scholarly journals The Act of Controlling Adult Stem Cell Dynamics: Insights from Animal Models

Biomolecules ◽  
2021 ◽  
Vol 11 (5) ◽  
pp. 667
Author(s):  
Meera Krishnan ◽  
Sahil Kumar ◽  
Luis Johnson Kangale ◽  
Eric Ghigo ◽  
Prasad Abnave

Adult stem cells (ASCs) are the undifferentiated cells that possess self-renewal and differentiation abilities. They are present in all major organ systems of the body and are uniquely reserved there during development for tissue maintenance during homeostasis, injury, and infection. They do so by promptly modulating the dynamics of proliferation, differentiation, survival, and migration. Any imbalance in these processes may result in regeneration failure or developing cancer. Hence, the dynamics of these various behaviors of ASCs need to always be precisely controlled. Several genetic and epigenetic factors have been demonstrated to be involved in tightly regulating the proliferation, differentiation, and self-renewal of ASCs. Understanding these mechanisms is of great importance, given the role of stem cells in regenerative medicine. Investigations on various animal models have played a significant part in enriching our knowledge and giving In Vivo in-sight into such ASCs regulatory mechanisms. In this review, we have discussed the recent In Vivo studies demonstrating the role of various genetic factors in regulating dynamics of different ASCs viz. intestinal stem cells (ISCs), neural stem cells (NSCs), hematopoietic stem cells (HSCs), and epidermal stem cells (Ep-SCs).

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 396-396
Author(s):  
Stephane Barakat ◽  
Julie Lambert ◽  
Guy Sauvageau ◽  
Trang Hoang

Abstract Abstract 396 Hematopoietic stem cells that provide short term reconstitution (ST-HSCs) as well as hematopoietic progenitors expand from a small population of long term hematopoietic stem cells (LT-HSCs) that are mostly dormant cells. The mechanisms underlying this expansion remain to be clarified. SCL (stem cell leukemia), is a bHLH transcription factor that controls HSC quiescence and long term competence. Using a proteomics approach to identify components of the SCL complex in erythroid cells, we and others recently showed that the ETO2 co-repressor limits the activity of the SCL complex via direct interaction with the E2A transcription factor. ETO2/CBF2T3 is highly homologous to ETO/CBFA2T1 and both are translocation partners for AML1. We took several approaches to identify ETO2 function in HSCs. We initially found by Q-PCR that ETO2 is highly expressed in populations of cells enriched in short-term HSC (CD34+Flt3-Kit+Sca+Lin-) and lympho-myeloid progenitors (CD34+Flt3+Kit+Sca+Lin-) and at lower levels in LT-HSCs (CD34-Kit+Sca+Lin- or CD150+CD48-Kit+Sca+Lin-). Next, the role of ETO2 was studied by overexpression or downregulation combined with transplantation in mice. Ectopic ETO2 expression induces a 100 fold expansion of LT-HSCs in vivo in transplanted mice associated with differentiation blockade in all lineages, suggesting that ETO2 overexpression overcomes the mechanisms that limit HSC expansion in vivo. We are currently testing the role of the NHR1 domain of ETO2 in this expansion. Conversely, shRNAs directed against ETO2 knock down ET02 levels in Kit+Sca+Lin- cells, causing a ten-fold decrease in this population after transplantation, associated with reduced short-term reconstitution in mice. Finally, proliferation assays using Hoechst and CFSE indicate that ETO2 downregulation affects cell division (CFSE) and leads to an accumulation of Kit+Sca+Lin-cells in G0/G1 state (Hoescht). In conclusion, we show that ETO2 is highly expressed in ST-HSCs and lymphoid progenitors, and controls their expansion by regulating cell cycle entry at the G1-S checkpoint. In addition, ETO2 overexpression converts the self-renewal of maintenance into self-renewal of expansion in LT-HSCs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Author(s):  
Lei Zhang ◽  
Jing Li ◽  
Hui Xu ◽  
Xianyu Shao ◽  
Li Fu ◽  
...  

