The GEF Lsc Regulates Hematopoietic Stem Cell Motility, Mobilization and Recruitment.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1346-1346
Author(s):  
Isabelle Petit ◽  
Prashant Kaul ◽  
Daniel J. Lerner ◽  
Shahin Rafii

Abstract Lsc is a Rho GTPase guanine nucleotide exchange factor (RhoGEF) that physically and functionally links G-protein coupled receptors (GPCR) to the monomeric GTPase RhoA in mature hematopoietic and other cells. Lsc−/− (LscKO) mice have a peripheral leukocytosis, abnormal neutrophil and B cell motility, and immune response deficiencies. Although Lsc is required for neutrophil homeostasis, its role in hematopoietic stem and progenitor cells is unknown. In this study, we have used LscKO mice to determine if Lsc is required for normal stem cell motility and mobilization. Initially, we used immunofluorescence labeling to demonstrate that hematopoietic stem and progenitor cells express Lsc. This suggested that Lsc may be required for normal hematopoietic stem and progenitor cell migration. Stromal-cell derived factor-1 (SDF-1) is a potent chemokine for hematopoietic stem cells and activates the CXCR4 GPCR. It has been reported that Lsc is not required for SDF-1-stimulated migration of mature murine T and B cells. However, using a bare-filter transwell assay, we found that while LscKO Sca-1+ cells and Sca-1+Lin- cells have normal spontaneous migration, they have significantly increased SDF-1-stimulated migration compared to their wild-type (WT) counterparts, 1.4 and 2.3 fold, respectively. We then demonstrated that adhesion of LscKO Sca-1+ cells to bone marrow (BM) stromal MS-5 cells was normal, indicating that impaired adhesion was not responsible for the abnormal SDF-1-stimulated migration. Using colony assay, we demonstrated that LscKO mice have a normal number of circulating peripheral stem and progenitor cells. Strikingly, after 5 days of G-CSF administration, LscKO mice have 1.6 fold and 2.3 fold the number of peripheral mature WBC and stem and progenitor cells (colony forming units), respectively, compared to WT mice. Recruitment of BM CXCR4+ pro-angiogenic stem and progenitor cells has been linked to enhanced tumor angiogenesis. Because LscKO BM cells had abnormal SDF-1-stimulated migration and mobilization, we hypothesized that Lsc might regulate tumor angiogenesis as well. To this end, we assessed tumor growth in LscKO mice by injecting congenic Lewis lung carcinoma cells subcutaneously into LscKO mice and WT controls. Preliminary experiments revealed that tumors were 3.3 times larger in the LscKO mice as compared to WT mice. Quantification of the tumor vessels with anti-CD31 staining demonstrated that the tumors in LscKO mice were 1.4 fold more vascularized than controls. In summary, our results demonstrate that the Rho GEF Lsc is essential for normal hematopoietic stem cell migration and mobilization. In addition, we propose that absence of Lsc facilitates tumor growth by promoting BM stem and progenitor cell recruitment to the neo-angiogenic vessels, possibly augmenting tumor vascularization.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1293-1293
Author(s):  
Hong Qian ◽  
Sten Eirik W. Jacobsen ◽  
Marja Ekblom

Abstract Within the bone marrow environment, adhesive interactions between stromal cells and extracellular matrix molecules are required for stem and progenitor cell survival, proliferation and differentiation as well as their transmigration between bone marrow (BM) and the circulation. This regulation is mediated by cell surface adhesion receptors. In experimental mouse stem cell transplantation models, several classes of cell adhesion receptors have been shown to be involved in the homing and engraftment of stem and progenitor cells in BM. We have previously found that integrin a6 mediates human hematopoietic stem and progenitor cell adhesion to and migration on its specific ligands, laminin-8 and laminin-10/11 in vitro (Gu et al, Blood, 2003; 101:877). Using FACS analysis, the integrin a6 chain was now found to be ubiquitously (>95%) expressed in mouse hematopoietic stem and progenitor cells (lin−Sca-1+c-Kit+, lin−Sca-1+c-Kit+CD34+) both in adult bone marrow and in fetal liver. In vitro, about 70% of mouse BM lin−Sca-1+c-Kit+ cells adhered to laminin-10/11 and 40% adhered to laminin-8. This adhesion was mediated by integrin a6b1 receptor, as shown by functional blocking monoclonal antibodies. We also used a functional blocking monoclonal antibody (GoH3) against integrin a6 to analyse the role of the integrin a6 receptor for the in vivo homing of hematopoietic stem and progenitor cells. We found that the integrin a6 antibody inhibited the homing of bone marrow progenitors (CFU-C) into BM of lethally irradiated recipients. The number of homed CFU-C was reduced by about 40% as compared to cells incubated with an isotype matched control antibody. To study homing of long-term repopulating stem cells (LTR), antibody treated bone marrow cells were first injected intravenously into lethally irradiated primary recipients. After three hours, bone marrow cells of the primary recipients were analysed by competitive repopulation assay in secondary recipients. Blood analysis 16 weeks after transplantation revealed an 80% reduction of stem cell activity of integrin a6 antibody treated cells as compared to cells treated with control antibody. These results suggest that integrin a6 plays an important role for hematopoietic stem and progenitor cell homing in vivo.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1387-1387
Author(s):  
Hong Qian ◽  
Sten Eirik W. Jacobsen ◽  
Marja Ekblom

