Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 6230 2023-06-06 15:22:27 |
2 The updated version is mainly focused on the molecular mechanisms of HSC. -3438 word(s) 2792 2023-06-09 12:28:27 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Sun, S.; Han, Y.; Lei, Y.; Yu, Y.; Dong, Y.; Chen, J. Hematopoietic stem cell: regulation and nutritional intervention. Encyclopedia. Available online: https://encyclopedia.pub/entry/45245 (accessed on 08 July 2024).
Sun S, Han Y, Lei Y, Yu Y, Dong Y, Chen J. Hematopoietic stem cell: regulation and nutritional intervention. Encyclopedia. Available at: https://encyclopedia.pub/entry/45245. Accessed July 08, 2024.
Sun, Siyuan, Yingxue Han, Yumei Lei, Yifei Yu, Yanbin Dong, Juan Chen. "Hematopoietic stem cell: regulation and nutritional intervention" Encyclopedia, https://encyclopedia.pub/entry/45245 (accessed July 08, 2024).
Sun, S., Han, Y., Lei, Y., Yu, Y., Dong, Y., & Chen, J. (2023, June 06). Hematopoietic stem cell: regulation and nutritional intervention. In Encyclopedia. https://encyclopedia.pub/entry/45245
Sun, Siyuan, et al. "Hematopoietic stem cell: regulation and nutritional intervention." Encyclopedia. Web. 06 June, 2023.
Hematopoietic stem cell: regulation and nutritional intervention
Edit

Hematopoietic stem cells (HSCs) are multipotent precursors with the unique ability to self-renew into all cell types and self-regenerate in order to resume proliferation in the blood-forming system. They are crucial for the life maintenance of bio-organisms. Investigation into the functioning of HSCs remains a prominent and dynamic area of exploration by researchers. Here we summarizes the intrinsic factors (i.e., RNA-binding protein, modulators in epigenetics and enhancer–promotor-mediated transcription) essential to the function of HSCs.

hematopoietic stem cell RNA-binding protein phase separation

1. Introduction

Hematopoietic stem cells (HSCs) are multipotent precursors with the unique ability to self-renew into all cell types and self-regenerate in order to resume proliferation in the blood-forming system. These cells were first discovered in the bone marrow. Specifically, scientists discovered that a set of stem cells showed a hematopoietic function when they were intravenously injected into normal adult mice that had received a lethal dose of radiation [1]. The transplanted cells exhibited the ability to reestablish the destroyed hematopoietic system by differentiating into enough lymphoid and myeloid cells to support life [2]. The HSC niche is perivascular, can be created by mesenchymal stromal cells and endothelial cells and is likely to be located near the trabecular bone [3]. Human diseases related to HSCs encompass a broad spectrum of disorders that impact the normal functioning and development of the blood-forming system. These diseases arise due to abnormalities or dysregulation within the HSC population, leading to various pathological conditions. Examples of HSC-related diseases include hematological malignancies such as leukemia, in which the uncontrolled proliferation and differentiation of HSCs result in the accumulation of abnormal blood cells. Since the homeostasis of HSCs determines health in humans, understanding the intrinsic regulatory factors in HSCs and the extrinsic nutritional interventions needed for human health is important.
Investigation into the functioning of HSCs remains a prominent and dynamic area of exploration by researchers. In the past, studies on the internal factors regulating HSCs have mainly focused on transcription factors; however, in recent years, the focus has changed to encompass the regulatory role of RNA-binding protein and noncoding RNA families in HSCs. Nonetheless, reviews that summarize the role that nutrients play in HSC function are rare. Nutrients, including carbohydrates, lipids, proteins, vitamins and minerals, which are either energy sources for growth and reproduction or metabolic regulators, play pivotal roles in maintaining all forms of life. Generally, organisms utilize nutrients through two mechanisms: catabolic reactions and anabolic reactions. Nutrients with large molecular weights are broken down into small molecules, and through catabolic reactions, these molecules generate energy. In addition, small molecules are the basis of larger molecules that function via anabolic reactions. The integrated regulation of both processes enables the bioactivity that supports life. Few studies have elaborated on the connection between HSC function and dietary patterns, such as a high-fat diet [4][5][6][7][8]. Moreover, basic nutrients such as the vitamins ingested daily have also been proven to promote the function of HSCs [9][10][11][12][13].

2. Factors Regulating the Homeostatic Function of Hematopoietic Stem Cells

An increasing number of regulators have been reported to change HSC function. In general, these regulators can be categorized into several groups according to their functions. 

