scholarly journals In vivo dynamics of human hematopoietic stem cells: novel concepts and future directions

2019 ◽  
Vol 3 (12) ◽  
pp. 1916-1924 ◽  
Author(s):  
Serena Scala ◽  
Alessandro Aiuti

Abstract Unveiling the mechanisms and the cellular dynamics at the basis of human hematopoietic homeostasis has been a main focus for the scientific community since the discovery of a pool of multipotent hematopoietic stem cells (HSCs) capable of sustaining the hematopoietic output throughout life and after transplantation. Recently, new works shed light on the (1) differentiation paths, (2) size and replication rate of human HSC population at steady state, and (3) role of the distinct subpopulations comprising the hematopoietic stem and progenitor cell reservoir after transplantation. These papers exploited cutting-edge technologies, including vector integration site clonal tracking, spontaneous mutations, and deep transcriptome profiling. Here we discuss the latest updates in human hematopoietic system biology and in vivo dynamics, highlighting novel concepts and common findings deriving from different approaches and the future directions of these studies. Taken together, this information contributed to partially resolving the complexity of the in vivo HSC behavior and has major implications for HSC transplantation and gene therapy as well as for the development of future therapies.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1182-1182
Author(s):  
Stephanie Laufs ◽  
Ursula Sorg ◽  
Veronika Kleff ◽  
Laila Gao ◽  
Michael Flasshove ◽  
...  

Abstract Gene transfer of the DNA repair protein O6-methylguanine-DNA-methyltransferase (MGMT) into hematopoietic stem cells has been shown to protect hematopoiesis from the toxic side effects of O6-guanine alkylating drugs such as BCNU, ACNU or temozolomide (TMZ). In addition, MGMT gene transfer allows efficient in vivo selection of transduced hematopoietic stem cells and enrichment of genetically corrected cells in the context of gene therapy for monogenetic diseases. We here have analysed the long-term effect of MGMT gene transfer on the hematopoietic stem cell compartment using an in vivo murine transplantation/gene therapy model and a retroviral vector carrying the gene for MGMTP140K, a mutant resistant to the wtMGMT-specific inhibitor O6-benzylguanine (BG). Serial transplants were performed and primary, secondary as well as tertiary recipients were treated with combined BG/ACNU, BG/BCNU or BG/TMZ chemotherapy at doses myeloablative in non-MGMT-protected hematopoiesis. Serial transplantation was performed with 1.8 – 3.0 x 106 mononuclear bone marrow cells and 2 to 3 animals were transplanted per primary or secondary animal. While initial gene transfer efficiency was low (1–5% of cells engrafted at week four) chemotherapy resulted in efficient selection of transduced cells in primary animals (70–90% transgene expression in peripheral blood). Secondary and tertiary recipients showed 40–80% transgene expression even before CTX. Efficient stem cell engraftment and protection from CTX was demonstrated in > 90% of secondary animals, while tertiary recipients clearly demonstrated compromised engraftment and a substantial number of animals did not survive CTX treatment. Retroviral vector integration site analysis to study the clonality of hematopoiesis of stem cells by ligation mediated PCR (LM-PCR) was performed in the serially transplanted mice. In three mice of the secondary transplantation cohort we detected 3, 0, and 6 clones, respectively. In three mice of the tertiary transplantation cohort 7, 2, and 2 clones, respectively, were found. Thus, an exhaustion of transduced hematopoiesis following regenerative stress by high dose chemotherapy was not evident. Of the total 20 detected clones one could not be mapped to the mouse genome, while the others could be blasted against the mouse genome (assembly 2004, http://genome.ucsc.edu/; >99.5% identity). It turned out that 5 of 8 integrations landed in RefSeq in the tertiary transplantation cohort, while 3 of 8 integrations occurred in RefSeq genes in the secondary transplantation cohort. This suggests that clones profit from the transcription machinery of their integration site. Thus, our LM-PCR results indicate that the multiclonality of hematopoiesis is conserved after serial transplants which may be considered a safety feature for drug-resistance gene therapy. Furthermore, vector integration in highly resistant stem cells is favored in actively transcribed genomic regions.


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.


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.