c-Myc (Myc hereafter) is found to be deregulated and/or amplified in most acute myeloid leukemias (AML). Almost all AML cells are dependent upon Myc for their proliferation and survival. Thus Myc has been proposed as a critical anti-AML target. Myc has Max-mediated trans-activational and Miz1-mediated trans-repressional activities. The role of Myc-Max-mediated trans-activation in the pathogenesis of AML has been well-studied; however the role of Myc-Miz1-mediated trans-repression in AML is still somewhat obscure. MycV394D is a mutant form of Myc which lacks trans-repressional activity due to a defect in its ability to interact with Miz1. We found that, compared to Myc, the oncogenic function of MycV394D is significantly impaired. The AML/myeloproliferative disorder which develops in mice receiving MycV394D-transduced hematopoietic stem/progenitor cells (HSPCs) is significantly delayed compared to mice receiving Myc-transduced HSPCs. Using a murine MLL-AF9 AML model, we found that AML cells expressing MycV394D (intrinsic Myc deleted) are partially differentiated and show reductions in both colony-forming ability in vitro and leukemogenic capacity in vivo. The reduced frequency of leukemia stem cells (LSCs) among MycV394D-AML cells and their reduced leukemogenic capacity during serial transplantation suggest that Myc-Miz1 interaction is required for the self-renewal of LSCs. In addition, we found that MycV394D-AML cells are more sensitive to chemotherapy than are Myc-AML cells. Mechanistically, we found that the Myc represses Miz1-mediated expression of Cebpα and Cebpδ, thus playing an important role in the pathogenesis of AML by maintaining the undifferentiated state and self-renewal capacity of LSCs.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1568-1568
Author(s):  
Yumi Fukuchi ◽  
Kana Kuroda ◽  
Ken Sadahira ◽  
Ryouichi Ono ◽  
Daniel G. Tenen ◽  
...  

Abstract Abstract 1568 MLL translocations found in acute leukemia possess unique clinical characteristics. They have over 50 different fusion partners and show poor prognosis. These MLL fusion proteins lost H3K4 methyltransferase activity of wild-type MLL, but gained the ability to induce aberrant expression of HoxA cluster genes. Moreover, these proteins are able to transform hematopoietic stem/progenitor cells into leukemic stem cells (LSCs). Previous studies have shown that C/EBPα and PU.1, well-known myeloid specific transcription factors, were common molecular targets of myeloid malignancies. We and others have recently shown that C/EBPα and PU.1 are negative regulators of hematopoietic stem cells, suggesting that these transcription factors may play a role in the generation of LSCs. Because we have little knowledge on the role of C/EBPα and PU.1 in MLL-leukemia, we asked whether these key myeloid transcription factors were involved in the leukemogenesis by MLL-fusion proteins, especially in the stages of leukemia initiation and/or progression. First, we investigated the role of C/EBPα and PU.1 by in vitro self-renewal capacity and in vivo leukemia formation by MLL-fusion oncogenes. Bone marrow (BM) cells were harvested from C57BL/6J mice treated with 5-FU (150 mg/kg), and pre-stimulated with recombinant mouse (rm) SCF, rmIL-6, rhFL, rhTPO (50 ng/ml each). Cells were then transduced with pMYs-IG-MLL-ENL or pMXs-IG-MLL-Septin6, serially replated in methylcellulose, and transferred to rmIL-3 (10 ng/ml) containing liquid culture (immortalized cells), or were transplanted into lethally irradiated recipients (primary leukemic cells). MLL-ENL (or MLL-Septin6) immortalized cells or MLL-ENL primary leukemic cells were transduced with pMXs-IRES-DsRed-C/EBPα-ER or pMXs-IRES-DsRed-PU.1-ER. GFP+DsRed+ cells were sorted and serially replated in methylcellulose with or without 4-hydroxytamoxifen (4-HT) (1 mM), or were treated with or without 4-HT (1 mM) for 5 days followed by transplantion into sublethally irradiated secondary recipients. The results showed that overexpression of PU.1, but not C/EBPα, completely suppressed the serial replating capacity of MLL-ENL- and MLL-Septin6-immortalized cells. Moreover, activation of PU.1 suppressed propagation of MLL-ENL leukemic cells in the secondary recipients. In contrast, activation of C/EBPα did not eradicate leukemic cells in the same settings. To elucidate the role of PU.1 in the initiation of leukemia by MLL-ENL, we took PU.1+/− BM cells, or E14.5 fetal liver (FL) cells from PU.1-/- or +/− mice, and examined their capability to initiate leukemia when they were transduced with MLL-ENL. The result showed that leukemia did not develop in the absence of PU.1, and PU.1 haploinsufficiency prolonged survival of the recipients. A role of PU.1 in leukemia progression/maintenance by MLL-ENL was also tested using PU.1 conditional knockout mice. BM cells from PU.1flox/flox or flox/- mice were transduced with pMYs-IG-MLL-ENL, and transplanted into lethally irradiated recipients. PU.1-flox allele was conditionally deleted in primary leukemia cells by induction of Cre recombinase, whose effect was assessed by transplanting Cre-treated cells into secondary recipients. The result showed that conditional inactivation of PU.1 perturbed propagation of MLL-ENL leukemic cells, indicating that PU.1 is absolutely required not only for initiation, but also for maintenance of MLL-leukemia. Taken together, these results suggest that the dosage of PU.1 activity has profound impact on the self-renewal of LSCs and in vivo leukemia formation induced by MLL-fusion oncogenes. Therefore, PU.1 may serve as a potential therapeutic target for MLL-leukemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3162-3162
Author(s):  
Fabienne A. Brenet ◽  
Joseph M. Scandura