2.1. RNA-Binding Protein

RNA-binding proteins bind specific RNAs to manipulate RNA bioactivity, which in turn regulates cell function. An increasing number of studies have revealed the regulatory functions of RNA-binding proteins in HSCs (Figure 1).
Figure 1. Different mechanisms of RNA-binding protein interacting with its target RNA. U6 biogenesis 1 (USB1) excises U6 and U6atac small nuclear RNAs, and USB1 deadenylates miRNAs to avoid their degradation. Wrong splicing of SF3B1 mutants results in nonsense-mediated decay of RNA. TRAF6 ubiquitinates of hnRNPA1, causing abnormal alternative splicing of Arhgap1. Igf2bp2 regulates translation of mRNA through Lin28b/Hmga2 pathway.
As one type of RNA-binding protein, splicing factors participate intron removal from mRNA, which enables proteins to be accurately translated. Four genes (SF3B1, SRSF2, U2AF1 and ZRSR2) encode splicing factors, and mutations in these genes are frequently reported in leukemia [14][15][16][17]. For example, a common mutation in SF3B1 (K700E) contributes to the expansion of long-term HSCs (LT-HSCs) and the phenotype acquisition of myelodysplastic syndrome (MDS). More than 80% of patients with myelodysplastic syndrome with ring sideroblasts (MDS-RS) carry mutations in the SF3B1 gene; during bone marrow transplantation, SF3B1-mutated MDS-RS HSCs differentiate into characteristic ring sideroblasts [18]. One study demonstrated that the K700E mutation contributed to abnormal 3′ splice-site selection and increased the nonsense-mediated decay of RNA [19]. Lieu et al. showed that this mutation promotes the mis-splicing of MAP3K7 and ultimately accelerates the death of erythrocytes and leads to the acquisition of the MDS phenotype [20]. In addition, mutations in SF3B1 and SRSF2 exert convergent effects by enhancing autophagy in hematopoietic stem and progenitor cells (HSPCs) and increasing the cell death rate through the hyperactivation of NF-κB signaling [21]. A study reported that heterozygous mice carrying the SRSF2 P95H mutation exhibit a significant reduction in the number of HSPCs and an increased number of HSPC differentiation defects under both steady-state conditions and after transplantation [22]. Fang et al. reported that hnRNPA1, an auxiliary splicing factor, is a substrate of TRAF6. Ubiquitination of hnRNPA1 by TRAF6 regulates the alternative splicing of Arhgap1, which is critical for the hematopoietic defects observed in TRAF6-expressing HSPCs [23].
In addition to these splicing factors, other RNA-binding proteins have also been reported to regulate the activity of HSCs. U6 biogenesis 1 (USB1) was discovered to be a 3′→5′ exoribonuclease that removes 3′-terminal uridine bases from U6 small nuclear RNA transcripts. In addition, USB1 has been recently reported to play a critical role in deadenylating microRNAs (miRNAs) and retarding their degradation, and mutations in USB1 can affect miRNA levels during blood development by inhibiting the removal of 3′-end adenylated tails, which impairs hematopoietic development [24]. Igf2bp2, which is located downstream of the Lin28b/Hmga2 pathway, is an RNA-binding protein that regulates the stability and translation of mRNA. A recent study showed that Igf2bp2-dependent gene regulation in young HSCs and a decline in its function in aged HSCs both contributed to the acquisition of distinct phenotypes associated with HSC aging [25]. Musashi 2 (Msi2), a translational inhibitor, maintains the stem cell compartment mainly by regulating the proliferation of primitive progenitors downstream in LT-HSCs. Through cell cycle and gene expression analyses, the decreased proliferation capacity of ST-HSCs and lymphoid myeloid-primed progenitors (LMPPs) was discovered in Msi2-defective mice. Moreover, HSCs with Msi2 knocked out exhibited significant defects in competitive repopulation experiments [26]. By overexpressing MSI2, one study showed that pro-self-renewal phenotypes of HSCs can be induced, resulting in a 17-fold increase in the number of ST-HSCs and a 23-fold increase in the number of LT-HSCs. Furthermore, in a comprehensive analysis of MSI2-RNA interactions, MSI2 was shown to dampen aryl hydrocarbon receptor (AHR) signaling directly in cord blood HSPCs. This regulatory effect was mediated through the posttranscriptional downregulation of key components involved in the canonical AHR pathway [27]. Zfp36l2 is a critical modulator of definitive hematopoiesis. Zfp36l2-KO mice showed a significant reduction in the levels of both definitive multilineage and hematopoietic progenitors, suggesting that Zfp36l2 functions as an essential target to control the stability of mRNA in HSPCs [28].
Taken together, these observations identify a class of RNA-binding proteins that maintain the self-renewal and differentiation of HSCs, and these RNA-binding proteins regulate the self-renewal and differentiation of HSC by regulating RNA transcription, splicing and translation. These intrinsic regulatory mechanisms provide a rich molecular basis for later nutritional interventions to extend HSC homeostasis.