2000 ◽  
Vol 191 (2) ◽  
pp. 253-264 ◽  
Author(s):  
Jos Domen ◽  
Samuel H. Cheshier ◽  
Irving L. Weissman

Hematopoietic stem cells (HSC) give rise to cells of all hematopoietic lineages, many of which are short lived. HSC face developmental choices: self-renewal (remain an HSC with long-term multilineage repopulating potential) or differentiation (become an HSC with short-term multilineage repopulating potential and, eventually, a mature cell). There is a large overcapacity of differentiating hematopoietic cells and apoptosis plays a role in regulating their numbers. It is not clear whether apoptosis plays a direct role in regulating HSC numbers. To address this, we have employed a transgenic mouse model that overexpresses BCL-2 in all hematopoietic cells, including HSC: H2K-BCL-2. Cells from H2K-BCL-2 mice have been shown to be protected against a wide variety of apoptosis-inducing challenges. This block in apoptosis affects their HSC compartment. H2K-BCL-2–transgenic mice have increased numbers of HSC in bone marrow (2.4× wild type), but fewer of these cells are in the S/G2/M phases of the cell cycle (0.6× wild type). Their HSC have an increased plating efficiency in vitro, engraft at least as well as wild-type HSC in vivo, and have an advantage following competitive reconstitution with wild-type HSC.


Blood ◽  
2004 ◽  
Vol 103 (11) ◽  
pp. 4084-4092 ◽  
Author(s):  
Claudia Orelio ◽  
Kirsty N. Harvey ◽  
Colin Miles ◽  
Robert A. J. Oostendorp ◽  
Karin van der Horn ◽  
...  

Abstract Apoptosis is an essential process in embryonic tissue remodeling and adult tissue homeostasis. Within the adult hematopoietic system, it allows for tight regulation of hematopoietic cell subsets. Previously, it was shown that B-cell leukemia 2 (Bcl-2) overexpression in the adult increases the viability and activity of hematopoietic cells under normal and/or stressful conditions. However, a role for apoptosis in the embryonic hematopoietic system has not yet been established. Since the first hematopoietic stem cells (HSCs) are generated within the aortagonad-mesonephros (AGM; an actively remodeling tissue) region beginning at embryonic day 10.5, we examined this tissue for expression of apoptosis-related genes and ongoing apoptosis. Here, we show expression of several proapoptotic and antiapoptotic genes in the AGM. We also generated transgenic mice overexpressing Bcl-2 under the control of the transcriptional regulatory elements of the HSC marker stem cell antigen-1 (Sca-1), to test for the role of cell survival in the regulation of AGM HSCs. We provide evidence for increased numbers and viability of Sca-1+ cells in the AGM and subdissected midgestation aortas, the site where HSCs are localized. Most important, our in vivo transplantation data show that Bcl-2 overexpression increases AGM and fetal liver HSC activity, strongly suggesting that apoptosis plays a role in HSC development.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1359-1359
Author(s):  
Prabal Banerjee ◽  
Lindsey Crawford ◽  
Michelle Sieburg ◽  
Patrick Green ◽  
Mark A Beilke ◽  
...  