Abstract Abstract 3162 The cyclin-dependent kinase inhibitor (CDKI) CDKN1C (p57) is a tumor suppressor gene with strong differential expression in both human and murine hematopoietic stem cells (HSC). Whereas the expression of other CDKIs is normal in most hematopoietic malignancies, p57 expression is silenced in 30 to 55% of acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), and B-Cell lymphoma patients suggesting a role as suppressor of hematopoietic cell transformation. p57 has a unique role in embryogenesis functioning in tissue-specific developmental programs to coordinate proliferation, differentiation and apoptosis. We have identified p57 as an early transcriptional target of the tumor suppressor TGFβ which is required for TGFβ-induced cytostasis of human CD34+ hematopoietic progenitor/stem cells; functions that provide a mechanistic basis for its expression being silenced in many aggressive human myeloid and lymphoid malignancies. To understand the role of p57 in HSC function, we used an engineered mouse strain deficient in p57. Using limiting-dilution HSC transplantation, we found that the fetal livers of p57-null mice have ∼4-fold fewer HSCs than their wild-type littermate controls. When we transplanted wt recipient mice with a mixture of p57-wt and p57-null fetal liver mononuclear cells (FLMC), we found that the p57-null hematopoietic cells were underrepresented in the blood of the recipient animals at steady-state. Both the limiting-dilution and competitive repopulation experiments indicate that the absence of p57 compromises HSC development. Strikingly, despite this quantitative deficit, we found that p57-null HSCs are qualitatively superior to p57-wt HSCs. Using serial transplantation, an assay of in vivo HSC self-renewal, we found that p57-null HSCs can be transplanted for two generations beyond which p57-wt HSCs are exhausted and unable to reconstitute hematopoiesis in the recipient animals. This result demonstrates under the tonic strain of serial transplantation, p57 normally serves to restrain HSC self-renewal. Importantly, mice with p57-null hematopoiesis are less sensitive to myelotoxic stress induced by the chemotherapeutics, 5-Fluorouracil (5FU) and cytarabine and have a shallower and shorter nadir following such treatment. Mice with p57-null hematopoiesis also recovered more briskly to the haemolytic agent phenylhydrazine suggesting that p57 serves a critical function restraining the stress-response of hematopoiesis. We next examined whether p57 might be involved in regulating the maintenance of LKSCD34- Flk2- long-term HSCs, LKSCD34+ Flk2- short-term HSCs, or LKSCD34+ Flk2+ MPPs (multipotent progenitors), LKSca-CD34+ FcRlow CMPs (common myeloid progenitors), LKSca- CD34+ FcR+ GMPs (granulocyte-monocyte progenitors) and LKSca- CD34- FcR- MEPs (megakaryocyte erythroid progenitors). Unlike in steady-state where the amount of cells in each subset in p57-null and wt bone marrow is comparable, we found significant variations in the p57-null bone marrow reconstitution after 5FU treatment. Furthermore when we administered 5FU to mice transplanted with mixtures of p57-null and p57-wt HSCs, we found that p57-null hematopoiesis contributed disproportionately to hematopoietic recovery and that this enhanced competition was durable. Our findings reveal a novel role of p57 to restrain HSC self-renewal during periods of hematopoietic stress. Deletion of p57 decelerates hematopoietic cells exhaustion due to serial transplantation and improves long-term engraftment, largely because of increased self-renewing divisions of HSCs in vivo. These results suggest that cells deficient in p57 have an advantage over p57-expressing cells only after hematopoietic stress suggesting that silenced p57 expression can serve to promote malignant hematopoiesis at the expense of normal hematopoiesis and contribute to chemotherapy resistance. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2309-2309
Author(s):  
Jian Huang ◽  
Peter S. Klein