2.2. Epigenetic Regulation of HSC

Several components of the m6A machinery, including METTL3, ALKBH5, a m6A eraser α-ketoglutarate-dependent dioxygenase (FTO) and the m6A reader YTH N6-methyladenosine RNA-binding protein 2 (YTHDF2), are commonly dysregulated in leukemia [29][30][31][32]. It has been reported that the m6A writer enzyme METTL3 is highly expressed in acute myeloid leukemia (AML) cells, and its loss promotes cell differentiation and decreases cell proliferation in HSPCs [30]. One study showed that either the downregulation of YTHDF2 or the activation of FTO inhibited CXCR4 decay in cord blood (CB) HSCs, promoting their homing and engraftment activity [33]. YTHDF2 is an m6A reader that recognizes m6A-modified transcripts and participates in m6A-mRNA degradation. Recent studies have revealed that YTHDF2 functions as a repressor of inflammatory pathways in HSCs and plays an essential role in long-term HSC maintenance [34][35]. Moreover, its inactivation leads to HSC expansion and ameliorates AML, while YTHDF2 deficiency exerts no effect on normal HSC function [31]. These results suggest a potential strategy to reduce leukemic stem cell (LSC) survival.
In vivo experiments showed that m6A demethylation by ALKBH5 fine-tunes the activity of splicing factor encoded by SF3B1 and other epigenetic regulators, indicating that ALKBH5 may delay HSC leukemic transformation in patients with MDS [36]. STED2, a histone H3 lysine 36 methyl-transferase, plays an important role in the pathogenesis of hematologic malignancies. A recent study showed that STED2 deficiency facilitated the self-renewal of NHD13+ HSPCs, which promoted the transformation of MDS into AML [37]. The Ψ writer PUS7 plays a dual role by modifying and activating a network of tRNA-derived small fragments (tRFs) that target the translation initiation complex. When PUS7 is inactivated in embryonic stem cells, the translational regulation mediated by tRFs is impaired, resulting in increased protein biosynthesis. Thus, the disruption of the posttranscriptional regulatory mechanism leads to a reduced commitment of hematopoietic stem cells and an increased susceptibility to MDS development in humans [38].
Highly expressed in adult HSCs, MLLT3 (also known as AF9) is a component of super elongation complex 6 and interacts with DOT1L to exert its effects. MLLT3 sustains the abundance of H3K79me2 and is an HSC maintenance factor that connects histone readers with other modification factors to regulate the expression of HSC-specific genes. Moreover, in a mouse model, the sustained expression of MLLT3 led to balanced multilineage reconstitution in both the primary and secondary recipients [39].
These epigenetic regulators regulate the division pattern of HSCs, which subsequently controls the self-renewal and differentiation of HSCs (Figure 2), thus maintaining the normal development and homeostasis of the hematopoietic system.
Figure 2. Factors involved in the regulation of different states of HSCs in epigenetics. In normal HSCs, YTHDF2, the m6A reader, maintains long-term HSCs, while α-ketoglutarate-dependent dioxygenase (FTO) acts against YTHDF2, inhibiting CXCR4 decay to promote homing and engraftment activity of HSCs. In addition, MLLT3 sustains H3K79me2 level to maintain HSCs. In the pathogenesis of hematopoietic malignancies, METTL3 increases the HSPC growth in acute myeloid leukemia (AML). ALKBH5 delays HSC leukemic transformation in myelodysplastic syndromes (MDS), and STED2 deficiency facilitates the transformation of MDS into acute myeloid leukemia (AML). Moreover, inactivation of the Ψ “writer” PUS7 also contributes to the impairment of HSCs and the development of MDS.

