies significantly reduced the selective advantage of DNMT3AMut-preL-HSPCs exposed to FBM. Overall, age related paracrine FBM inflammatory signals promote DNMT3A-driven clonal hematopoiesis, which can be inhibited by blocking the IL-6 receptor.

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 596-596
Author(s):  
Naama Zioni ◽  
Noa Chapal Ilani ◽  
Ekaterina Petrovich-Kopitman ◽  
Mehmet Saçma ◽  
Hartmut Geiger ◽  
...  

Abstract With aging, humans accumulate preleukemic mutations (pLMs) in hematopoietic stem and progenitor cells (HSPCs) which was termed age-related clonal hematopoiesis (ARCH). To gain a better insight changes in the HSPCs-environment crosstalk upon aging that might contribute to ARCH, it is critical to develop a multilayer perspective that integrates information on mutations, epigenetics the cellular context and the bone marrow (BM) microenvironment, since all these layers are changing during ageing. Accordingly, a key question in the field is how the ageing BM microenvironment influences clonal expansion of HSPCs. Fatty bone marrow (FBM) is one of the environmental factors that may influence clonal hematopoiesis (CH) with age. As we age, our bone marrow shifts from red to adipocyte-enriched yellow BM. We hypothesize that age related BMF accumulation may provide a selective advantage to specific pre-leukemic stem and progenitor cells (preL-HSPCs) carrying pLM. To support this hypothesis, we established a FBM model in NSG mice to enable the study of both human and rodent preL-HSPCs. Transplantation of primary human preL-HSPCs from AML patients (DNMT3A, NPM1 mutations ) into FBM resulted in enhanced engraftment compared to control mice without FBM. We further demonstrate that DNMT3A-R882H+/- mice derived BM HSPCs, engrafted significantly higher in NSG mice with FBM compared to controls. Interestingly, when DNMT3A-R882H+/- derived BM cells from middle-aged mice (12-month old) were injected into FBM mice, engraftment increased tenfold. Secondary engraftment of aged DNMT3A-R882H +/-BM derived cells resulted in an increase in engraftment upon transplantation into to FBM, suggesting enhanced in vivo self-renewal capacity of HSPCs in FBM. To study the underlying molecular mechanisms provided by the FBM to preL-HSPCs carrying DNMT3A-R882H +/-, we used a multiplex cytokine assay. In this approach we analyzed 17 common cytokines in BM following transplantation of young, two-month old, or middle-aged, 12-month old, DNMT3A-R882H +/-or control - BM derived cells into FBM. Our results show that transplanting two months old, middle-aged DNMT3A-R882H +/-or control BM derived cells to FBM resulted in a significant increase in BM mIL-6 secretion when compared to transplants into control, non-FBM mice. mIL-6 was secreted by adipocytes following irradiation regardless of which cells are transplanted. We then transplanted middle-aged DNMT3A-R882H+/- BM derived cells to FBM mice that had been treated intraperitoneally with a neutralizing IL-6 Ab. The administration of neutralizing IL-6 Ab resulted in a significant decrease in engraftment of DNMT3A-R882H+/- BM derived cells, confirming that IL6 contributes to the expansion of the DNMT3a-R882H+/- cells in FBM. In summary, these results demonstrate for the first time that the FBM provides a selective advantage to pre-leukemic cells carrying DNMT3A-R882H. Importantly, we show that IL-6 is a one of the major players in the molecular mechanism that confers the FBM advantage specifically to preL-HSPCs carrying R882H both in vitro and in vivo. Disclosures Mueller-Tidow: Janssen Cilag: Consultancy, Research Funding; Bioline: Consultancy, Research Funding; Pfizer: Consultancy, Research Funding.


