scholarly journals e0193 A critical role of ckit in CXCR4-mediated progenitor cell niche maintenance and mobilisation

Heart ◽  
2010 ◽  
Vol 96 (Suppl 3) ◽  
pp. A62-A62
Author(s):  
C. Min ◽  
Z. Qiutang ◽  
L. Douglas
Blood ◽  
2011 ◽  
Vol 118 (10) ◽  
pp. 2733-2742 ◽  
Author(s):  
Cristina Mazzon ◽  
Achille Anselmo ◽  
Javier Cibella ◽  
Cristiana Soldani ◽  
Annarita Destro ◽  
...  

Abstract Hematopoiesis is the process leading to the sustained production of blood cells by hematopoietic stem cells (HSCs). Growth, survival, and differentiation of HSCs occur in specialized microenvironments called “hematopoietic niches,” through molecular cues that are only partially understood. Here we show that agrin, a proteoglycan involved in the neuromuscular junction, is a critical niche-derived signal that controls survival and proliferation of HSCs. Agrin is expressed by multipotent nonhematopoietic mesenchymal stem cells (MSCs) and by differentiated osteoblasts lining the endosteal bone surface, whereas Lin−Sca1+c-Kit+ (LSK) cells express the α-dystroglycan receptor for agrin. In vitro, agrin-deficient MSCs were less efficient in supporting proliferation of mouse Lin−c-Kit+ cells, suggesting that agrin plays a role in the hematopoietic cell development. These results were indeed confirmed in vivo through the analysis of agrin knockout mice (Musk-L;Agrn−/−). Agrin-deficient mice displayed in vivo apoptosis of CD34+CD135− LSK cells and impaired hematopoiesis, both of which were reverted by an agrin-sufficient stroma. These data unveil a crucial role of agrin in the hematopoietic niches and in the cross-talk between stromal and hematopoietic stem cells.


2012 ◽  
Vol 197 (3) ◽  
pp. 196-208 ◽  
Author(s):  
Rosa M. Ceinos ◽  
Eva Torres-Nuñez ◽  
Ruben Chamorro ◽  
Beatriz Novoa ◽  
Antonio Figueras ◽  
...  

Blood ◽  
2011 ◽  
Vol 117 (20) ◽  
pp. 5350-5361 ◽  
Author(s):  
Helen He Zhu ◽  
Kaihong Ji ◽  
Nazilla Alderson ◽  
Zhao He ◽  
Shuangwei Li ◽  
...  

Abstract The stem cell factor (SCF)/Kit system has served as a classic model in deciphering molecular signaling events in the hematopoietic compartment, and Kit expression is a most critical marker for hematopoietic stem cells (HSCs) and progenitors. However, it remains to be elucidated how Kit expression is regulated in HSCs. Herein we report that a cytoplasmic tyrosine phosphatase Shp2, acting downstream of Kit and other RTKs, promotes Kit gene expression, constituting a Kit-Shp2-Kit signaling axis. Inducible ablation of PTPN11/Shp2 resulted in severe cytopenia in BM, spleen, and peripheral blood in mice. Shp2 removal suppressed the functional pool of HSCs/progenitors, and Shp2-deficient HSCs failed to reconstitute lethally irradiated recipients because of defects in homing, self-renewal, and survival. We show that Shp2 regulates coordinately multiple signals involving up-regulation of Kit expression via Gata2. Therefore, this study reveals a critical role of Shp2 in maintenance of a functional HSC/progenitor pool in adult mammals, at least in part through a kinase-phosphatase-kinase cascade.


2014 ◽  
Vol 51 (1) ◽  
pp. 142-154 ◽  
Author(s):  
Ting Li ◽  
Dehua Yang ◽  
Jia Li ◽  
Yu Tang ◽  
Juan Yang ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 309-309
Author(s):  
Alan M. Hanash ◽  
Jarrod A Dudakov ◽  
Guoqiang Hua ◽  
Lauren F. Young ◽  
Maggie O'connor ◽  
...  