2.3. Other Factors Regulating HSC Dormancy and Maintenance

Generally, cells in organisms must strike a balance between self-renewal and differentiation. In a stable environment, most HSCs are in a state of quiescence, as frequent differentiation of HSCs reduces the HSC pool. HSCs maintain dormancy through the action of multiple transcription factors. SIRT7 facilitates the transcription of DNA, and its expression is reduced in aged HSCs. Researchers found that its inactivation reduced the number of HSCs in a quiescent state and compromised HSC regenerative capacity. SIRT7 upregulation increased the regenerative capacity of aged HSCs [40]. Hu et al. demonstrated that steroid receptor coactivator 3 (SRC-3) was highly expressed in HSCs. SRC-3−/− mice were used to measure the function of SRC-3, and it was revealed that SRC-3 maintained the quiescence of HSCs and removed ROS from mitochondria [41]. Adenosine-to-inosine RNA editing and the enzyme adenosine deaminase play crucial roles in hematopoietic cell development and differentiation. Specifically, antizyme inhibitor 1 (Azin1) has been observed to undergo extensive editing in hematopoietic stem and progenitor cells (HSPCs). This editing process leads to an amino acid change, resulting in the translocation of the Azin1 protein (AZI) to the nucleus. In the nucleus, AZI exhibits an enhanced binding affinity for DEAD box polypeptide 1 and modulates the expression of multiple hematopoietic regulators, ultimately promoting HSPC differentiation [42].
Zpf70 is a transcription factor that regulates the self-renewal and differentiation of hematopoietic stem cells. By using CRISPR/Cas9 technology to delete Zfp90, researchers found that Zpf70 promotes HSC self-renewal in a Hoxa9-dependent manner. Specifically, Zfp90 binds to the promoter of Hoxa9 to initiate its expression via the NURF complex [43]. Autophagy suppresses the metabolism of hematopoietic stem cells by selectively eliminating functional and healthy mitochondria. This process is crucial for maintaining the quiescent state and stemness of cells. Additionally, autophagy becomes increasingly important with age, as it preserves the regenerative capacity of aging hematopoietic stem cells [44]. HSCs with a high receptor tyrosine kinase Tie2 expression tend to be quiescent and resist undergoing apoptosis. The binding of Tie2 with its ligand angiopoietin-1 (Ang-1) maintains the long-term repopulation of HSCs in vivo [45]. The activation of mitophagy, a quality control mechanism in mitochondria, is crucial for the self-renewal and expansion of Tie2+ hematopoietic stem cells (HSCs). The PPAR (peroxisome proliferator-activated receptor)-fatty acid oxidation pathway actively promotes the expansion of Tie2+ HSCs by facilitating the recruitment of Parkin to mitochondria [46]. One study reported that the activity of chaperone-mediated autophagy (CMA) in HSCs decreased with age and showed that the genetic or pharmacological activation of CMA restored the functionality (lower intracellular ROS levels, increased PK and GAPDH activities, etc.) of HSCs in both old mice and elderly humans [47]. These findings focused on the regulatory mechanisms of HSCs mediated by enhancer–promoter interactions and provided a theoretical basis for the future exploration of HSCs in this spatial field, leading to greater resolution of the multidimensional genetic regulatory mechanisms underlying HSC production.