Abstract Human T-lymphotropic virus type-1 (HTLV-1) is a human retrovirus linked to cancer and is the etiologic agent of Adult T-cell leukemia/lymphoma (ATLL), an aggressive CD4+/CD25+ T cell malignancy. The early molecular events induced by HTLV-1 infection as well as the role of various viral genes in the induction of leukemia remain unclear, predominantly due to the lack of an animal model that recapitulates ATLL development. HTLV-1 infection of humanized NOD/SCID mice (HTLV-1- HU-SCID) was achieved by inoculation of NOD/SCID mice with CD34+ hematopoietic progenitor cells and stem cells (CD34+ HP/HSCs) infected ex vivo with HTLV-1. HTLV-1-HU-NOD/SCIDmice consistently developed CD4+CD25+ T cell lymphomas with clinical characteristics associated with ATLL and infected mice showed hyperproliferation of infected human stem cells (CD34+CD38−) in the bone marrow. Inoculation of NOD/SCID mice withCD34+ HP/HSCs transduced with a lentivirus vector (LV) expressing the HTLV-1oncoprotein (Tax1) also developed CD4+CD25+ lymphomas. The HTLV-1 bZIP protein(HBZ), encoded by the minus strand of the HTLV-1 genome, is expressed in all ATLL cells and has been implicated in the maintenance of leukemogenesis. HBZ has previously been previously shown to interact with numerous cellular factors and can modulate Tax1 activity in vitro. To establish the role of HBZ in HTLV-1 replication and leukemogenesis in vivo, HU-SCID mice were infected with an infectious proviral clone lacking functional HBZ (HTLV-1ΔHBZ). HTLV-1ΔHBZ-infected HU-SCID mice developed lymphoproliferations with an immature preleukemic CD4−CD8−CD90+ phenotype starting at ~10 weeks post-reconstitution. In contrast wild type HTLV-1 infection reproducibly induces a mature CD4+CD25+ CD90− lymphoma. Lymphoma cells successfully engrafted naïve NOD/SCID mice when injected into the peritoneal cavity and these cells maintain the expression of viral proteins, gp46env and p19gag. HTLV-1 infection of CD34+ HP/HSCs and the recapitulation of a lymphoma similar to ATLL in HU-NOD/SCID mice suggest that hematopoietic stem cells provide a relevant cellular target and viral reservoir in vivo and that infection of these cells contribute to viral lymphomagenesis in humans. The HTLV-1-HU-SCID mouse model presents a compelling in vivo model to characterize molecular initiation and progression of events in the generation of ATL and to establish the role of HTLV-1 auxiliary proteins in viral pathogenesis.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1593-1593
Author(s):  
Lanelle V. Nakamura ◽  
Malini Mukherjee ◽  
Margaret A. Goodell ◽  
Debananda Pati

Abstract Abstract 1593 Introduction: Cohesin is an evolutionarily conserved protein complex that forms during the replication of sister chromatids. It is a multi-protein complex that consists of four proteins, Smc1, Smc3, Rad21, and Scc3. Resolution of sister chromatid cohesion at the onset of anaphase depends on Separase, an endopeptidase that separates sister chromatids by cleaving cohesion Rad21. A recent study suggests a new role of Cohesin proteins in gene expression and development with implications in hematopoiesis. Our data indicates that cohesin-resolving protease Separase may play a critical role in hematopoiesis. HYPOTHESIS: We hypothesize that Separase plays a role in hematopoiesis by increasing the quantity of hematopoietic stem cells (HSC). METHODS: Our experimental approach was to isolate murine long-term HSC from WT mice and mice with one mutated copy of Separase (i.e. Separase heterozygotes). In addition, in vivo competitive long term repopulation assays were used assess the function of HSC in Separase heterozyotes. RESULTS: Separase heterozygote have increased HSC numbers (p<0.05) as compared to WT mice. In addition, an improved engraftment in a competitive repopulation assay (p < 0.001) was seen in the Separase heterozyotes. Analysis of the engrafted cells demonstrated no difference between the wild type and Separase heterozygote animals, indicating the increased engraftment may be due to unique features in the primitive hematopoietic stem cells. CONCLUSION: Investigation of the mechanism for improved HSC engraftment in Separase heterozygote mice will significantly contribute to our understanding of marrow engraftment and function. Elucidating the mechanisms of hematopoietic dysregulation will provide insights into the development of life-threatening disorders such as leukemia and, in the setting of bone marrow transplant, engraftment failure. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2003 ◽  
Vol 102 (9) ◽  
pp. 3129-3135 ◽  
Author(s):  
Jonas Larsson ◽  
Ulrika Blank ◽  
Hildur Helgadottir ◽  
Jon Mar Björnsson ◽  
Mats Ehinger ◽  
...  

Abstract Studies in vitro implicate transforming growth factor β (TGF-β) as a key regulator of hematopoiesis with potent inhibitory effects on progenitor and stem cell proliferation. In vivo studies have been hampered by early lethality of knock-out mice for TGF-β isoforms and the receptors. To directly assess the role of TGF-β signaling for hematopoiesis and hematopoietic stem cell (HSC) function in vivo, we generated a conditional knock-out model in which a disruption of the TGF-β type I receptor (TβRI) gene was induced in adult mice. HSCs from induced mice showed increased proliferation recruitment when cultured as single cells under low stimulatory conditions in vitro, consistent with an inhibitory role of TGF-β in HSC proliferation. However, induced TβRI null mice show normal in vivo hematopoiesis with normal numbers and differentiation ability of hematopoietic progenitor cells. Furthermore HSCs from TβRI null mice exhibit a normal cell cycle distribution and do not differ in their ability long term to repopulate primary and secondary recipient mice following bone marrow transplantation. These findings challenge the classical view that TGF-β is an essential negative regulator of hematopoietic stem cells under physiologic conditions in vivo.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4744-4744
Author(s):  
Tina M Schnoeder ◽  
Patricia Arreba-Tutusaus ◽  
Inga Griehl ◽  
Daniel B Lipka ◽  
Florian H Heidel ◽  
...  