Abstract Abstract 309 Despite decades of intensive research, graft vs. host disease (GVHD) remains a major complication of allogeneic transplantation. Much progress has been made toward understanding GVHD pathophysiology and the mechanisms regulating the donor immune response, however no factors have been identified to regulate the response of transplant recipients to GVHD and its concomitant damage. Furthermore, virtually all strategies available to reduce clinical GVHD do so by limiting the donor immune system at the expense of therapeutic graft vs. leukemia/lymphoma (GVL) responses. IL-22 is a recently characterized cytokine that is produced by both helper T cells and innate lymphoid cells (ILC). Expression of its receptor (IL-22R) is generally limited to epithelial and other non-hematopoietic cells, and it has demonstrated a protective role for intestinal epithelium during experimental colitis, providing a signal for intestinal epithelial cell survival, proliferation, and wound healing. We investigated the role of IL-22 in GVHD during MHC-mismatched C57BL/6 (B6) into BALB/c murine bone marrow transplantation (BMT). Elimination of IL-22 with anti-IL-22 neutralizing antibody lead to increased GVHD mortality post-BMT in comparison to isotype control (p<.01). Interestingly, use of IL-22 knockout (KO) donor marrow or T cells had no impact on survival or on the ability to clear A20 lymphoma post-BMT, but IL-22 KO BMT recipients demonstrated significantly increased mortality during GVHD (p<.01). The critical role of recipient-derived IL-22 was confirmed in a minor antigen-mismatched LP into B6 BMT model (Figure 1). IL-22 KO recipients again demonstrated significantly increased mortality during GVHD (p<.001) and histopathologic evidence of GVHD in the small (p<.01) and large (p<.001) intestine and liver (p<.001). Furthermore, BMT into hematopoietic IL-22 KO chimeras also demonstrated increased GVHD mortality (p<.05) and clinical scoring (p<.001), indicating that the source of protective IL-22 is residual host-derived radio-resistant hematopoietic cells. IL-22 ELISA on tissue homogenates following BMT with wild type donors and recipients indicated that IL-22 levels are increased three weeks post-BMT in both small and large intestine (p<.05), as well as following sublethal radiation without transplantation (p<.05 small intestine; p<.01 large) in comparison to normal controls. However, IL-22 was significantly reduced in both tissues during GVHD (2 weeks post-BMT in small intestine, Figure 2A; 3 weeks post-BMT in large, p<.05), suggesting that IL-22 was produced by residual host hematopoietic cells that were being eliminated during GVHD. Consistent with this, we identified IL-22-producing host ILC (CD45+CD3−RORg+) in small intestine lamina propria two weeks post-BMT, whereas no IL-22 production could be identified in CD3+ or RORg− subsets, and these IL-22-producing ILC were significantly reduced during GVHD (p<.05, Figure 2B). Interestingly, although host IL-22 deficiency was associated with increased GVHD morbidity, mortality, and pathology, no major differences were observed in donor lymphocyte infiltration of recipient intestines, frequencies of donor lymphoid subsets, or donor T cell inflammatory cytokines post-BMT. In contrast, IL-22R expression on recipient intestinal epithelium was increased post-BMT, and GVHD in IL-22 KO recipients led to increased damage of the intestinal epithelium as measured by FITC-dextran absorption (p<.05). Additionally, IL-22R expression was identified by immunohistochemistry in intestinal crypts where the stem/progenitor cell niche is located, and use of LGR5-LacZ reporter mice indicated that intestinal stem cells (ISC) were indeed targets of GVHD. Finally, histologic assessment of the ISC niche demonstrated exacerbated loss of both ISC (p<.001, Figure 2C) and Paneth cells (p<.05) in IL-22 KO mice during GVHD. In summary, IL-22 is produced post-BMT by host ILC that are eliminated during GVHD, and IL-22 deficiency increases GVHD morbidity and mortality. While IL-22 deficiency did not significantly alter the donor immune response, it did lead to increased GVHD pathology, loss of epithelial integrity, and damage to the ISC niche. IL-22 is thus critical for protection of host epithelium during GVHD but is not involved in GVL. This may be exploited in the future to reduce clinical GVHD without limiting the curative potential of the transplant. Disclosures: Fouser: Pfizer: Employment.