2.4. Phase Separation

Phase separation has recently become a hot topic since a consensus has been reached suggesting that biomolecular condensates with membranes are reaction platforms for biological functions [48]. Limited research on the influence of phase separation on HSCs has been reported, and the mechanisms established are shown in Figure 3. One study showed that the transdifferentiation of spermatogonial stem cells (SSCs) into induced neural stem-cell-like cells (iNSCs) is mediated by the methylation reader protein YTHDF1 interacting with IKBα/β mRNAs, which promotes the formation of phase-separated condensates. The formation of condensates inhibits the translation of IKBα/β mRNAs and activates nuclear factor κB (NF-κB) p65, which ultimately facilitates the transdifferentiation of SSCs mediated through the expression of Eya1 [49]. Shao et al. revealed a mechanism in embryonic stem cells (ESCs), in which promoter-associated RNAs and their binding proteins cooperatively initiate the phase separation of polymerase condensates to promote transcription. Specifically, the paraspeckle protein PSPC1 combines with RNA, which functions as a multivalent molecule. This interaction results in the formation of transcript condensates and leads to the subsequent phosphorylation and release of RNA polymerase (Pol) II, increasing the activity of the polymerase [50].
Figure 3. Several proteins participating in the formation of phase separation in stem cells. YTHDF1 interacts with IKBα/β mRNAs to form phase separation, which ultimately promotes the transformation from spermatogonial stem cells (SSC) to induced neural stem-cell-like cells (iNSC). Phase separation of OCT4 reorganizes the structure of topological-associated domains (TADs), which manipulates induced pluripotent stem cell (iPSC) generation. PSPC1 binds RNA and forms transcription condensates, increasing transcription process in embryonic stem cells (ESC).
During the development of acute promyelocytic leukemia (APL), the aberrant phase separation of PML/RARα caused by neddylation of the RARα moiety results in the failure of PML nuclear body (NB) assembly, which is essential for tumor suppression. Moreover, when PML/RARα was deneddylated, phase separation was reinitiated and functional NBs were formed, and PML/RARα-driven leukemogenesis was also impeded [51]. Phase separation has been found to influence epigenetic modifications in cancer stem cells (CSCs), specifically regulating their self-renewal activity. It has been hypothesized that phase separation promotes the tumorigenicity of CSCs through ubiquitination. For example, the mutation of speckle-type POZ protein (SPOP), which prevents the recruitment of ligase substrates, facilitates the accumulation of proto-oncoproteins and triggers the phase separation of SPOP, maintaining the ubiquitination of ubiquitin-dependent proteins [52]. OCT4 is known as a master transcription factor for somatic cell reprogramming. Wang et al. recently demonstrated that the phase separation of OCT4 contributes to topological-associated domain (TAD) reorganization, which affects the efficiency of induced pluripotent stem cell (iPSC) generation [53].
The dysregulation of protein homeostasis plays a pivotal role in the aging process of hematopoietic stem cells (HSCs). In a recent study, researchers examined the proteome of HSCs and identified crucial protease inhibitors. Their findings revealed that the genetic elimination of prolyl isomerase resulted in the accelerated aging of HSCs. Interestingly, prolyl isomerase facilitates phase separation, thereby enhancing cellular stress resistance. These discoveries highlight the significance of maintaining the protein balance in HSCs and shed light on the role of prolyl isomerase in regulating HSC aging [54]. This study linked a family of widely expressed chaperones to phase transitions and revealed that macromolecular condensation dynamics drive the aging of blood stem cells.