Abstract Abstract 4744 Erythropoiesis is a complex multistage process in which the development of red blood cells occurs through expansion and differentiation of hematopoietic stem cells (HSCs) into more committed progenitors. Regulation of survival, expansion and differentiation of erythroid progenitors is dependent on a well-coordinated cohort of transcription factors and an intricate network of finely tuned regulatory signalling pathways. In vivo and in vitro studies have highlighted erythropoietin receptor (EpoR) signaling through JAK2 tyrosine kinase as a crucial regulator of erythropoiesis. This leads to the subsequent activation of downstream effectors such as STAT5, MAPK, and PI-3K/Akt pathways. However, detailed knowledge about signalling pathways involved in EPO/EpoR induced differentiation of erythroid progenitors remain elusive. Phosphatidylinositol-specific phospholipase C gamma1 (PLCg1) is known to act as key mediator of calcium-signalling that can substitute for PI-3K/AKT signalling in oncogenic models. Moreover, its loss is associated with lack of erythropoiesis in a straight knockout mouse model. As it is tempting to speculate on the role of Plcg1/Ca-signalling downstream of EpoR/JAK in regulation of erythroid development we aimed to investigate its influence on differentiation and proliferation of hematopoietic cells in vitro and in vivo. Using different cellular models (Ba/F3, 32D) stably transfected with EpoR and wildtype JAK2 we could provide evidence that PLCg1 is a downstream target of EpoR/JAK2 signalling. Knockdown of PLCg1 led to a decreased proliferation of PLCg1-deficient cells compared to control cells whereas survival of these cells was not affected. In contrast, other downstream targets of EpoR signalling were not affected by PLCg1 knockdown. In order to assess specifically its role in erythroid development, we used the murine pro-erythroblast cell line I-11 as well as primary fetal liver cells (FLC). The I-11 cell line was isolated from p53-deficient fetal livers and is able to differentiate upon dexamethasone-/stem cell factor-withdrawal combined with erythropoietin stimulation; primary FLC were harvested at E13.5. PLCg1 knockdown by using RNA-interference technology led to a significant delay in erythroid differentiation and accumulation of immature erythroid progenitors (e.g. pro-erythroblasts) as assessed by cytology and flow cytometry technology. In addition, we tested the colony-forming potential of PLCg1-deficient I-11 and fetal liver cells compared to controls. Colony formation was significantly impaired in both - I-11 and primary FLC - when compared to control cells (shRNA-scr). We performed gene-expression analysis by Q-RT-PCR on sorted hematopoietic stem and progenitor cells and found a higher expression in MEP compared to GMP or CMP. To clarify, whether the effects of Plcg1 knockdown are restricted to erythroid development at the stage of MEP or erythroid progenitors, we aimed to investigate adult hematopoietic stem cells in erythroid development. We infected lineage-depleted/erythroid-enriched (Gr1-, B220-, CD3/4/8, CD19-/ IL7Ra- negative) bone marrow cells with either PLCg1 or control shRNA. Using flow cytometry analysis to examine differentiation we could observe a reduction of megakaryocyte/erythroid progenitor cells (MEP) in PLCg1 knockdown cells compared to control cells while development of other lineages (e.g. GMP) remained unaffected. Currently, competitive repopulation assays investigating the repopulation and differentiation capacity of hematopoietic stem cells after Plcg1 knockdown (or scr controls) are under way to explore the role of Plcg1 signalling in hematopoietic and erythroid development in vivo. Taken together, our findings presume PLCg1 to be a key regulator in erythroid development and understanding of its relevance in development and maintenance of normal hematopoiesis will be a crucial prerequisite for targeting this important pathway in myeloproliferative disease. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document