scholarly journals Genetic and Epigenetic Mechanisms That Maintain Hematopoietic Stem Cell Function

2016 ◽  
Vol 2016 ◽  
pp. 1-14 ◽  
Author(s):  
Christian Kosan ◽  
Maren Godmann

All hematopoiesis cells develop from multipotent progenitor cells. Hematopoietic stem cells (HSC) have the ability to develop into all blood lineages but also maintain their stemness. Different molecular mechanisms have been identified that are crucial for regulating quiescence and self-renewal to maintain the stem cell pool and for inducing proliferation and lineage differentiation. The stem cell niche provides the microenvironment to keep HSC in a quiescent state. Furthermore, several transcription factors and epigenetic modifiers are involved in this process. These create modifications that regulate the cell fate in a more or less reversible and dynamic way and contribute to HSC homeostasis. In addition, HSC respond in a unique way to DNA damage. These mechanisms also contribute to the regulation of HSC function and are essential to ensure viability after DNA damage. How HSC maintain their quiescent stage during the entire life is still matter of ongoing research. Here we will focus on the molecular mechanisms that regulate HSC function.

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 772-772
Author(s):  
Zhenhua Yang ◽  
Hao Jiang

Fate determination of hematopoietic stem cells (HSCs), including their maintenance and differentiation, is profoundly influenced by their metabolic state. How HSCs control their metabolism to ensure correct decision making of cell fate remains an outstanding question. Metabolism is regulated by expression and activities of many rate-limiting metabolic enzymes. Histone modifications shape many aspects of DNA-based processes including transcription and DNA damage responses (DDR). H3K4 methylation is best known for its intimate association with active transcription, and is also implicated in DDR, but its role in DDR for stem cell function is unclear. The Set1/Mll complexes comprise one of six different catalytic subunits and several shared core subunits including Dpy30. We have previously shown that Dpy30 directly facilitates genome-wide H3K4 methylation (Jiang et al., Cell 2011), and that Dpy30 knockout (KO) in mouse hematopoietic system disables differentiation and long-term maintenance HSCs (Yang et al., J Exp Med, 2016). While we have identified dysregulation of multiple genes known to be important for HSC maintenance and differentiation, it is unclear what pathways functionally mediate Dpy30's role in HSC fate determination. Our analyses revealed dysregulation of many metabolic genes upon Dpy30 loss in HSCs, prompting us to examine if and how metabolism is affected by Dpy30 loss in HSCs. We found that Dpy30 loss resulted in increased AMPK activation, suggesting a low cellular energy state. Dpy30 loss resulted in significantly decreased mitochondrial membrane potential, while mitochondrial mass was insignificantly reduced, suggesting impaired mitochondrial function in energy production upon Dpy30 loss. Moreover, Dpy30 loss resulted in significant decrease in oxygen consumption in lineage-negative hematopoietic cells. In further support of diminished oxidative phosphorylation, we also found that reactive oxygen species (ROS) was significantly reduced in all hematopoietic lineage cells upon Dpy30 loss. Consistent with the reduced energy production, glucose uptake was found to be significantly reduced in Dpy30 deficient HSCs. Interestingly, we found that the Dpy30 KO HSCs were more quiescent than control HSCs. As HSCs are usually kept quiescent and they increase oxidative phosphorylation and energy production upon activation, our results suggest that Dpy30 plays important role in enabling HSC activation by metabolic reprogramming. In addition to dysregulated energy metabolism, we also found significant increase of γ-H2AX in the Dpy30 KO lineage negative bone marrow cells, suggesting increase in DDR. As the major source of DNA damage, ROS, is decreased in Dpy30 KO HSCs, we examined if the DNA damage repair was affected and thus led to sustained DDR upon Dpy30 loss. We found that Dpy30 KO cells resolved irradiation-induced γ-H2AX foci with significantly lower efficiency, suggesting that Dpy30 and its associated H3K4 methylation is important for efficient DNA damage repair. Importantly, inhibition of DDR by ATM inhibitor partially rescued the colony formation capacity of the Dpy30 KO cells, suggesting that sustained DDR functionally mediates stem cell activities. As we also saw dramatic upregulation of CDK inhibitor p21 upon Dpy30 loss, we reasoned that increased DDR may affect stem cell activity via p21. To test this hypothesis, we have been breeding to get p21 and Dpy30 double KO mice, and will soon (within a month or so) be able to test if loss of p21 can partially rescue the functional defect of Dpy30 KO stem cells, which will demonstrate an important role of CDK inhibitors in stem cell function. Taken together, our results demonstrate that a key chromatin modulator exerts a profound control of stem cell fate determination through regulating energy metabolism and genome integrity. The functional relationship between metabolic dysregulation, DDR, and stem cell function warrants further studies. Moreover, as we previously showed a critical role of Dpy30 in leukemogenesis and Myc-driven lymphomagenesis, it will be of great interest to investigate whether and how loss or inhibition of this key epigenetic modulator affects cellular metabolism and genome integrity as part of cancer-inhibitory mechanisms. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 12-12
Author(s):  
Nan Wang ◽  
Jing Yin ◽  
Na You ◽  
Dan Guo ◽  
Yangyang Zhao ◽  
...  

