A Forward RNAi Screen Identifies p38 MAP Kinase As a Druggable Target for Expansion of Human Hematopoietic Stem Cells

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2352-2352
Author(s):  
Aurelie Baudet ◽  
Jonas Larsson

Abstract Abstract 2352 While the mechanisms regulating key fate decisions such as self-renewal and differentiation in hematopoietic stem and progenitor cells (HSPC) remain poorly understood, intense efforts are being devoted to develop conditions that would enable ex vivo amplification of transplantable stem cells. We have developed a screening strategy to assess modulators of human HSCPs using pooled lentiviral shRNA libraries transduced into cord blood-derived stem/progenitor cells. We use the limited persistence of HSPCs under ex vivo culture conditions as a baseline for functional selection of shRNAs leading to prolonged maintenance or expansion of undifferentiated HSPCs. This approach enables complex, pooled screens in large numbers of cells. We further take advantage of next generation sequencing to track shRNA-transduced cells with high fidelity, allowing thousands of perturbations to be tested in parallel in a single pool of cells. Here we used a library of 2500 shRNAs targeting around 800 genes, mainly kinases and phosphatases, which include large numbers of “druggable” genes. The shRNAs composing the library were monitored by next generation sequencing in cord blood CD34+ cells sampled one day after transduction and following 20 days of culture, to determine their relative change in distribution during the culture period. The sequencing of all integrated proviruses containing shRNAs generated over 3 million sequences per sample. Analysis of the shRNA distribution before and after culture in 3 replicate screens revealed a dramatic enrichment of 3 independent shRNAs targeting MAPK14 (p38α). We could confirm that inhibition of MAPK14, mediated by RNA interference, leads to a proliferation advantage of CD34+ cells in culture, identifying p38 as a possible target for ex vivo stem cell expansion. We next used the chemical inhibitor SB203580 to inhibit p38 without genetic perturbation and in a non-permanent fashion. Culture of CD34+ cells under optimized conditions for expansion (serum-free medium supplemented with SCF, TPO and FLT3) with or without SB203580 showed a 3-fold increase of the stem cell enriched CD34+CD90+ cell population during 5 days of culture in SB203580 treated cells compared to control cells. Furthermore, when transplanted to immune-deficient NSG mice, SB203580 treated cells showed a dramatic increase in repopulating activity, as evidenced by the percentage of human engraftment 10 weeks after transplantation (SB203580: 30±6.4% vs control: 7.5±3.6%, p< 0,001). Thus, under otherwise optimized culture conditions for stem cell expansion, the addition of the p38 inhibitor leads to a significant increase in stem cell activity. To understand the basis for the increase in stem cell activity, we assayed SB203580 treated cells with respect to cell cycling and survival rate, but found a similar cell division history (shown by cytoplasmic dye dilution assays) and similar levels of apoptotic cells (shown by Annexin V staining) compared to control cells. Interestingly, however, when the cells were assayed for reactive oxygen species (ROS), we detected significantly reduced levels of ROS in SB203580 treated cells, implicating modulation of ROS as a possible mechanism behind the enhanced stem cell output. Taken together, using a functional forward genetic screen, we have been able to identify p38 MAP kinase as a highly promising target to enhance hematopoietic stem cell activity in ex vivo expansion settings. These results further support the feasibility of pooled RNAi screens in conjunction with next generation sequencing to identify genes and pathways that regulate primary human stem cell populations. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 36-36
Author(s):  
Mehrnaz Safaee Talkhoncheh ◽  
Fredrik Ek ◽  
Aurelie Baudet ◽  
Christine Karlsson ◽  
Roger Olsson ◽  
...  

