SRC within the Lin-CD34+CD38+/Lo Population Possess Heterogeneous Migration, Repopulation, and Self-Renewal Potential.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2670-2670
Author(s):  
Joby L. McKenzie ◽  
Olga I. Gan ◽  
Monica Doedens ◽  
John E. Dick

Abstract The SCID repopulating cell (SRC) xenotransplant assay is a powerful tool for characterizing human hematopoietic stem cells. Injection of hematopoietic cells directly into the intrafemoral (IF) cavity along with injection of a neutralizing antibody against residual murine NK cells and macrophages provides additonal improvements to the method (ASH 2004). IF injection permitted identification of a novel rapid-SRC (R-SRC) within the Lin-CD34+CD38+/Lo population that generated an erythromyeloid graft within 2 weeks post-transplant (Nat Med 2003). We found that this population also provides multi-lineage engraftment at 6 weeks post-transplant raising the question of whether the R-SRC had self-renewal potential. Lentivector-mediated clonal tracking was used to determine the self-renewal capacity of the individual cells within the Lin-CD34+CD38+/Lo population. Clonal analysis in primary recipients injected by IF with 5 x 10e4 Lin-CD34+CD38+/Lo and analyzed at 6 weeks showed that a subset of clones present in the injected femur were found in other bones, indicating that some individual SRC had self-renewed in the injected femur and migrated to other hematopoietic tissues. To directly test for self-renewal of migrating and non-migrating SRC, the original injected femur and the other bones (non-injected femur, two tibias and the pelvis) from each primary mouse was injected by IF into two individual secondary mice, respectively. 1) At 6 weeks post-transplant, each cell source produced substantial secondary grafts establishing that the Lin-CD34+CD38+/Lo population contains SRC with self-renewal potential. 2) Clonal analysis revealed heterogeneous self-renewal properties of individual SRC found in the primary mice; some made major contributions to all hematopoietic territories of secondary mice while others did not engraft secondary mice. 3) Interestingly, in some cases clones were detected in secondary mice that had been below detection in the primary mouse, suggesting that upon transplant into primary mice the SRC either did not divide or if they divided they returned to quiescence. Secondary transplantation was a stimulus for their activation to produce a graft of differentiated progeny. 4) Cases were observed where an active clone was found in a secondary mouse (transplanted from the primary injected femur) that had been below detection within the primary injected femur. However, the non-injected bones from this primary mouse as well as secondary mice derived from these bones all contained that same clone. We conclude that upon IF injection this SRC underwent self-renewal divisions and some of these progeny migrated to other bones and established a graft and also self-renewed, while in the injected bone the SRC likely returned to quiescence, only to be reactivated by secondary transplant. 5) Evaluation of two secondary recipients derived from one primary injected femur at 3 weeks when the graft is mainly myeloerythroid and at 6 weeks when it is mainly B cell and myeloid demonstrated that different lineage compositions were initiated by the same stem cell. The combination of clonal marking and IF injection provides an unprecedented insight into the earliest steps of stem cell function following transplantation. Although our clonal analysis is ongoing, it appears that rapid self-renewal and migration following IF injection represents a hallmark of a primitive subclass of SRC. It is essential to gain insight into the complex composition of the human stem cell compartment to develop effective stem cell-based therapies.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 249-249 ◽  
Author(s):  
Joby L. McKenzie ◽  
Olga I. Gan ◽  
Monica Doedens ◽  
John E. Dick