Author(s):  
Oriol Pich ◽  
Iker Reyes-Salazar ◽  
Abel Gonzalez-Perez ◽  
Nuria Lopez-Bigas

AbstractMutations in genes that confer a selective advantage to hematopoietic stem cells (HSCs) in certain conditions drive clonal hematopoiesis (CH). While some CH drivers have been identified experimentally or through epidemiological studies, the compendium of all genes able to drive CH upon mutations in HSCs is far from complete. We propose that identifying signals of positive selection in blood somatic mutations may be an effective way to identify CH driver genes, similarly as done to identify cancer genes. Using a reverse somatic variant calling approach, we repurposed whole-genome and whole-exome blood/tumor paired samples of more than 12,000 donors from two large cancer genomics cohorts to identify blood somatic mutations. The application of IntOGen, a robust driver discovery pipeline, to blood somatic mutations across both cohorts, and more than 24,000 targeted sequenced samples yielded a list of close to 70 genes with signals of positive selection in CH, available at http://www.intogen.org/ch. This approach recovers all known CH genes, and discovers novel candidates. Generating this compendium is an essential step to understand the molecular mechanisms of CH and to accurately detect individuals with CH to ascertain their risk to develop related diseases.


2020 ◽  
Vol 12 (526) ◽  
pp. eaax6249 ◽  
Author(s):  
Wing Hing Wong ◽  
Sima Bhatt ◽  
Kathryn Trinkaus ◽  
Iskra Pusic ◽  
Kevin Elliott ◽  
...  

Clonal hematopoiesis is associated with various age-related morbidities. Error-corrected sequencing (ECS) of human blood samples, with a limit of detection of ≥0.0001, has demonstrated that nearly every healthy individual >50 years old harbors rare hematopoietic clones below the detection limit of standard high-throughput sequencing. If these rare mutations confer survival or proliferation advantages, then the clone(s) could expand after a selective pressure such as chemotherapy, radiotherapy, or chronic immunosuppression. Given these observations and the lack of quantitative data regarding clonal hematopoiesis in adolescents and young adults, who are more likely to serve as unrelated hematopoietic stem cell donors, we completed this pilot study to determine whether younger adults harbored hematopoietic clones with pathogenic mutations, how often those clones were transferred to recipients, and what happened to these clones over time after transplantation. We performed ECS on 125 blood and marrow samples from 25 matched unrelated donors and recipients. Clonal mutations, with a median variant allele frequency of 0.00247, were found in 11 donors (44%; median, 36 years old). Of the mutated clones, 84.2% of mutations were predicted to be molecularly pathogenic and 100% engrafted in recipients. Recipients also demonstrated de novo clonal expansion within the first 100 days after hematopoietic stem cell transplant (HSCT). Given this pilot demonstration that rare, pathogenic clonal mutations are far more prevalent in younger adults than previously appreciated, and they engraft in recipients and persist over time, larger studies with longer follow-up are necessary to correlate clonal engraftment with post-HSCT morbidity.


Hematology ◽  
2019 ◽  
Vol 2019 (1) ◽  
pp. 294-302 ◽  
Author(s):  
Nils B. Leimkühler ◽  
Rebekka K. Schneider

Abstract Self-renewing hematopoietic stem cells and their progeny, lineage-specific downstream progenitors, maintain steady-state hematopoiesis in the bone marrow (BM). Accumulating evidence over the last few years indicates that not only primitive hematopoietic stem and progenitor cells (HSPCs), but also cells defining the microenvironment of the BM (BM niche), sense hematopoietic stress signals. They respond by directing and orchestrating hematopoiesis via not only cell-intrinsic but also cell-extrinsic mechanisms. Inflammation has many beneficial roles by activating the immune system in tissue repair and as a defense mechanism. However, chronic inflammation can have detrimental effects by stressing HSPCs, leading to cell (DNA) damage resulting in BM failure or even to leukemia. Emerging data have demonstrated that the BM microenvironment plays a significant role in the pathogenesis of hematopoietic malignancies, in particular, through disrupted inflammatory signaling, specifically in niche (microenvironmental) cells. Clonal selection in the context of microenvironmental alterations can occur in the context of toxic insults (eg, chemotherapy), not only aging but also inflammation. In this review, we summarize mechanisms that lead to an inflammatory BM microenvironment and discuss how this affects normal hematopoiesis. We pay particular attention to the process of aging, which is known to involve low-grade inflammation and is also associated with age-related clonal hematopoiesis and potentially malignant transformation.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1213-1213
Author(s):  
Yuko Kawano ◽  
Daniel K Byun ◽  
Benjamin J. Frisch ◽  
Hiroki Kawano ◽  
Mark W LaMere ◽  
...  