Abstract Abstract 2309 Hematopoietic stem cells (HSCs) maintain the ability to self-renew and to differentiate into all lineages of the blood. The signaling pathways regulating hematopoietic stem cell (HSCs) self-renewal and differentiation are not well understood. We are very interested in understanding the roles of glycogen synthase kinase-3 (Gsk3) and the signaling pathways regulated by Gsk3 in HSCs. In our previous study (Journal of Clinical Investigation, December 2009) using loss of function approaches (inhibitors, RNAi, and knockout) in mice, we found that Gsk3 plays a pivotal role in controlling the decision between self-renewal and differentiation of HSCs. Disruption of Gsk3 in bone marrow transiently expands HSCs in a b-catenin dependent manner, consistent with a role for Wnt signaling. However, in long-term repopulation assays, disruption of Gsk3 progressively depletes HSCs through activation of mTOR. This long-term HSC depletion is prevented by mTOR inhibition and exacerbated by b-catenin knockout. Thus GSK3 regulates both Wnt and mTOR signaling in HSCs, with opposing effects on HSC self-renewal such that inhibition of Gsk3 in the presence of rapamycin expands the HSC pool in vivo. In the current study, we found that suppression of the mammalian target of rapamycin (mTOR) pathway, an established nutrient sensor, combined with activation of canonical Wnt/ß-catenin signaling, allows the ex vivo maintenance of human and mouse long-term HSCs under cytokine-free conditions. We also show that combining two clinically approved medications that activate Wnt/ß-catenin signaling and inhibit mTOR increases the number of long-term HSCs in vivo. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Hong-Chen Yan ◽  
Yu Sun ◽  
Ming-Yu Zhang ◽  
Shu-Er Zhang ◽  
Jia-Dong Sun ◽  
...  

Abstract Background Skin-derived stem cells (SDSCs) are a class of adult stem cells (ASCs) that have the ability to self-renew and differentiate. The regulation mechanisms involved in the differentiation of ASCs is a hot topic. Porcine models have close similarities to humans and porcine SDSCs (pSDSCs) offer an ideal in vitro model to investigate human ASCs. To date, studies concerning the role of yes-associated protein (YAP) in ASCs are limited, and the mechanism of its influence on self-renewal and differentiation of ASCs remain unclear. In this paper, we explore the link between the transcriptional regulator YAP and the fate of pSDSCs. Results We found that YAP promotes the pluripotent state of pSDSCs by maintaining the high expression of the pluripotency genes Sox2, Oct4. The overexpression of YAP prevented the differentiation of pSDSCs and the depletion of YAP by small interfering RNA (siRNAs) suppressed the self-renewal of pSDSCs. In addition, we found that YAP regulates the fate of pSDSCs through a mechanism related to the Wnt/β-catenin signaling pathway. When an activator of the Wnt/β-catenin signaling pathway, CHIR99021, was added to pSDSCs overexpressing YAP the ability of pSDSCs to differentiate was partially restored. Conversely, when XAV939 an inhibitor of Wnt/β-catenin signaling pathway, was added to YAP knockdown pSDSCs a higher self-renewal ability resulted. Conclusions our results suggested that, YAP and the Wnt/β-catenin signaling pathway interact to regulate the fate of pSDSCs.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 91-91
Author(s):  
Yuichi Hirata ◽  
Kazuhiro Furuhashi ◽  
Hiroshi Ishi ◽  
Hao-Wei Li ◽  
Sandra Pinho ◽  
...  