Abstract The conventional NOD/SCID xenotransplant model provides a powerful tool to characterize human hematopoietic stem cells. This system relies on IV injection of transplanted cells, with subsequent circulation through the blood prior to homing to appropriate niches. Two major limitations of this model are the presence of residual host factors that resist engraftment (i.e. NK cells and macrophages) and inability to detect stem cells that are incapable of homing or surviving in the circulation. We previously showed that rapid-SRC (R-SRC) were more efficiently detected by direct intrafemoral (IF) injection compared to IV transplantation (Nat Med 2003). Additionally, others showed that depletion of NK cell activity detects a short-term repopulating cell indicating that immune recognition is also important. R-SRC are found in the Lin-CD34+CD38+/lo population and produce a robust human erythromyeloid graft 2 weeks post-transplant. R-SRC are critical for stem cell therapies that require rapid engraftment and their characterization requires an efficient assay. To determine the role of cellular resistance factors we compared human engraftment in NOD/SCID mice, NOD/SCID-B2 microglobulin-null (NOD/SCID-B2m−/−) mice that are depleted of NK cells, or we administered a neutralizing antibody against the IL-2R B-chain (CD122) to NOD/SCID mice. CD122 depletes several populations including NK cells and macrophages. 4–5 x 104 Lin-CD34+CD38lo cells purified from CB were injected IF or IV into these recipients and human engraftment was determined at 2 weeks post-transplant to assay for R-SRC. In addition to determining engraftment levels, we also used the IF assay to gain insight into migration/mobilization function of R-SRC by examining human engraftment in other bones. Human myelolymphoid (CD45+) engraftment in the injected femur (RF) was significantly higher (p<0.05) in IF injected anti-CD122 treated NOD/SCID mice compared to all other groups. Since IF NOD/SCID-B2M−/ − mice had the next highest engraftment levels, these data indicate that R-SRC are very sensitive to NK activity. However the data clearly show that CD122+ cells also play a significant role in resisting stem cell engraftment. Importantly, CD122+ cells markedly affected R-SRC migration/mobilization since there was significantly higher engraftment in non-injected bones from anti-CD122 treated mice even when compared to the NOD/SCID-B2M−/ − mice. Our previous clonal analysis showed that R-SRC that are found in non-injected bones also self-renew in the injected bone before migration. We conclude that in addition to NK cells, CD122+ cells (likely macrophages) prevent the direct engraftment of R-SRC when delivered by IF or IV injection as well as their subsequent in vivo self-renewal and/or migration. Modification to the standard NOD/SCID assay by IF injection in combination with anti-CD122 provides a powerful tool to identify novel populations of stem cells as well as insight into fundamentally important properties of stem cell biology and transplantation. Mouse (n) Injection Tissue CD45+ (%) Erythroid (CD45-CD36+glyA+ (%) Total * RF-injected rt femur;BM-noninjected lt femur, pelvis, two tibiae;glyA-glycophorinA;*-total CD45+plus CD45-erythroid engraftment NOD/SCID anti-CD122 (15) IF RF 13.2 41.4 54.6 BM 4.9 23.3 28.1 NOD/SCID anti-CD122 (18) IV RF 5.3 26.5 31.8 BM 6.6 33.5 40.1 NOD/SCID (13) IF RF 3.6 11.9 15.5 BM 0.9 2.6 3.5 NOD/SCID (15) IV RF 1.4 3.9 5.3 BM 1.3 4.2 5.5 NOD/SCID/B2M−/ − (6) IF RF 7.2 31.9 39.1 BM 1.8 7.0 8.8 NOD/SCID/B2M−/ − (9) IV RF 3.5 4.9 8.3 BM 3.4 9.4 12.8


Blood ◽  
2021 ◽  
Author(s):  
Yuqing Yang ◽  
Andrew J Kueh ◽  
Zoe Grant ◽  
Waruni Abeysekera ◽  
Alexandra L Garnham ◽  
...  