Radiotherapy is used in the treatment of ~50% of tumors. We and others have reported long-term suppression of hematopoietic stem and progenitor cells (HSPCs) in the setting of total body irradiation; however, it has been shown that even relatively small irradiation volumes can result in systemic adverse events, such as myeloablation and secondary malignancies. The mechanisms underlying these effects are unclear. We hypothesize that localized radiation may activate a systemic inflammatory response that can acutely alter HSPCs and bone marrow microenvironment (BMME) components, including marrow stromal cells (MSCs), thereby contributing to late effects. We therefore established a murine model of targeted irradiation (TR) using a small animal radiation research platform (SARRP). Methods: We administered local irradiation to a single tibia of 6-8 week old C57BL/6 male mice using a single dose of 15 Gy. Subsequently, we analyzed peripheral blood, BM, BM extracellular fluid (BMEF), collagenase-1 digested bone associated cells of both the irradiated (TR) and non-irradiated, contralateral (CONT) tibiae at 2, 6, 48 hours, 1 and 3 weeks post-TR, performing phenotypic (flow cytometry) and cytokine analyses. For all studies, n = 10-13 mice/time point. Results: In the TR tibia at 2 hours, although total cell numbers were unchanged, there was a significant upregulation of inflammatory cytokines (interleukin 1β (IL1b), IL18), chemokines (CXCL2, CXCL10, CCL2, CCL3) and macrophage colony stimulating factor (M-CSF). Of note, most of these changes normalized by 48 hours (M-CSF at 1 week). Changes in mediator expression were followed, at 6 hours post-TR, by significant increases in macrophage (macs) numbers, including CD206 phagocytic macs, neutrophils (PMNs) and cytotoxic lymphocytes, including CD8+ cells expressing CXCR3+, the receptor for CXCL9 and CXCL10. Interestingly, similar to the TR tibia, CXCL2 expression was also increased significantly in the CONT at 2 hours, followed (6 hours) by significant increases in macs and CD8+ cells, suggesting a systemic or abscopal effect. With respect to the effects of radiation on HSPCs, by 6 hours, most of the stem and progenitor cell (HSPC) populations in the TR marrow were significantly decreased; the decrease in long-term-HSCs was delayed until 48 hours post-TR. All populations remained severely depleted until 3 weeks post RT, demonstrating a rapid and sustained effect of TR on all HSPCs within the irradiation volume. In comparison, in the CONT tibia at 6 hours, CD41+ HSCs were expanded; this is consistent with previous demonstrations that CD41+ LT-HSCs expand with inflammatory signals and suggests that TR-induced signals induced a systemic impact on the non-irradiated HSPCs. By 1 week post-radiation, short term-HSCs were significantly decreased in the CONT marrow, likely due to mobilization since CFU-Cs were correspondingly significantly increased in the circulation. Finally, MSCs, previously shown to support HSCs, were found to be significantly increased in the TR tibia starting at 6 hours and peaking at 48 hours post-radiation. Surprisingly, MSCs were also expanded in the CONT marrow at 48 hours; this expansion was likely associated with the increased CXCL12 levels seen in both TR and CONT marrow, although the CXCL12 levels were higher in the irradiated tibia. Taken together, these changes indicate TR-induced global disruption of the HSC niche. Furthermore, in addition to the transient effects of localized irradiation, we observed a second wave of inflammatory signals, including a significant increase in CCL3, at 1 week post-TR and increased IL1b in the CONT marrow at 3 weeks, changes that may have contributed to the sustained loss of HSPC populations. Conclusions: We present the effects of local irradiation on global hematopoiesis, showing that, in addition to the anticipated acute local changes in the irradiated bone marrow, TR-induced persistent and, more importantly, systemic inflammation. We believe that using this murine model will allow us to dissect the contribution of direct (local) and indirect (systemic) responses to radiation on treatment effects, such as marrow failure and secondary malignancies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2308-2308
Author(s):  
Martin F. Ryser ◽  
Joachim Roesler ◽  
Harry L. Malech ◽  
Angela Roesen-Wolff ◽  
Sebastian Brenner