Abstract Homing of transplanted hematopoietic stem cells (HSC) in the bone marrow (BM) is a prerequisite for establishment of hematopoiesis following transplantation. However, although multiple adhesive interactions of HSCs with BM microenviroment are thought to critically influence their homing and subsequently their engraftment, the molecular pathways that control the homing of transplanted HSCs, in particular, of fetal HSCs are still not well understood. In experimental mouse stem cell transplantation models, several integrins have been shown to be involved in the homing and engraftment of both adult and fetal stem and progenitor cells in BM. We have previously found that integrin a6 mediates human hematopoietic stem and progenitor cell adhesion to and migration on its specific ligands, laminin-8 and laminin-10/11 in vitro (Gu et al, Blood, 2003; 101:877). Furthermore, integrin a6 is required for adult mouse HSC homing to BM in vivo (Qian et al., Abstract American Society of Hematology, Blood 2004 ). We have now found that the integrin a6 chain like in adult HSC is ubiquitously (>99%) expressed also in fetal liver hematopoietic stem and progenitor cells (lin−Sca-1+c-Kit+, LSK ). In vitro, fetal liver LSK cells adhere to laminin-10/11 and laminin-8 in an integrin a6b1 receptor-dependent manner, as shown by function blocking monoclonal antibodies. We have now used a function blocking monoclonal antibody (GoH3) against integrin a6 to analyse the role of the integrin a6 receptor for the in vivo homing of fetal liver hematopoietic stem and progenitor cells to BM. The integrin a6 antibody inhibited homing of fetal liver progenitors (CFU-C) into BM of lethally irradiated recipients. The number of homed CFU-C in BM was reduced by about 40% as compared to the cells incubated with an isotype matched control antibody. To study homing of long-term repopulating stem cells, BM cells were first incubated with anti-integrin alpha 6 or anti-integrin alpha 4 or control antibody, and then injected intravenously into lethally irradiated primary recipients. After three hours, BM cells of the primary recipients were analysed by competitive repopulation assay in secondary recipients. Blood analysis up to 16 weeks after transplantation showed that no reduction of stem cell reconstitution from integrin a6 antibody treated cells as compared to cells treated with control antibody. In accordance with this, fetal liver HSC from integrin a6 gene deleted embryos did not show any impairment of homing and engraftment in BM as compared to normal littermates. These results suggest that integrin a6 plays an important developmentally regulated role for homing of distinct hematopoietic stem and progenitor cell populations in vivo.


Blood ◽  
2015 ◽  
Vol 125 (12) ◽  
pp. 1890-1900 ◽  
Author(s):  
Sarah A. Kinkel ◽  
Roman Galeev ◽  
Christoffer Flensburg ◽  
Andrew Keniry ◽  
Kelsey Breslin ◽  
...  

Key Points Depletion of Jarid2 in mouse and human hematopoietic stem cells enhances their activity. Jarid2 acts as part of PRC2 in hematopoietic stem and progenitor cells.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2413-2413 ◽  
Author(s):  
Jingmei Hsu ◽  
Hsuan-Ting Huang ◽  
Chung-Tsai Lee ◽  
Shuqian Yu ◽  
Leonard I. Zon ◽  
...  