Abstract A crucial player in immune regulation, FoxP3+ regulatory T cells (Tregs) are drawing attention for their heterogeneity and noncanonical functions. For example, specific subsets of Tregs in the adipose tissue control metabolic indices; muscle Tregs potentiate muscle repair, and lung Tregs prevent tissue damage. These studies, together with a previous finding that Tregs are enriched in the primary site for hematopoiesis, the bone marrow (BM), prompted us to examine whether there is a special Treg population which controls hematopoietic stem cells (HSCs). We showed that HSCs within the BM were frequently adjacent to distinctly activated FoxP3+ Tregs which highly expressed an HSC marker, CD150. Moreover, specific reduction of BM Tregs achieved by conditional deletion of CXCR4in Tregs, increased reactive oxygen species (ROSs) in HSCs. The reduction of BM Tregs further induced loss of HSC quiescence and increased HSC numbers in a manner inhibited by anti-oxidant treatment. Additionally, this increase in HSC numbers in mice lacking BM Tregs was reversed by transfer of CD150high BM Tregs but not of CD150low BM Tregs. These results indicate that CD150high niche-associated Tregs maintain HSC quiescence and pool size by preventing oxidative stress. We next sought to identify an effector molecule of niche Tregs which regulates HSCs. Among molecules highly expressed by niche Tregs, we focused on CD39 and CD73, cell surface ecto-enzymes which are required for generation of extracellular adenosine, because 1) CD39highCD73high cells within the BM were prevalent among CD150high Tregs and 2) HSCs highly expressed adenosine 2a receptors (A2AR). We showed that both conditional deletion of CD39 in Tregs and in vivo A2AR antagonist treatment induced loss of HSC quiescence and increased HSC pool size in a ROS-dependent manner, which is consistent with the findings in mice lacking BM Tregs. In addition, transfer of CD150high BM Tregs but not of CD150low BM Tregs reversed the increase in HSC numbers in FoxP3cre CD39flox mice. The data indicate that niche Treg-derived adenosine regulates HSCs. We further investigated the protective role of niche Tregs and adenosine in radiation injury against HSCs. Conditional deletion of CD39 in Tregs increased radiation-induced HSC apoptosis. Conversely, transfer of as few as 15,000 CD150high BM Tregs per B6 mouse (iv; day-1) rescued lethally-irradiated (9.5Gy) mice by preventing hematopoiesis failure. These observations indicate that niche Tregs protect HSCs from radiation stress. Finally, we investigated the role of niche Tregs in allogeneic (allo-) HSC transplantation. Our previous study showed that allo-hematopoietic stem and progenitor cells but not allo-Lin+ cells persisted in the BM of non-conditioned immune-competent recipients without immune suppression in a manner reversed by systemic Treg depletion1. This observation suggests that HSCs have a limited susceptibility to immune attack, as germline and embryonic stem cells are located within immune privileged sites. Because the study employed systemic Treg depletion and non-conditioned recipients, it remains unknown whether niche Tregs play a critical role in immune privilege of HSCs and in allo-HSC engraftment following conditioning. We showed here that the reduction of BM Tregs and conditional deletion of CD39 in Tregs abrogated allo-HSC persistence in non-conditioned immune-competent mice as well as allo-HSC engraftment following nonmyeloablative conditioning. Furthermore, transfer of CD150high BM Tregs but not of other Tregs (15,000 cells/recipient; day -2) significantly improved allo-HSC engraftment. This effect of niche Treg transfer is noteworthy given that 1-5 million Tregs per mouse were required in case of transfer of spleen or lymph node Tregs. These observations suggest that niche Tregs maintain immune privilege of HSCs and promote allo-HSC engraftment. In summary, our studies identify a unique niche-associated Treg subset and adenosine as regulators of HSC quiescence, numbers, stress response, engraftment, and immune privilege, further highlighting potential clinical utility of niche Treg transfer in radiation-induced hematopoiesis failure and in allo-HSC engraftment (under revision in Cell Stem Cell). 1 Fujisaki, J. et al. In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature474, 216-219, doi:10.1038/nature10160 (2011). Disclosures No relevant conflicts of interest to declare.


2014 ◽  
Vol 115 (suppl_1) ◽  
Author(s):  
Hyun Sook Hong ◽  
Suna Kim ◽  
Youngsook Son