Abstract Despite extensive studies over the last decades, little is known about the mechanisms governing human hematopoietic stem cell (HSC) fate decisions. In particular, it has been challenging to define culture conditions in which HSCs can be expanded for clinical benefit. Application of small molecule screening to modulate stem cells has emerged as a useful tool for identification of new compounds with ability to expand hematopoietic stem and progenitor cells (HSPCs). Such screens have mainly relied on the expression of CD34 as predictor of stem cell activity in cultured cells. However, CD34 defines a broad repertoire of progenitor cells and does not define stem cell function. We found that the long-term repopulation potential of cultured human HSPCs is exclusively contained within a discrete cell population co-expressing CD34 and CD90, while the vast majority of progenitor cells are found in the CD34+CD90- population. Tracking the CD34+ CD90+ population is therefore a sensitive and specific tool to predict stem cell activity in cultured hematopoietic cells and provides a good basis for a screen aimed at discovering modifiers of stem cell expansion. To search broadly for novel and potential modifiers of ex vivo HSCs expansion we next developed and optimized a small molecule screen in human cord blood (CB) derived CD34+ cells. We screened >500 small molecules from 8 different annotated chemical libraries for the phenotypic expansion of CD34+ CD90+ cells following a 6-day culture in serum-free medium supplemented with stem cell factor (SCF), thrombopoietin (TPO) and fms-like tyrosine kinase 3 ligand (FL). The numbers of CD34+ CD90+ cells for each molecule, tested at two different concentrations, was compared to DMSO treated controls. Following the initial screen, several candidate hits were selected and subjected to a dose response validation experiment from which we selected four top candidate molecules. Two of these molecules were histone deacetylase (HDAC) inhibitors, which recently have been reported to facilitate expansion of CB derived HSCs. One of the top candidates, Ciclopirox ethanolamine (CE), had previously not been implicated in HSC expansion. Ciclopirox ethanolamine is known as an antifungal agent and iron chelator. It has further been shown to suppress cancer cell survival through inhibition of Wnt/beta catenin signaling. We found that CB cells cultured with CE had a 4-fold increase in CD34+90+ cell number compared to DMSO treated controls following 6 days of culture. Interestingly, the total cell count was not different, suggesting a specific increase in CD34+ CD90+ cell number rather than an overall higher proliferation rate. When plated in methylcellulose, CE cultured cells generated increased numbers of myeloid colonies. Moreover, CE treated cells gave rise to multilineage colonies (CFU-GEMM) that could not be detected from the control cultures. To further test the functional capacity of cells cultured with CE, we transplanted cultured equivalents of 30,000 CB CD34+ cells (cultured with or without CE) into sub lethally irradiated NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mice. Human hematopoietic reconstitution in peripheral blood was determined 16 weeks later. Mice transplanted with CE cultured cells showed higher human CD45 engraftment 16 weeks post transplant compared to control cells (33.2±6.7% vs 14.6±5% p=0.04). The engrafted cells contributed to both myeloid and lymphoid lineages. This shows that Ciclopirox ethanolamine enhances the long-term engraftment capacity of ex vivo cultured HSCs and suggests that it should be considered in stem cell expansion protocols, either alone or in combination with other molecules. We are currently addressing the basis for the increased stem cell activity mediated by Ciclopirox ethanolamine using parameters for differentiation, cell cycling and apoptosis. In addition, we are comparing Ciclopirox ethanolamine with other recently defined modifiers of HSC expansion. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5044-5044
Author(s):  
Manja Wobus ◽  
Guruchandar Arulmozhivarman ◽  
Martin Kraeter ◽  
Jens Friedrichs ◽  
Martin Stoeter ◽  
...  