Abstract Hematopoietic stem cell (HSC) egress to the circulation and homing to the bone marrow (BM) are regulated by interactions between CXCR4 and SDF-1. Serum albumin is the major protein component of plasma. Concentration gradients of albumin between plasma and interstitial fluid account for the colloid osmotic pressure, which is a central regulator of the intravasal blood volume. SDF-1/CXCR4 dependent migration of mobilized peripheral blood CD34+ hematopoietic stem cells (PBSC) was studied in transwell migration assays. We compared the effect of RPMI diluted Heparin-plasma (50% plasma, 50% RPMI) versus undiluted plasma as a migration medium. To our surprise the use of undiluted plasma in the upper chamber containing the cells and diluted plasma in the lower chamber containing 100ng/ml SDF-1, resulted in a 3 fold increase of migrating PBSCs compared to experiments without a gradient (undiluted versus undiluted plasma or diluted versus diluted plasma) while diluted plasma in the upper chamber and undiluted in the lower chamber inhibited the SDF-1 dependent migration 3,5 fold. To further characterize this observation we removed high molecular weight proteins (>50kd) by filtration of the plasma. The filtrate was used to dilute plasma to obtain samples with reduced protein contents. Transwell experiments showed that negative gradients of plasma proteins (high concentration in the upper chamber vs low concentration in the lower chamber) stimulate (up to 2 fold) while positive gradients inhibit SDF-1/CXCR4 dependent migration of PBSC (up to 3 fold). Migration experiments were repeated with RPMI, supplemented with varying concentrations of serum albumin as migration medium. Negative gradients of albumin stimulated migration while positive gradients were inhibiting. Interestingly, a gradient of 4% albumin in the upper chamber and 1% albumin in the lower chamber enabled migration in the absence of a SDF-1 gradient (100ng/ml SDF-1 in both upper and lower chamber). Albumin gradients did not stimulate migration in the absence of SDF-1. The absence of serum albumin in both chambers abolished the SDF-1/CXCR4 dependent migration of PBSCs. Our results show that gradients of serum albumin strongly influence the SDF-1/CXCR4 dependent migration of PBSCs. Negative gradients of albumin between blood and bone marrow might be supportive for the homing of PBSC to the stem cell niche, which suggests a new function of the multitask protein serum albumin.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4040-4040
Author(s):  
Szabolcs Fatrai ◽  
Simon M.G.J. Daenen ◽  
Edo Vellenga ◽  
Jan J. Schuringa

Abstract Mucin1 (Muc1) is a membrane glycoprotein which is expressed on most of the normal secretory epithelial cells as well as on hematopoietic cells. It is involved in migration, adhesion and intracellular signalling. Muc1 can be cleaved close to the membrane-proximal region, resulting in an intracellular Muc1 that can associate with or activate various signalling pathway components such as b-catenin, p53 and HIF1a. Based on these properties, Muc1 expression was analysed in human hematopoietic stem/progenitor cells. Muc1 mRNA expression was highest in the immature CD34+/CD38− cells and was reduced upon maturation towards the progenitor stage. Cord blood (CB) CD34+ cells were sorted into Muc1+ and Muc1− populations followed by CFC and LTC-IC assays and these experiments revealed that the stem and progenitor cells reside predominantly in the CD34+/Muc1+ fraction. Importantly, we observed strongly increased Muc1 expression in the CD34+ subfraction of AML mononuclear cells. These results tempted us to further study the role of Muc1 overexpression in human CD34+ stem/progenitor cells. Full-length Muc1 (Muc1F) and a Muc1 isoform with a deleted extracellular domain (DTR) were stably expressed in CB CD34+ cells using a retroviral approach. Upon coculture with MS5 bone marrow stromal cells, a two-fold increase in expansion of suspension cells was observed in both Muc1F and DTR cultures. In line with these results, we observed an increase in progenitor counts in the Muc1F and DTR group as determined by CFC assays in methylcellulose. Upon replating of CFC cultures, Muc1F and DTR were giving rise to secondary colonies in contrast to empty vector control groups, indicating that self-renewal was imposed on progenitors by expression of Muc1. A 3-fold and 2-fold increase in stem cell frequencies was observed in the DTR and Muc1F groups, respectively, as determined by LTC-IC assays. To determine whether the above mentioned phenotypes in MS5 co-cultures were stroma-dependent, we expanded Muc1F and DTR-transduced cells in cytokine-driven liquid cultures. However, no proliferative advantage or increase in CFC frequencies was observed suggesting that Muc1 requires bone marrow stromal cells. In conclusion, our data indicate that HSCs as well as AML cells are enriched for Muc1 expression, and that overexpression of Muc1 in CB cells is sufficient to increase both progenitor and stem cell frequencies.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3444-3444
Author(s):  
Luanne L. Peters ◽  
Shirng-wern Tsaih ◽  
Rong Yuan