The histone acetyltransferase HBO1 (MYST2, KAT7) is indispensable for postgastrulation development, histone H3 lysine 14 acetylation (H3K14Ac) and the expression of embryonic patterning genes. In this study, we report the role of HBO1 in regulating hematopoietic stem cell function in adult hematopoiesis. We used two complementary cre-recombinase transgenes to conditionally delete Hbo1 (Mx1-Cre and Rosa26-CreERT2). Hbo1 null mice became moribund due to hematopoietic failure with pancytopenia in the blood and bone marrow two to six weeks after Hbo1 deletion. Hbo1 deleted bone marrow cells failed to repopulate hemoablated recipients in competitive transplantation experiments. Hbo1 deletion caused a rapid loss of hematopoietic progenitors (HPCs). The numbers of lineage-restricted progenitors for the erythroid, myeloid, B-and T-cell lineages were reduced. Loss of HBO1 resulted in an abnormally high rate of recruitment of quiescent hematopoietic stem cells (HSCs) into the cell cycle. Cycling HSCs produced progenitors at the expense of self-renewal, which led to the exhaustion of the HSC pool. Mechanistically, genes important for HSC functions were downregulated in HSC-enriched cell populations after Hbo1 deletion, including genes essential for HSC quiescence and self-renewal, such as Mpl, Tek(Tie-2), Gfi1b, Egr1, Tal1(Scl), Gata2, Erg, Pbx1, Meis1 and Hox9, as well as genes important for multipotent progenitor cells and lineage-specific progenitor cells, such as Gata1. HBO1 was required for H3K14Ac through the genome and particularly at gene loci required for HSC quiescence and self-renewal. Our data indicate that HBO1 promotes the expression of a transcription factor network essential for HSC maintenance and self-renewal in adult hematopoiesis.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1345-1345
Author(s):  
Erin J. Oakley ◽  
Gary Van Zant

Abstract It is well documented that both quantitative and qualitative changes in the murine hematopoietic stem cell (HSC) population occur with age. In mice, the effect of aging on stem cells is highly strain-specific, thus suggesting genetic regulation plays a role in HSC aging. We have previously mapped a quantitative trait locus (QTL) to murine Chr 2 that is associated with the variation in frequency of HSCs between aged B6 and D2 mice. In C57BL/6 (B6) mice the HSC population steadily increases with age, whereas in DBA/2 mice, this population declines. A QTL regulating the natural variation in lifespan between the two strains was mapped to the same location on mouse Chr 2, thus leading to the hypothesis that stem cell function affects longevity. B6 alleles, associated with expansion of the stem cell pool, are also associated with a ~50% increase in lifespan. Using a congenic mouse model, in which D2 alleles in the QTL interval were introgressed onto a B6 background, genome wide gene expression analyses were performed using sorted lineage negative hematopoietic cells, which are enriched for primitive stem and progenitor cells. Three variables were examined using Affymetrix M430 arrays:the effect of strain--congenic versus background;the effect of age--2 months versus 22 months; andthe effects of 2 Gy of radiation because previous studies indicated that congenic animals were highly sensitive to the effects of mild radiation compared to B6 background animals. Extensive analysis of the expression arrays pointed to a single strong candidate, the gene encoding ribosome binding protein 1 (Rrbp1). Real-time PCR was used to validate the differential expression of Rrbp1 in lineage negative, Sca-1+, c-kit+ (LSK) cells, a population highly enriched for stem and progenitor cells. Further analysis revealed the presence eight non-synonymous, coding single nucleotide polymorphisms (SNPs), and at least one of them because of its location and nature may significantly alter protein structure and function. The Rrbp1 gene consists of 23 exons in mouse and is highly conserved among mammalian species including mouse, human, and canine. The Rrbp1 protein is present on the surface of the rough endoplasmic reticulum where it tethers ribosomes to the membrane, stabilizes mRNA transcripts, and mediates translocation of nascent proteins destined for the cell secretory pathway. It is well established that the interaction of HSCs with microenvironmental niches in the bone marrow is crucial for their maintenance and self-renewal, and that this interaction is mediated in part by the molecular repertoires displayed on the cell surfaces of both HSCs and niche stromal cells. Therefore, we hypothesize that age and strain specific variation in Rrbp1, through its role in the secretory pathway, affects the molecular repertoire at the cell surface of the HSC, thus altering the way stem cells interact with their niches. This altered microenvironmental interaction could have profound effects on fundamental properties relevant to stem cell aging such as pluripotency, self-renewal, and senescence.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 611-611
Author(s):  
Erin J. Oakley ◽  
Hartmut Geiger ◽  
Gary Van Zant