The mitochondria of hematopoietic stem cell (HSC) play crucial roles in regulating cell fate and in preserving HSC functionality and survival. However, the mechanism underlying its regulation remain poorly understood. Here, we identify transcription factor TWIST1 as a novel regulator of HSC maintenance through modulating mitochondrial function. We demonstrate that Twist1 deletion results in a significantly decreased long-term HSC (LT-HSC) frequency, markedly reduced dormancy and self-renewal capacities and skewed myeloid differentiation in steady-state hematopoiesis. Twist1-deficient LT-HSC are more compromised in tolerance of irradiation and 5 fluorouracil-induced stresses, and exhibit typical phenotypes of senescence and higher levels of DNA damage and apoptosis. Mechanistically, Twist1 deficiency upregulates the expression of voltage-gated calcium channel Cacna1b in HSC, leading to noticeable increases in mitochondrial calcium levels, biogenesis, metabolic activity and reactive oxygen species production. Suppression of voltage-gated calcium channel by a calcium channel blocker largely rescues the phenotypic and functional defects in Twist1-deleted HSCs under both steady-state and stress conditions. Collectively, our data, for the first time, characterize TWIST1 as a critical regulator of HSC function acting through CACNA1B/Ca2+/mitochondria axis, and highlight the importance of Ca2+ in HSC maintenance. These observations provide new insights into the mechanisms for the control of HSC fate. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 24 (4) ◽  
pp. 551-565.e8 ◽  
Author(s):  
Giulia Schiroli ◽  
Anastasia Conti ◽  
Samuele Ferrari ◽  
Lucrezia della Volpe ◽  
Aurelien Jacob ◽  
...  

Author(s):  
Tanja Schuster ◽  
Hartmut Geiger

Septins were first described in yeast. Due to extensive research in non-yeast cells, Septins are now recognized across all species as important players in the regulation of the cytoskeleton, in the establishment of polarity, for migration, vesicular trafficking and scaffolding. Stem cells are primarily quiescent cells, and this actively maintained quiescent state is critical for proper stem cell function. Equally important though, stem cells undergo symmetric or asymmetric division, which is likely linked to the level of symmetry found in the mother stem cell. Due to the ability to organize barriers and be able to break symmetry in cells, Septins are thought to have a significant impact on organizing quiescence as well as the mode (symmetric vs asymmetric) of stem cell division to affect self-renewal versus differentiation. Mechanisms of regulating mammalian quiescence and symmetry breaking by Septins are though still somewhat elusive. Within this overview article, we summarize current knowledge on the role of Septins in stem cells ranging from yeast to mice especially with respect to quiescence and asymmetric division, with a special focus on hematopoietic stem cells.


Blood ◽  
2011 ◽  
Vol 118 (6) ◽  
pp. 1534-1543 ◽  
Author(s):  
Matilda Rehn ◽  
André Olsson ◽  
Kristian Reckzeh ◽  
Eva Diffner ◽  
Peter Carmeliet ◽  
...  