Abstract Introduction The identification of compounds which increase the number but also keep or enhance the activity of hematopoietic stem and progenitor cells (HSPCs) could improve the clinical outcome after autologous and allogeneic hematopoietic stem cell transplantation (HSCT). So far, most attempts to increase HSPC numbers ex vivo have been unsuccessful because of either inadequate cell numbers and/or loss of engraftment capacity and HSPC quality during expansion. Executing drug discovery screens in vertebrate systems is generally expensive, technically challenging and time consuming. Therefore, the zebrafish represents a versatile vertebrate model allowing HSPC regulation and development studies during embryogenesis and adulthood. Methods We used a semi-automated chemical screen to identify modulators of HSPC activity by transgenic (cmyb:EGFP) zebrafish embryos. Verification of identified histone deacetylase (HDAC) inhibitor candidates was carried out in vitro using human CD34+ HSPCs which were isolated from apharesis samples of healthy donors after mobilization with G-CSF by anti-CD34 coupled magnetic beads. The influence of HDAC inhibitors on HSPC phenotype, gene expression pattern as well as adhesion and migration capacity was analyzed after 5 days of treatment either in single or in co-culture with bone marrow-derived mesenchymal stromal cells (MSCs). Results The HDAC inhibitors valproic acid (VPA), resminostat and entinostat were shown to significantly amplify the number of hematopoietic precursors in a chemical in vivo zebrafish embryo screen (Arulmozhivarman et al. 2016). Treatment of human CD34+ HSPCs with these compounds in vitro resulted in a significantly increased percentage of CD34+CD90+ cells up to 60% compared to controls which showed only 2% of double positive cells as well as in 3-fold higher CD34+ and about 12-fold higher CD34+CD90+ absolute cell numbers. CD34 is a well-known surface marker for human immature HSPCs and in combination with CD90 it defines a potentially pluripotent subpopulation. In a co-culture setting, we found that VPA treated cells showed 2 to 3-fold higher attachment capacity on MSCs compared to the control cells. This finding led us to quantify the adhesive capacity of cells using static adhesion assay and atomic force microscopy based single-cell force spectroscopy (AFM-SCFS). Interestingly, detachment forces of VPA treated HSPCs were 3 times increased on MSCs compared to control cells and a similar phenotype was observed by static adhesion assay. Accordingly, the chemokine-mediated migration of VPA treated HSPCs towards SDF-1/CXCL12 was inhibited. To reveal underlying downstream molecules and mechanisms mediating the modified cellular characteristics, a whole genome expression array was carried out for HSPCs treated with VPA in comparison to untreated controls. Amongst a panel of regulated genes, the melanoma cell adhesion molecule (MCAM/CD146), Notch 3 and its downstream effector Hes-1 as well as the SDF-1 receptor CXCR-4 were found to be significantly changed. Whereas the decreased expression of CXCR4 correlates with the inhibited migration potential of VPA-treated HSPCs and Notch-3/Hes-1 have a known role in normal and malignant hematopoiesis (Gu et al. 2016), the induced expression of MCAM on HSPCs was not described so far. The result was confirmed by flow cytometry which revealed a 40% MCAM-positive cell population when treated with VPA, whereas the control showed only negative cells. Additionally, significant higher transcript levels were detected for MCAM by quantitative real-time PCR in VPA expanded cells. Recently, we described a role of MCAM in MSCs for the hematopoietic support (Stopp et al. 2013). The inducible expression in HSPCs may reflect homotypic interactions which preserve a more immature subpopulation with high stem cell activity. Conclusion We describe for the first time the ability of the HDAC inhibitors VPA, resminostat and entinostat to efficiently expand CD34+ HSPCs ex vivo especially supporting a CD34+CD90+ subpopulation with potentially high stem cell activity. Moreover, a potential role of MCAM in this context may offer new perspectives of the HSPC expansion ex vivo for the improvement of HSCT. Disclosures No relevant conflicts of interest to declare.


2003 ◽  
Vol 75 (2) ◽  
pp. 314-323 ◽  
Author(s):  
Yuk Yin Ng ◽  
Berris van Kessel ◽  
Henk M. Lokhorst ◽  
Miranda R. M. Baert ◽  
Caroline M. M. van den Burg ◽  
...  

Blood ◽  
2012 ◽  
Vol 119 (26) ◽  
pp. 6255-6258 ◽  
Author(s):  
Aurélie Baudet ◽  
Christine Karlsson ◽  
Mehrnaz Safaee Talkhoncheh ◽  
Roman Galeev ◽  
Mattias Magnusson ◽  
...  

We report on a forward RNAi screen in primary human hematopoietic stem and progenitor cells, using pooled lentiviral shRNA libraries deconvoluted by next generation sequencing. We identify MAPK14/p38α as a modulator of ex vivo stem cell proliferation and show that pharmacologic inhibition of p38 dramatically enhances the stem cell activity of cultured umbilical cord blood derived hematopoietic cells. p38 inhibitors should thus be considered in strategies aiming at expanding stem cells for clinical benefit.


Sign in / Sign up

Export Citation Format

Share Document