Abstract It is well documented that both quantitative and qualitative changes in the murine hematopoietic stem cell (HSC) population occur with age. We have previously mapped a quantitative trait locus (QTL) to murine chromosome 2 that is associated with the variation in frequency of HSCs between aged C57BL/6 (B6) and DBA/2 (D2) mice. In B6 mice the HSC population steadily increases with age, whereas in D2 mice, this population declines. A QTL regulating the natural variation in lifespan between the two strains was mapped to the same location on mouse Chr 2, thus leading to the hypothesis that stem cell function affects longevity. B6 alleles of this locus, associated with expansion of the stem cell pool, are also associated with a ~50% increase in lifespan. In the present study, we characterize a congenic mouse model which was generated by introgressing D2 alleles in the QTL onto a B6 background. Using a surrogate assay to mimic aging, we analyzed the cell cycle, apoptotic and self-renewal capabilities of congenic and B6 HSCs and show that D2 alleles in the QTL affect the apoptotic and self-renewal capabilities of HSCs. Next, we used oligonucleotide arrays to compare the differential expression of B6 and congenic cells using a population enriched for primitive stem and progenitor cells. Three variables were examined using Affymetrix M430 arrays: the effect of strain—congenic versus background; the effect of age—2 months versus 22 months; and the effects of 2 Gy of irradiation because previous studies indicated that congenic animals were highly sensitive to the effects of mild radiation compared to B6 background animals. Extensive analysis of the expression arrays pointed to a strong candidate, the gene encoding Retinoblastoma like protein 1, otherwise known as p107. The B6 allele is associated with increased p107 expression in old HSCs therefore p107 in this context is a positive regulator of stem cell number in aged mice. Real-time PCR was used to validate the differential expression of p107 in lineage negative and lineage negative Sca-1+, c-kit+ (LSK) cells. Detailed sequence analysis of the gene revealed the presence of 4 non-synonymous, coding region single nucleotide polymorphisms (SNPs) between B6 and D2 mice, which may contribute to the differential expression of the gene and function of the protein. Perhaps most importantly, we show that overexpression of p107 in congenic HSCs increases day 21, day 28, and day 35 CAFC numbers in vivo by 2- to 4-fold, therefore confirming its role as a positive regulator of primitive progenitor populations including HSCs. These studies uncover a novel role for p107 and provide additional clues in the complex regulation of stem cell aging.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2647-2647 ◽  
Author(s):  
Linda Resar ◽  
Lingling Xian ◽  
Tait Huso ◽  
Amy Belton ◽  
Leslie Cope ◽  
...  