Bone marrow stem cells, especially, endothelial precursor cells (EPC), mesenchymal stem cells (MSC) or hematopoietic stem cell (HSC) are expected as reparative cells for the repair of a variety of tissue damages such as stroke and myocardial infarction, even though their role in the repair is not demonstrated. This report was investigated to find a role of Substance-p (SP) as a reparative agent in the tissue repair requiring EPC and MSC. In order to examine EPC (EPC SP ) and MSC (MSC SP ) mobilized by SP, we injected SP intravenously for consecutive 2 days and saline was injected as a vehicle. At 3 post injection, peripheral blood (PB) was collected.To get mesenchymal stem cells or endothelial progenitor cells, MNCs were incubated in MSCGM or EGM-2 respectively for 10 days. Functional characteristics of the EPC SP were proven by the capacity to form endothelial tubule network in the matrigel in vitro and in the matrigel plug assay in vivo. In contrast, MSC SP did not form a tube-like structure but formed a pellet-structure on matrigel. However, when both cells were premixed before the matrigel assay, much longer and branched tubular network was formed, in which a-SMA expressing MSC SP were decorating outside of the endothelial tube, especially enriched at the bifurcating point. MSC SP may contribute and reinforce elaborate vascular network formation in vivo by working as pericyte-like cells. Thus, the EPC SP and MSC SP were labeled with PKH green and PKH red respectively and their tubular network was examined. Well organized tubular network was formed, which was covered by PKH green labeled cells and was decorated in a punctate pattern by PKH red labeled cells. In order to investigate the role of EPC SP and MSC SP specifically in vivo, rabbit EPC SP and MSC SP were transplanted to full thickness skin wound. The vessel of EPC SP -transplanted groups was UEA-lectin+, which was not covered with a-SMA+ pericytes but EPC SP + MSC SP -transplanted groups showed, in part, a-SMA+ pericyte-encircled UEA-lectin+ vessels. This proved the specific role of MSC SP as pericytes. From these data, we have postulated that the collaboration of MSC and EPC is essential for normal vessel structure and furthermore, accelerated wound healing as ischemia diseases, which can be stimulated through by SP injection.


2019 ◽  
Vol 2019 ◽  
pp. 1-16 ◽  
Author(s):  
Qing Xia ◽  
Tao Han ◽  
Pinghua Yang ◽  
Ruoyu Wang ◽  
Hengyu Li ◽  
...  

Background. MicroRNAs (miRNAs) play a critical role in the regulation of cancer stem cells (CSCs). However, the role of miRNAs in liver CSCs has not been fully elucidated. Methods. Real-time PCR was used to detect the expression of miR-miR-28-5p in liver cancer stem cells (CSCs). The impact of miR-28-5p on liver CSC expansion was investigated both in vivo and in vitro. The correlation between miR-28-5p expression and sorafenib benefits in HCC was further evaluated in patient-derived xenografts (PDXs). Results. Our data showed that miR-28-5p was downregulated in sorted EpCAM- and CD24-positive liver CSCs. Biofunctional investigations revealed that knockdown miR-28-5p promoted liver CSC self-renewal and tumorigenesis. Consistently, miR-28-5p overexpression inhibited liver CSC’s self-renewal and tumorigenesis. Mechanistically, we found that insulin-like growth factor-1 (IGF-1) was a direct target of miR-28-5p in liver CSCs, and the effects of miR-28-5p on liver CSC’s self-renewal and tumorigenesis were dependent on IGF-1. The correlation between miR-28-5p and IGF-1 was confirmed in human HCC tissues. Furthermore, the miR-28-5p knockdown HCC cells were more sensitive to sorafenib treatment. Analysis of patient-derived xenografts (PDXs) further demonstrated that the miR-28-5p may predict sorafenib benefits in HCC patients. Conclusion. Our findings revealed the crucial role of the miR-28-5p in liver CSC expansion and sorafenib response, rendering miR-28-5p an optimal therapeutic target for HCC.


2019 ◽  
Vol 116 (4) ◽  
pp. 1447-1456 ◽  
Author(s):  
Rong Lu ◽  
Agnieszka Czechowicz ◽  
Jun Seita ◽  
Du Jiang ◽  
Irving L. Weissman

While the aggregate differentiation of the hematopoietic stem cell (HSC) population has been extensively studied, little is known about the lineage commitment process of individual HSC clones. Here, we provide lineage commitment maps of HSC clones under homeostasis and after perturbations of the endogenous hematopoietic system. Under homeostasis, all donor-derived HSC clones regenerate blood homogeneously throughout all measured stages and lineages of hematopoiesis. In contrast, after the hematopoietic system has been perturbed by irradiation or by an antagonistic anti-ckit antibody, only a small fraction of donor-derived HSC clones differentiate. Some of these clones dominantly expand and exhibit lineage bias. We identified the cellular origins of clonal dominance and lineage bias and uncovered the lineage commitment pathways that lead HSC clones to different levels of self-renewal and blood production under various transplantation conditions. This study reveals surprising alterations in HSC fate decisions directed by conditioning and identifies the key hematopoiesis stages that may be manipulated to control blood production and balance.


Sign in / Sign up

Export Citation Format

Share Document