Abstract Hypoxia is emerging as an important characteristic of the hematopoietic stem cell (HSC) niche, but the molecular mechanisms contributing to quiescence, self-renewal, and survival remain elusive. Vascular endothelial growth factor A (VEGFA) is a key regulator of angiogenesis and hematopoiesis. Its expression is commonly regulated by hypoxia-inducible factors (HIF) that are functionally induced in low-oxygen conditions and that activate transcription by binding to hypoxia-response elements (HRE). Vegfa is indispensable for HSC survival, mediated by a cell-intrinsic, autocrine mechanism. We hypothesized that a hypoxic HSC microenvironment is required for maintenance or up-regulation of Vegfa expression in HSCs and therefore crucial for HSC survival. We have tested this hypothesis in the mouse model Vegfaδ/δ, where the HRE in the Vegfa promoter is mutated, preventing HIF binding. Vegfa expression was reduced in highly purified HSCs from Vegfaδ/δ mice, showing that HSCs reside in hypoxic areas. Loss of hypoxia-regulated Vegfa expression increases the numbers of phenotypically defined hematopoietic stem and progenitor cells. However, HSC function was clearly impaired when assessed in competitive transplantation assays. Our data provide further evidence that HSCs reside in a hypoxic microenvironment and demonstrate a novel way in which the hypoxic niche affects HSC fate, via the hypoxia-VEGFA axis.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. SCI-34-SCI-34
Author(s):  
Danica Chen

Abstract Cell cycle checkpoints are surveillance mechanisms in eukaryotic cells that monitor the condition of the cell, repair cellular damages, and allow the cell to progress through the various phases of the cell cycle when conditions become favorable. Recent advances in hematopoietic stem cell (HSC) biology highlight a mitochondrial metabolic checkpoint that is essential for HSCs to return to the quiescent state. As quiescent HSCs enter the cell cycle, mitochondrial biogenesis is induced, which is associated with increased mitochondrial protein folding stress and mitochondrial oxidative stress. Mitochondrial unfolded protein response and mitochondrial oxidative stress response are activated to alleviate stresses and allow HSCs to exit the cell cycle and return to quiescence. Other mitochondrial maintenance mechanisms include mitophagy and asymmetric segregation of aged mitochondria. Because loss of HSC quiescence results in the depletion of the HSC pool and compromised tissue regeneration, deciphering the molecular mechanisms that regulate the mitochondrial metabolic checkpoint in HSCs will increase our understanding of hematopoiesis and how it becomes dysregulated under pathological conditions and during aging. More broadly, this knowledge is instrumental for understanding the maintenance of cells that convert between quiescence and proliferation to support their physiological functions. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Pradeep Ramalingam ◽  
Jason M. Butler ◽  
Michael G. Poulos

Abstract Purpose of Review Hematopoietic stem cells (HSCs) sit at the top of the hierarchy that meets the daily burden of blood production. HSC maintenance relies on extrinsic cues from the bone marrow (BM) microenvironment to balance stem cell self-renewal and cell fate decisions. In this brief review, we will highlight the studies and model systems that define the centralized role of BM vascular endothelium in modulating HSC activity in health and stress. Recent Findings The BM microenvironment is composed of a diverse array of intimately associated vascular and perivascular cell types. Recent dynamic imaging studies, coupled with single-cell RNA sequencing (scRNA-seq) and functional readouts, have advanced our understanding of the HSC-supportive cell types and their cooperative mechanisms that govern stem cell fate during homeostasis, regeneration, and aging. These findings have established complex and discrete vascular microenvironments within the BM that express overlapping and unique paracrine signals that modulate HSC fate. Summary Understanding the spatial and reciprocal HSC-niche interactions and the molecular mechanisms that govern HSC activity in the BM vascular microenvironment will be integral in developing therapies aimed at ameliorating hematological disease and supporting healthy hematopoietic output.


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.


Leukemia ◽  
2018 ◽  
Vol 32 (5) ◽  
pp. 1211-1221 ◽  
Author(s):  
Juliane Mohr ◽  
Banaja P. Dash ◽  
Tina M. Schnoeder ◽  
Denise Wolleschak ◽  
Carolin Herzog ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1156-1156
Author(s):  
Matteo Marchesini ◽  
Yamini Ogoti ◽  
Irene Ganan-Gomez ◽  
Yue Wei ◽  
Carlos E. Bueso-Ramos ◽  
...  