Abstract ATP-dependent chromatin remodeling enzymes alter histone/DNA interactions, and are involved in the regulation of transcription, chromosome segregation, DNA replication and repair. We identified Chromodomain Helicase DNA binding protein 7 (CHD7) as a negative regulator of hematopoietic stem cell function. Autosomal dominant CHD7 mutations are associated with CHARGE syndrome (Coloboma of the eye, Heart defects, Atresia of the choanae, Retardation of growth and/or development, Genital and/or urinary abnormalities, and Ear abnormalities and deafness). Although several cases of T and B cell immunodeficiency in a subset of CHARGE syndrome patients have been reported, no previous role for CHD7 in hematopoiesis had been proposed. We show that morpholino knockdown of Chd7 in zebrafish embryos results in an increased number of runx1 and c-myb expressing hematopoietic stem and progenitor cells in the aorta/gonad/mesonephros (AGM) region, and this effect is cell autonomous as determined by blastula transplantation. Heterozygous germline Chd7 deletion in mice also results in an increased number of phenotypic hematopoietic stem and progenitor cells (Runx1+c-kit+CD31+) in the AGM region. Downstream lineages such as myeloid and erythroid cells are expanded in zebrafish, and similarly conditional pan-hematopoietic deletion of Chd7 in mice with Vav1-Cre results in myeloid lineage expansion and increased granulocyte/monocyte progenitors. Consistent with these results, microarray analysis of murine CD48- CD150+ Lin- Sca-1+ c-kit+ (SLAM LSK) phenotypic long term repopulating HSCs shows up-regulation of genes in several subclasses of the myeloid lineages. Interestingly, although CHD7 deficient mouse bone marrow had a normal frequency of SLAM LSK cells, it had a two-fold higher frequency of functional LT-HSCs as determined by whole bone marrow, purified LSK and SLAM LSK limiting dilution transplants. ChIP-seq performed in human CD34+ hematopoietic stem and progenitor cells show CHD7 localizes to genes encoding many key hematopoietic transcription factors including MYB and RUNX1. The most abundant transcription factor motif under CHD7 genomic binding sites is a RUNX motif, indicating that CHD7 and Runx1 function together. We show CHD7, Runx1 and c-Myb interact both physically and genetically. CHD7 function in hematopoietic stem cells is dependent on both Runx1 and c-Myb since the increase in hematopoiesis in fish upon morpholino knockdown of Chd7 is abolished when either Runx1 or c-Myb is mutated. In summary, our study identifies CHD7 as a novel evolutionarily conserved negative epigenetic regulator of HSCs and progenitors through its interaction with Runx1 and c-Myb. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 214 (10) ◽  
pp. 2967-2983 ◽  
Author(s):  
Matthias Kollek ◽  
Gesina Voigt ◽  
Christian Molnar ◽  
Fabronia Murad ◽  
Daniela Bertele ◽  
...  

During hematopoietic stem cell transplantation, a substantial number of donor cells are lost because of apoptotic cell death. Transplantation-associated apoptosis is mediated mainly by the proapoptotic BCL-2 family proteins BIM and BMF, and their proapoptotic function is conserved between mouse and human stem and progenitor cells. Permanent inhibition of apoptosis in donor cells caused by the loss of these BH3-only proteins improves transplantation outcome, but recipients might be exposed to increased risk of lymphomagenesis or autoimmunity. Here, we address whether transient inhibition of apoptosis can serve as a safe but efficient alternative to improve the outcome of stem cell transplantation. We show that transient apoptosis inhibition by short-term overexpression of prosurvival BCL-XL, known to block BIM and BMF, is not only sufficient to increase the viability of hematopoietic stem and progenitor cells during engraftment but also improves transplantation outcome without signs of adverse pathologies. Hence, this strategy represents a promising and novel therapeutic approach, particularly under conditions of limited donor stem cell availability.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 98-98
Author(s):  
Rebekka K. Schneider ◽  
Dirk Heckl ◽  
Marcus Järås ◽  
Lisa Chu ◽  
McConkey Marie ◽  
...  