Abstract Introduction: Nuclear chromatin structure is a key determinant of stem cell function and cell fate, although factors that regulate this are only beginning to emerge. While High Mobility Group A1(HMGA1) chromatin remodeling proteins are among the most abundant, nonhistone chromatin binding proteins in adult stem cells (ASCs), their role in this setting has been unknown. HMGA1/2 proteins modulate gene expression by binding to DNA, bending chromatin, and recruiting transcription factor complexes to enhancers throughout the genome. The HMGA1 gene is highly expressed during embryogenesis with low or undetectable levels in mature, differentiated tissues. In cancer, HMGA1 re-expression occurs through oncogenic transcription factors, other epigenetic alterations, or in rare cases, chromosomal translocation events. Importantly, HMGA1 levels correlate with adverse clinical outcomes in diverse malignancies. We previously reported that Hmga1 transgenic mice develop leukemic transformation by inducing transcriptional networks involved in stem cell function and cell cycle progression. Methods: To elucidate the role of Hmga1 in normal development and ASCs in vivo, we generated mouse models with transgenic overexpression or deletion of Hmga1. To define the function of Hmga1 in adult stem cells (ASCs), we used gain-of-function (overexpression) and loss-of-function (silencing or genetic deletion) approaches in human and murine intestinal stem cells (ISCs) and hematopoietic stem and progenitor cells. Results:Transgenic mice overexpressing Hmga1 in ISCs develop hyperproliferation, aberrant crypt formation, and polyposis in the intestinal epithelium by expanding the ISC and niche compartments. Hmga1 enhances self-renewal in ISCs by amplifying Wnt/β-catenin signaling, inducing genes that encode both Wnt agonist receptors and downstream Wnt effectors. Surprisingly, Hmga1 also "builds" an epithelial niche by directly up-regulating Sox9 to induce Paneth cell differentiation. Paneth cells constitute the epithelial ISC niche by secreting Wnt agonists. This is the first example of Hmga1 fostering terminal differentiation to establish a stem cell niche. In human intestine, HMGA1 and SOX9 are highly correlated, and both become up-regulated in colorectal cancer. Human CD34+ cells engineered to overexpress Hmga1 expand more efficiently, while those with Hmga1 deficiency have defective proliferation and colony forming capability. Both colony number and size were decreased, and differentiation was skewed towards myeloid lineages. In mice, Hmga1 deletion causes partial embryonic lethality; over 50% of expected offspring die before mid-gestation. Those that survive develop premature aging phenotypes with early kyphosis, decreased bone density, grip strength, gait velocity, and hearing deficits. Knock-out mice also have early thymic aplasia, decreased numbers of early T-cell precursors, as well as decreased B-cell differentiation. Long-term (LT)-hematopoietic stem cells were decreased and preliminary data suggests aberrant regenerative function in serial, competitive transplant experiments.Preliminary ChIP-seq and gene expression studies in CD34+ cells suggest that Hmga1 regulates transcriptional networks involved in Wnt, JAK-STAT, and PI3K signaling. Conclusions:Our results in ASCs reveal a novel role for Hmga1 in tissue homeostasis by inducing pathways involved in Wnt and regenerative function. In ISCs, Hmga1 maintains both the stem cell pool and niche compartment whereas deregulated Hmga1 may perturb this equilibrium during carcinogenesis. Functional studies in HSCs suggest that Hmga1 also regulates self-renewal, regenerative potential, and the capacity for balanced differentiation. These findings indicate that HMGA1 is required for normal stem cell function, both during embryogenesis, and postnatally, in ASCs. Our prior work in tumor models demonstrates that a subset of HMGA1 stem cell pathways are hi-jacked by cancer cells to drive tumor progression. Together, these studies provide compelling rationale for further research to determine how to harness HMGA1 for regenerative medicine and to target it in cancer therapy. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2017 ◽  
Vol 130 (5) ◽  
pp. 619-624 ◽  
Author(s):  
Brian S. Garrison ◽  
Adrian P. Rybak ◽  
Isabel Beerman ◽  
Balthasar Heesters ◽  
Francois E. Mercier ◽  
...  

Key Points ZFP521 regulates HSC self-renewal and differentiation. ZFP521 facilitates leukemogenesis in an MLL-AF9–mediated leukemia model.


2013 ◽  
Vol 24 (16) ◽  
pp. 2582-2592 ◽  
Author(s):  
Kim-Hien T. Dao ◽  
Michael D. Rotelli ◽  
Brieanna R. Brown ◽  
Jane E. Yates ◽  
Juha Rantala ◽  
...  

Fanconi anemia hematopoietic stem cells display poor self-renewal capacity when subjected to a variety of cellular stress. This phenotype raises the question of whether the Fanconi anemia proteins are stabilized or recruited as part of a stress response and protect against stem cell loss. Here we provide evidence that FANCL, the E3 ubiquitin ligase of the Fanconi anemia pathway, is constitutively targeted for degradation by the proteasome. We confirm biochemically that FANCL is polyubiquitinated with Lys-48–linked chains. Evaluation of a series of N-terminal–deletion mutants showed that FANCL's E2-like fold may direct ubiquitination. In addition, our studies showed that FANCL is stabilized in a complex with axin1 when glycogen synthase kinase-3β is overexpressed. This result leads us to investigate the potential regulation of FANCL by upstream signaling pathways known to regulate glycogen synthase kinase-3β. We report that constitutively active, myristoylated-Akt increases FANCL protein level by reducing polyubiquitination of FANCL. Two-dimensional PAGE analysis shows that acidic forms of FANCL, some of which are phospho-FANCL, are not subject to polyubiquitination. These results indicate that a signal transduction pathway involved in self-renewal and survival of hematopoietic stem cells also functions to stabilize FANCL and suggests that FANCL participates directly in support of stem cell function.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 34-34
Author(s):  
Laura Garcia Prat ◽  
Kerstin B Kaufmann ◽  
Florin Schneiter ◽  
Veronique Voisin ◽  
Alex Murison ◽  
...  