Abstract Anemia of aging is now recognized as a significant medical problem. The National Health and Nutrition Examination Survey (NHANES III) revealed a steady increase in anemia in both males and females after the age of 50. Based upon the WHO definition of anemia (<13 g/dL hemoglobin (Hgb) in men; <12 g/dL in women), ~10% of the community dwelling population ≥ 65 years of age are anemic. Underlying causes fall into three broad groups, each representing ~1/3 of cases: nutritional deficits/blood loss; inflammation, kidney disease and myelodysplasia; and unexplained anemia. Although anemia of aging is usually mild, it is no longer considered a normal part of aging. It is associated with poor health and increased vulnerability to adverse outcomes in a multitude of circumstances, placing an enormous burden on the healthcare system that will only grow as the population continues to age. As part of The Jackson Laboratory Aging Center (http://agingmice.jax.org/), we are performing an extensive phenotypic analysis of multiple traits related to aging in 32 inbred mouse strains. All data are, or will be upon completion, publicly available via the Mouse Phenome Database (MPD, www.jax.org/phenome). Complete blood counts were obtained at 6, 12, 18, and 24 months of age in 30 strains. Two-way ANOVA reveals that both strain and age significantly impact Hgb in mice. A highly significant strain-by-age interaction is also seen. Substantial inter-strain and within strain sex variability in the decline in Hgb levels with age is seen among the strains analyzed, suggesting genetic influences. Significant declines in Hgb levels in females at 18 and/or 24 months vs. 6 months occurred in 21 of the 30 strains and, in males, 17 strains. Haplotype association mapping (HAM) using a dense SNP panel identified multiple distinct, age-related loci influencing Hgb levels. For example, a locus on chromosome (Chr) 13 significantly associated with Hgb levels at 12 months of age in males was not detected even at the suggestive level at 18 months of age where two new highly significant loci emerged (Chrs 14, 17). Only two strains show a statistically significant increase in percent circulating reticulocytes with age, indicative of a proliferative anemia. Failure of a significant reticulocyte response in all other strains suggests that an age-related compromise in bone marrow function (hematopoiesis-restricted anemia) predominates in aged, anemic mice. The ratio of urinary albumin to creatinine (ACR) is commonly used as an indicator of kidney damage in mice. In females, the ACR is stable and does not rise significantly with age in the majority of strains, suggesting that declining kidney function is not a major cause of anemia of aging in female inbred mice. Significant increases in IL-6 and TNFα are seen in strains 129SvImJ, C3H/HeJ, and DBA/2J, suggesting a pro-inflammatory state. From this preliminary analysis of a large ongoing project, we can conclude: Hgb levels in mice vary significantly by strain and sex, and decline significantly with age in many strains. Other baseline hematological traits (e.g., red blood cell counts, platelet counts) likewise vary by strain, age and sex. These data are available via the Mouse Phenome Database (project Peters4). The anemia of aging seen in most strains correlates most closely with restricted hematopoiesis, as indicated by the failure of the reticulocyte count to increase in response to declining Hgb levels. There is growing evidence that decrements in hematopoietic stem cell number and function play a role in the aging process in humans. Notably, hematopoietic stem cell numbers and bone marrow cellularity data will be available on the MPD as these analyses are completed. HAM analysis suggests that distinct age-related loci influence Hgb levels in mice. In a small subset of strains, anemia of aging may reflect declining kidney function, as occurs in humans. Preliminary data suggests an increase in cytokine levels in some strains, again mimicking the aging human population. Increased IL-6 levels as a cause of anemia of aging is of particular interest due to its inhibition of hepcidin and thus iron availability. Overall, the data indicate that anemia of aging occurs in mice and models that seen in elderly human populations. Additional data including iron levels, T4, BUN, and more on aging inbred mouse strains will be posted to the MPD in the near future.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3492-3492
Author(s):  
Laura A. Paganessi ◽  
Andrew L. Walker ◽  
Stephanie A. Gregory ◽  
Henry C. Fung ◽  
Kent W. Christopherson