Abstract Casein kinase 1α (Ck1α) is a serine/threonine kinase located in the common deleted region (5q32) in del(5q) myelodysplastic syndrome (MDS). Ck1α is a regulator of the canonical WNT signaling pathway and may play a role in the clonal advantage of del(5q) cells. In addition, we identified CK1α as a therapeutic target in myeloid malignancies in an in vivo RNA interference screen, and haploinsufficiency for CK1α could further sensitize del(5q) cells to CK1α inhibition. To explore the biology and therapeutic potential of CK1α in MDS, we generated a conditional Ck1α knockout mouse model. Conditional homozygous inactivation of Ck1α resulted in bone marrow failure, ablation of hematopoietic stem and progenitor cells, a severe anemia and rapid lethality within 7-12 days, confirming that Ck1α is essential for hematopoietic stem and progenitor cell survival. In contrast, mice with haploinsufficiency of Ck1α developed a hypercellular bone marrow, as is typical in MDS, a significantly elevated white blood cell count (p=0.002) and normal hemoglobin levels. The hematopoietic stem cells (LSK, LT-HSC, ST-HSC) as well as progenitor cells (LK, pre-GMP, GMP, pre-CFU-e, CFU-e, pre-megakaryocytes-erythrocytes) were not affected by Ck1α haploinsufficiency 14 days after induction. Only the megakaryocytic progenitor cells (p=0.04) were significantly reduced. This finding was in line with severe dysplasia and hypolobulated micromegakaryocytes observed in the bone marrow, another typical histomorphological feature of del(5q) MDS. In long-term experiments up to 8 months, the survival of mice with Ck1α haploinsufficiency was not impaired, although we observed an exhaustion of the stem cell pool with significant reduction of ST-HSC (p<0.001), LT-HSC (p=0.003), and MPP (p=0.007). We were able to demonstrate that this significant reduction is a cell-extrinsic effect. In transplantation and HSC repopulation assays, an intact HSC function and even a significant expansion of hematopoietic stem cells and progenitor cells with Ck1α haploinsufficiency was confirmed in comparison to MxCre controls (LSK p=0.019; LK p=0.035; CMP p=0.036; GMP p=0.027; MEP p=0.005), suggesting a repopulation advantage of HSC with Ck1α haploinsufficiency. In contrast, Ck1α homozygous deletion leads to a cell-autonomous, p53-mediated HSC failure in transplantation assays. To dissect the mechanism of hematopoietic stem cell expansion in Ck1α haploinsufficiency on the one hand and the hematopoietic stem cell ablation after Ck1α ablation on the other hand, we analyzed regulatory mechanisms including proliferation and apoptosis in LK cells (myeloid progenitor cells) and LSK cells (enriched for hematopoietic stem cells). Ablation of Ck1α led to a significant increase (p=0.001) in the number of LSK and LK in the S/M/G2 phase, accompanied by a significant reduction in the G0/G1 fraction, suggesting their exit from quiescence. Ck1α haploinsufficiency led to a significant increase in the fraction of cycling cells in myeloid progenitor cells (LK, p=0.052), the quiescent hematopoietic stem cells were not significantly affected. In Western Blots of ckit+ hematopoietic stem and progenitor cells, a significant increase of intracellular ß-catenin levels was detected in both Ck1α haploinsufficient and even stronger in Ck1α ablated cells, accompanied by an exit from stem cell quiescence shown by loss of p21-mediated growth arrest and up-regulation of phosphorylated retinoblastoma protein indicating cell cycle progression from G0 to G1 in comparison to the MxCre+ control cells. Ck1α ablation led to p53-mediated apoptosis in stem and progenitor cells (Annexin V/7-AAD). In Ck1α haploinsufficient cells, apoptosis was not significantly induced in neither LK cells or in LSK cells although p53 induction was observed in the bone marrow. Taken together, our results indicate that Ck1α is essential for hematopoietic stem and progenitor cell survival, but that Ck1α haploinsufficiency does not decrease, and may increase, hematopoietic stem cell function. This finding highlights the potential of preferential elimination of the del(5q) hematopoietic stem cells through Ck1α inhibtion and thus provides a potential therapeutic window. Consistent with this hypothesis, targeting the haploinsufficient kinase activity in vitro with the Ck1α small molecule inhibitor D4476, selectively targets CK1α haploinsufficient cells relative to wild-type cells. Disclosures: Järås: Cantargia: Equity Ownership.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2418-2418
Author(s):  
Anja Köhler ◽  
Vince Schmithorst ◽  
Marie-Dominique Filippi ◽  
Marnie A. Ryan ◽  
Deidre Daria ◽  
...  

Abstract Hematopoiesis, the process in which blood cells are generated from hematopoietic stem and progenitor cells (HSPCs) is primarily confined to the bone cavities. The interactions of hematopoietic cells with stroma cells forming niches inside the bone cavities are central to hematopoiesis, as these regulate cell proliferation, self-renewal and differentiation. Hematopoietic cell/stroma interactions have thus been, in analogy to the immunological synapse, named stem/progenitor cell synapses. So far, visualization of the behavior of somatic stem and progenitor cells in an undisturbed in vivo environment has not been reported for the mammalian system and consequently, the cellular dynamics of stem, progenitor and differentiated cells in vivo are only poorly defined. We developed and performed intravital time-lapse 2-photon microscopy in the marrow of the long bones (tibia) of mice to study the behavior and dynamics of differentiated hematopoietic cells as well as HPCs and HSCs in close vicinity to the endosteum in vivo over time. We demonstrate that HPCs as well as HSCs reside in close vicinity to the endosteum, further supporting the notion of an endosteal stem cell niche, and that they are, in contrast to differentiated macrophages and dendritic cells, solitary and immobile. Both HPCs and HSCs occupy distinct positions relative to the endosteum and show cell protrusion movement consistent with an active stem/progenitor cell synapse. Lastly, we report that aged HSCs show increased protrusion movement and localize more distantly to the endosteum compared to young HSCs. In addition, aged HSCs present with reduced adhesion to stroma as well as reduced polarity upon adhesion in vitro, implying a connection between altered stem cell dynamics in vivo and stem cell aging. The intravital imaging technology developed might establish a basis for further delineating additional important questions in stem cell biology like cellular mechanisms of hematopoietic stem cell self-renewal and differentiation in the context of the stroma/niche in vivo.


Sign in / Sign up

Export Citation Format

Share Document