Human long-term hematopoietic stem cells (LT-HSC) residing at the top of the hematopoietic hierarchy must meet enormous daily demand (~10e11 cells daily) while also sustaining life-long maintenance of the stem cell pool through self-renewal. This hierarchical organization is widely thought to protect LT-HSC from exhaustion by maintaining them in a quiescent and undifferentiated state, activating only in response to microenvironment signals to generate highly proliferative but more short-lived populations including short-term HSC (ST-HSC) and committed progenitors. When called upon to exit this dormant state, HSC must respond and adapt their metabolism and nutrient uptake to meet increased bioenergetic demands for cell growth and differentiation. At the same time, the events underlying cellular and metabolic activation must also be suppressed to allow LT-HSC to re-enter quiescence and ultimately maintain the LT-HSC pool through self-renewal. Thus, proper sensing of cellular output demands must be coordinated with the cell cycle and metabolic machinery of LT-HSC to balance stem cell fates and maintain hematopoietic homeostasis. However, the regulatory circuits of this demand-adapted regulation of early hematopoiesis are largely unknown. The ability of cells to receive signals or take up nutrients depends on proteins that are embedded within the plasma membrane. These proteins move to the cell's interior through endocytosis and can be degraded in the lysosomes or rerouted back to the cell surface and reused. Moreover, lysosomes are the terminal catabolic stations of the autophagy pathway that is essential for preserving stem cell function through clearance of toxic cellular components. However, little is known about the regulation and role of lysosomes in the stem cell context. Here, we describe the unexpected finding that lysosomes, whose activity is intricately balanced by TFEB and MYC, are instrumental for regulating the stemness and differentiation properties of human LT-HSC. Furthermore, we found that TFEB, which is normally implicated in stress response, induces a constitutive lysosomal flux in unperturbed LT-HSC that actively maintains quiescence, preserves self-renewal and governs lineage commitment. These effects are accompanied by endolysosomal degradation of membrane receptors, such as the transferrin receptor 1 (TfR1), pointing to a role for TFEB in coordinating how LT-HSC sense environmental changes and initiate the earliest steps of their fate transitions and lineage commitment decisions. These transitions are regulated by a TFEB/MYC dichotomy where MYC is a driver of LT-HSC anabolism and activation and counteracts TFEB function by serving as a negative transcriptional regulator of lysosomes. Moreover, our findings further suggest that active suppression of TFEB and its downstream lysosomal degradation of TfR1 within LT-HSC is required for commitment along the erythroid lineage: activation of TFEB can abolish erythroid differentiation even after lineage commitment has occurred. In summary, we uncovered a MYC-TFEB-mediated dichotomous regulation of lysosomal activity that is required to balance anabolic and catabolic processes that ultimately impact human LT-HSC fate determination. Figure Disclosures Takayanagi: Kirin Holdings Company, Ltd: Current Employment. Dick:Bristol-Myers Squibb/Celgene: Research Funding.


Blood ◽  
1988 ◽  
Vol 72 (4) ◽  
pp. 1193-1196 ◽  
Author(s):  
P Mauch ◽  
M Rosenblatt ◽  
S Hellman

A technique of irradiating the entire mouse except for one hind limb was developed to provide repeated proliferative demand on the stem cell pool. Animals received 200 cGY weekly for a total dose of 3,400 to 4,000 cGy. During irradiation, shielded bone marrow cellularity was similar to that of unirradiated controls. Shielded marrow colony- forming unit (CFUs) content increased while marrow CFUs self renewal capacity decreased as compared with unirradiated age-matched controls. Following irradiation experimental animals were monitored monthly for 10 to 12 months for marrow cellularity, CFUs content, and self renewal capacity. Shielded marrow cellularity and CFUs content remained elevated over age-matched controls throughout the period of observation. These findings are compatible with the requirement of the shielded hind limb to provide hematopoietic support for the remainder of the animal. Shielded marrow self renewal capacity, a measurement reflecting primitive hematopoietic stem cell function, remained depressed and did not recover with time. These experiments provide evidence for there being limitations on the self renewal capacity of the stem cell compartment. While the small amount of shielded marrow had sufficient capacity to support the animal its average self renewal capacity was permanently reduced.


Sign in / Sign up

Export Citation Format

Share Document