Abstract The exopeptidase CD26 (also known as DPPIV/dipeptidylpeptidase IV) cleaves dipeptides from the N-terminus of proteins that contain the required X-Pro or X-Ala motif. We have previously reported that inhibition or loss of CD26 activity results in a deficiency in normal granulocyte-colony stimulating factor (G-CSF) induced mobilization, suggesting that CD26 is a necessary component of mobilization (Christopherson, et al Blood 2003 and Christopherson, et al Exp Hematol 2003). The chemokine CXCL12 (SDF-1, stromal cell derived factor-1) contains the appropriate recognition sequence for CD26 induced cleavage. This combined with the importance of CXCL12 in the trafficking of hematopoietic stem and progenitor cells (HSC/HPC) suggests CXCL12 as a likely functional target of CD26 during G-CSF induced mobilization. For this reason we therefore decided to investigate whether genetically engineered mice lacking CD26 (CD26−/−) could be mobilized utilizing the CXCR4 antagonist, AMD3100. To evaluate this, ten week old C57BL/6 and CD26−/− mice (also on a C57BL/6 background) received a single subcutaneous injection of AMD3100 (1mg/1kg). One hour following injection the mice were euthanized by CO2 inhalation. Peripheral blood was then obtained by heart stick with a 1.2 ml syringe containing EDTA as an anticoagulant. A complete blood count was taken for each peripheral blood sample. Following red blood cell lysis, cells were plated for myeloid colony formation in a standard 1% methylcellulose colony assay containing the appropriate cytokines. Following 7 days of incubation at 5% O2, 5% CO2 and 37°C plates were scored for colony-forming units-granulocyte macrophage (CFU-GM), burst-forming units-erythroid (BFU-E), and colony-forming units-granulocyte, erythroid, macrophage, and megakaryocytic (CFU-GEMM). Data is presented as the number of colonies per femur for the bone marrow and as the number of colonies per ml of whole blood for the peripheral blood. AMD3100 treatment resulted in an increase in white blood cell (WBC) counts from 5.05±0.48 × 106/ml in untreated mice to 10.21±0.88×106/ml in treated mice (p≤0.01). An increase in WBC counts was also observed during AMD3100 treatment in CD26−/− mice from 7.77±1.28×106/ml in untreated mice to 16.7 ±2.11 × 106/ml in treated mice (p<0.01). AMD3100 treatment resulted in an increase in circulating myeloid progenitors in the peripheral blood of C57BL/6 and CD26−/− mice as compared to untreated C57BL/6 and CD26−/− mice respectively (p≤0.01). Specifically, a 2.38, 3.75, 12.33 fold increase in CFU-GM, BFU-E, and CFU-GEMM were observed in the peripheral blood of C57BL/6 mice after treatment. A 2.63, 5.48, 14.29 fold increase in CFU-GM, BFU-E, and CFU-GEMM were observed in the peripheral blood of CD26−/− mice after treatment. Existing pre-clinical and clinical data suggest that the CXCR4 antagonist, AMD3100, rapidly mobilizes hematopoietic progenitor cells from the bone marrow into the periphery. The results presented here provide pre-clinical evidence that disruption of the interaction between the CXCR4 chemokine receptor and CXCL12, via sub-cutaneous injection of AMD3100, mobilizes significant numbers of myeloid progenitors in mice, even in the absence of CD26. These results support the notion that CD26 is downstream of G-SCF treatment. Additionally, these results support the potential use of AMD3100 to treat patients that may have an altered ability to respond to G-CSF treatment as a result of a reduction or loss in CD26 activity. Future studies are warranted to evaluate potential variations in CD26 levels or activity in the general population, in differing patient populations, and during different treatment regimens.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1249-1249
Author(s):  
Mariusz Z Ratajczak ◽  
Kasia Mierzejewska ◽  
Janina Ratajczak ◽  
Magdalena Kucia