Abstract Accumulating evidence supports the view that DNA damage checkpoints activated by telomere erosion can drive hematopoietic stem cell (HSC) decline, thereby compromising HSC self-renewal, repopulating capacity, and differentiation. However, the precise mechanisms underlying telomere dysfunction-related HSC defects are still largely unknown. In this study, we employed the inducible telomerase deficient mice TERTER/ER to molecularly define the adverse effects of wide-spread endogenous telomere dysfunction-induced DNA damage signaling on stem cell function in vivo. The HSC compartment of 3-month-old telomere dysfunctional mice (G4/G5 TERTER/ER) showed an increased expansion in the steady-state absolute number of long-term HSCs (LT-HSC) and short-term HSCs with a concomitant decrease of multipotent progenitor cells. Accordingly, telomere dysfunctional LT-HSC showed a significant decrease of the quiescence state (p=0.018) associated with an increase of cells in the G1/G2-M phase of the cell cycle (p=0.038), although the preferential accumulation of phospho-H2AX foci (p=7x10-4). Furthermore, peripheral blood analysis revealed that the total CD45.2-derived reconstitution was significantly compromised in mice competitively transplanted with G4/G5 TERTER/ER LT-HSC, which shows that they have a finite potential for self-renewal under regenerative stress. Overall, these findings suggest the existence of a telomere dysfunction-induced differentiation checkpoint, which occurs at the level of LT-HSC and is responsible for their premature exhaustion. Correspondingly, aged telomere-dysfunctional mice (n=20) showed a significant decrease in the absolute number of LT-HSC in comparison to aged mice with intact telomeres (n=10) (p=0.04). On the contrary, leukemic transformation which occurred in about 5% of G4/G5 TERTER/ER mice both in homeostatic conditions and in the setting of competitive transplantation induced a significant expansion of the HSC pool, suggesting the existence of secondary events able to overcome the decline of telomere dysfunction-induced HSC self-renewal capability. One way in which cells can balance renewal with differentiation is through the control of asymmetric and symmetric division. During asymmetric division, one daughter cell remains a stem cell, while the other becomes a committed progenitor cell. In contrast, during symmetric divisions, a stem cell divides to become two HSCs (symmetric self-renewal) or two committed cells (symmetric commitment). Asymmetric cell division involves the polarized distribution of determinants, such as Numb, within the mother cell and their unequal inheritance by each daughter cell; in contrast, symmetric division allows both daughter cells to adopt equivalent fates. To determine if telomere dysfunction-induced DNA damage was directly responsible for HSC exhaustion by altering the mechanism of HSC self-renewal versus differentiation cell fate decisions, we evaluated Numb inheritance and expression in sorted telomere dysfunctional LT-HSC (n=310 LT-HSC isolated from 12 mice) in comparison to LT-HSC with intact telomeres (n=273 LT-HSC, isolated from 7 mice) induced to proliferate in culture. Specifically, we found that the frequency of symmetric self-renewal divisions was approximately 1.5-fold lower in telomere dysfunctional LT-HSC compared with those with intact telomeres (p=0.02), with a concomitant 2-fold increase in the frequency of symmetric commitment (p=0.006). Thus, telomere dysfunction-induced DNA damage is associated with a cell-intrinsic skewing toward symmetric commitment, which leads to compromised self-renewal capability. In contrast, and consistent with our in vivo data, LT-HSC isolated from G4/G5 TERTER/ER mice in leukemic transformation preferentially underwent symmetric self-renewal divisions. Next, we performed unbiased RNA sequencing on sorted G4/G5 TERTER/ER LT-HSC induced to proliferate in vitro, which underwent to preferential symmetric commitment or symmetric self-renewal divisions. Results of these analyses will provide insights into the mechanistic basis of how telomere dysfunction-induced DNA damage drives aberrant commitment of HSC, which results in their exhaustion, whereas leukemic transformation leads to deregulated and enhanced self-renewal, which results in their expansion and suppression of normal hematopoiesis. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document