2015 ◽  
Vol 14 (2) ◽  
pp. 133-143 ◽  
Author(s):  
Yuning Hou ◽  
Yanning Wu ◽  
Shukkur M. Farooq ◽  
Xiaoqing Guan ◽  
Shuo Wang ◽  
...  

2013 ◽  
Vol 41 (12) ◽  
pp. 1050-1061 ◽  
Author(s):  
Sarah L. Ellis ◽  
Shen Y. Heazlewood ◽  
Brenda Williams ◽  
Andrea J. Reitsma ◽  
Jochen Grassinger ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3674-3674
Author(s):  
Michihiro Kobayashi ◽  
Yuanshu Dong ◽  
Hao Yu ◽  
Yunpeng Bai ◽  
Sisi Chen ◽  
...  

Abstract The phosphatase of regenerating liver family of phosphatases, consisting of PRL1, PRL2 and PRL3, represents an intriguing group of proteins implicated in cell proliferation and tumorigenesis. However, the role of PRLs in normal and malignant hematopoiesis is largely unknown. While SCF/KIT signaling plays an important role in hematopoietic stem and progenitor cell (HSPC) maintenance, how SCF/KIT signaling is regulated in HSPCs remains poorly understood. We here report that PRL2 regulates HSPC maintenance through regulating SCF/KIT signaling. To define the role of PRL2 in hematopoiesis, we analyzed the hematopoietic stem cell (HSC) behavior in Prl2 deficient mice generated by our group. Prl2 deficiency results in ineffective hematopoiesis and impairs the long-term repopulating ability of HSCs. In addition, Prl2 null HSPCs are less proliferative and show decreased colony formation in response to SCF stimulation. Furthermore, Prl2 null HSPCs show reduced activation of the PI3K/AKT and ERK signaling in steady state and following SCF stimulation. Importantly, we found that PRL2 associates with KIT and the ability of PRL2 to enhance SCF signaling depends on its enzymatic activity, demonstrating that PRL2 mediates SCF/KIT signaling in HSPCs. Thus, PRL2 plays a critical role in hematopoietic stem and progenitor cell maintenance through regulating SCF/KIT signaling. Furthermore, loss of Prl2 decreased the ability of oncogenic KITD814V mutant in promoting hematopoietic progenitor cell proliferation and in activation of signaling pathways. We also checked the expression of PRL2 proteins in human AML cell lines and found increased level of PRL2 proteins in some acute myeloid leukemia (AML) cells compared with normal human bone marrow cells, indicating that PRL2 may play a pathological role in AML. Our results suggest that the PRL2 phosphatase may be a druggable target in myeloproliferative disease (MPD) and acute myeloid leukemia (AML) with oncogenic KIT mutations. Disclosures: No relevant conflicts of interest to declare.


Hematology ◽  
2014 ◽  
Vol 2014 (1) ◽  
pp. 71-76 ◽  
Author(s):  
Sophia R. Balderman ◽  
Laura M. Calvi

Abstract The BM microenvironment and its components regulate hematopoietic stem and progenitor cell (HSC) fate. An abnormality in the BM microenvironment and specific dysfunction of the HSC niche could play a critical role in initiation, disease progression, and response to therapy of BM failure syndromes. Therefore, the identification of changes in the HSC niche in BM failure syndromes should lead to further knowledge of the signals that disrupt the normal microenvironment. In turn, niche disruption may contribute to disease morbidity, resulting in pancytopenia and clonal evolution, and its understanding could suggest new therapeutic targets for these conditions. In this chapter, we briefly review the evidence for the importance of the BM microenvironment as a regulator of normal hematopoiesis, summarize current knowledge regarding the role of dysfunctions in the BM microenvironment in BM failure syndromes, and propose a strategy through which niche stimulation can complement current treatment for myelodysplastic syndrome.


Sign in / Sign up

Export Citation Format

Share Document