Abstract Abstract 1249 Background. Several phenotypes have been proposed for long-term repopulating hematopoietic stem cells (LT-HSCs) in murine bone marrow (BM). However, evidence from our and other laboratories has accumulated that adult murine tissues contain a population of developmentally early, so-called very small embryonic-like stem cells (VSELs), which we have proposed as playing an important role as precursors of LT-HSCs (Exp. Hematology 2011;39:225, Leukemia 2012;25,1278). As we reported, these cells are kept quiescent in the BM microenvironment by erasure of the somatic imprint in differentially methylated regions (DMRs) of some developmentally crucial, paternally imprinted genes (Igf2-H19, RasGRF1, and p57Kip2), which proper expression is required for proliferation and expansion of pluripotent stem cells (e.g., embryonic stem cells) (Leukemia 2009;23:2042). However, we also demonstrated that these cells may be specified into the hematopoietic lineage in vitro in co-cultures over OP9 stromal cells. Hypothesis. We hypothesized that these very small cells, which can be specified into the hematopoietic lineage ex vivo in an “artificial” OP9 microenvironment, should also be able to become specified into HSPCs in vivo in the normal BM microenvironment in situations of hematopoietic stress that promote the formation of new HSPCs. Experimental strategies. Normal C57Bl6 mice were bled (twice a week, 200 μl/bleeding) for 4 weeks, and by the end of each week were injected with bromodeoxyuridine (BrdU) to label cells that are in the cell cycle. These mice were subsequently sacrificed and BM cells, flushed from BM cavities as well as from crushed/collagenase-treated bones to recover cells associated with endosteal niches, were obtained from both control and bled mice. In these cell suspensions, we measured i) the total number of Sca-1+Lin–CD45+ HSPCs and small Sca-1+Lin–CD45– VSELs by FACS and ii) the number of cycling BrdU+ HSPCs and VSELs. Moreover, by employing RQ-PCR, we measured the expression of genes regulating the early stages of hematopoiesis and imprinted genes that keep VSELs quiescent in the cell cycle. We also tested the ability of VSELs from control and bled mice to differentiate into CD45+ HSPCs in OP9 co-cultures and their ability to reconstitute hematopoiesis in lethally irradiated mice. Salient results. We observed that the number of cycling BrdU+ VSELs increased from ∼1 ± 0.03% (control) to ∼26 ± 4% and ∼32 ± 6% among BM cells derived from flushed and crushed bones, respectively. Furthermore, in comparison with control animals, BM VSELs isolated from mice after chronic bleeding expressed lower levels of pluripotency markers such as Oct-4 and Nanog, upregulated expression of pro-proliferative mRNA whose expression is regulated by paternal imprinting (Igf2, IGF-1R, and RasGRF1), and downregulated expression of mRNA for paternally imprinted, proliferation-inhibiting H19 and p57Kip2genes. At the same time, the number of BM HSPCs increased from ∼17 ± 3% to 35 ± 7% and 1 ± 0.02% to 40 ± 5% in flushed and crushed bone-derived cells, respectively. Most importantly, we observed that VSELs isolated from bled mice highly upregulated the expression of genes involved in early stages of hematopoiesis, including Ikaros, Lmo2, GATA-2, HoxB4, PU.1, Scl and c-myb, and this correlated with their accelerated ability to become specified into CD45+ HSPCs in co-cultures over OP9 stroma. Finally, VSEL-derived CD45+ HSPCs, when isolated from OP9 cultures, grew methylocelulose colonies from all major hematopoietic lineages and were able to reconstitute hematopoiesis in lethally irradiated recipients. Conclusions. Our data, obtained in an in vivo murine model of hematopoietic stress from chronic bleeding, strongly support the notion that developmentally early murine Sca-1+Lin–CD45– VSELs represent a population of quiescent stem cells in BM that become specified into the hematopoietic lineage in vivo. We propose that, in order to establish the relationship of these cells to other LT-HSC phenotypes described in BM as well as to construct a complete developmental hierarchy, their hematopoietic potential should be compared side-by-side with other BM-derived stem cells isolated using different phenotypic criteria. Disclosures: Ratajczak: Neostem Inc: Member of SAB Other.


Sign in / Sign up

Export Citation Format

Share Document