References

  1. Jacobson, L.O.; Simmons, E.L.; Marks, E.K.; Eldredge, J.H. Recovery from radiation injury. Science 1951, 113, 510–511.
  2. Ford, C.E.; Hamerton, J.L.; Barnes, D.W.; Loutit, J.F. Cytological identification of radiation-chimaeras. Nature 1956, 177, 452–454.
  3. Morrison, S.J.; Scadden, D.T. The bone marrow niche for haematopoietic stem cells. Nature 2014, 505, 327–334.
  4. Yin, N.; Jin, G.; Ma, Y.; Zhao, H.; Zhang, G.; Li, M.O.; Peng, M. SZT2 maintains hematopoietic stem cell homeostasis via nutrient-mediated mTORC1 regulation. J. Clin. Investig. 2022, 132, e146272.
  5. Tadokoro, Y.; Hoshii, T.; Yamazaki, S.; Eto, K.; Ema, H.; Kobayashi, M.; Ueno, M.; Ohta, K.; Arai, Y.; Hara, E.; et al. Spred1 safeguards hematopoietic homeostasis against diet-induced systemic stress. Cell Stem Cell 2018, 22, 713–725.e718.
  6. Hermetet, F.; Buffiere, A.; Aznague, A.; Pais de Barros, J.P.; Bastie, J.N.; Delva, L.; Quere, R. High-fat diet disturbs lipid raft/TGF-beta signaling-mediated maintenance of hematopoietic stem cells in mouse bone marrow. Nat. Commun. 2019, 10, 523.
  7. Luo, Y.; Chen, G.L.; Hannemann, N.; Ipseiz, N.; Kronke, G.; Bauerle, T.; Munos, L.; Wirtz, S.; Schett, G.; Bozec, A. Microbiota from obese mice regulate hematopoietic stem cell differentiation by altering the bone niche. Cell. Metab. 2015, 22, 886–894.
  8. Kamimae-Lanning, A.N.; Krasnow, S.M.; Goloviznina, N.A.; Zhu, X.; Roth-Carter, Q.R.; Levasseur, P.R.; Jeng, S.; McWeeney, S.K.; Kurre, P.; Marks, D.L. Maternal high-fat diet and obesity compromise fetal hematopoiesis. Mol. Metab. 2015, 4, 25–38.
  9. Rzepecki, P.; Barzal, J.; Sarosiek, T.; Szczylik, C. Biochemical indices for the assessment of nutritional status during hematopoietic stem cell transplantation: Are they worth using? A single center experience. Bone Marrow Transpl. 2007, 40, 567–572.
  10. Chanda, B.; Ditadi, A.; Iscove, N.N.; Keller, G. Retinoic acid signaling is essential for embryonic hematopoietic stem cell development. Cell 2013, 155, 215–227.
  11. Cabezas-Wallscheid, N.; Buettner, F.; Sommerkamp, P.; Klimmeck, D.; Ladel, L.; Thalheimer, F.B.; Pastor-Flores, D.; Roma, L.P.; Renders, S.; Zeisberger, P.; et al. Vitamin A-retinoic acid signaling regulates hematopoietic stem cell dormancy. Cell 2017, 169, 807–823.e819.
  12. Sun, X.; Cao, B.; Naval-Sanchez, M.; Pham, T.; Sun, Y.B.Y.; Williams, B.; Heazlewood, S.Y.; Deshpande, N.; Li, J.; Kraus, F.; et al. Nicotinamide riboside attenuates age-associated metabolic and functional changes in hematopoietic stem cells. Nat. Commun. 2021, 12, 2665.
  13. Agathocleous, M.; Meacham, C.E.; Burgess, R.J.; Piskounova, E.; Zhao, Z.; Crane, G.M.; Cowin, B.L.; Bruner, E.; Murphy, M.M.; Chen, W.; et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis. Nature 2017, 549, 476–481.
  14. Ghram, M.; Morris, G.; Culjkovic-Kraljacic, B.; Mars, J.C.; Gendron, P.; Skrabanek, L.; Revuelta, M.V.; Cerchietti, L.; Guzman, M.L.; Borden, K.L.B. The eukaryotic translation initiation factor eIF4E reprograms alternative splicing. EMBO J. 2023, 42, e110496.
  15. McCarter, J.G.W.; Nemirovsky, D.; Famulare, C.A.; Farnoud, N.; Mohanty, A.S.; Stone-Molloy, Z.S.; Chervin, J.; Ball, B.J.; Epstein-Peterson, Z.D.; Arcila, M.E.; et al. Interaction between myelodysplasia-related gene mutations and ontogeny in acute myeloid leukemia. Blood Adv. 2023.
  16. Willekens, C.; Laplane, L.; Dagher, T.; Benlabiod, C.; Papadopoulos, N.; Lacout, C.; Rameau, P.; Catelain, C.; Alfaro, A.; Edmond, V.; et al. SRSF2-P95H decreases JAK/STAT signaling in hematopoietic cells and delays myelofibrosis development in mice. Leukemia 2023.
  17. Wang, Y.H.; Lin, C.C.; Yao, C.Y.; Amaral, F.; Yu, S.C.; Kao, C.J.; Shih, P.T.; Hou, H.A.; Chou, W.C.; Tien, H.F. High BM plasma S100A8/A9 is associated with a perturbed microenvironment and poor prognosis in myelodysplastic syndromes. Blood Adv. 2023.
  18. Mortera-Blanco, T.; Dimitriou, M.; Woll, P.S.; Karimi, M.; Elvarsdottir, E.; Conte, S.; Tobiasson, M.; Jansson, M.; Douagi, I.; Moarii, M.; et al. SF3B1-initiating mutations in MDS-RSs target lymphomyeloid hematopoietic stem cells. Blood 2017, 130, 881–890.
  19. Obeng, E.A.; Chappell, R.J.; Seiler, M.; Chen, M.C.; Campagna, D.R.; Schmidt, P.J.; Schneider, R.K.; Lord, A.M.; Wang, L.; Gambe, R.G.; et al. Physiologic expression of Sf3b1(K700E) causes impaired erythropoiesis, aberrant splicing, and sensitivity to therapeutic spliceosome modulation. Cancer Cell 2016, 30, 404–417.
  20. Lieu, Y.K.; Liu, Z.; Ali, A.M.; Wei, X.; Penson, A.; Zhang, J.; An, X.; Rabadan, R.; Raza, A.; Manley, J.L.; et al. SF3B1 mutant-induced missplicing of MAP3K7 causes anemia in myelodysplastic syndromes. Proc. Natl. Acad. Sci. USA 2022, 119, e2111703119.
  21. Lee, S.C.; North, K.; Kim, E.; Jang, E.; Obeng, E.; Lu, S.X.; Liu, B.; Inoue, D.; Yoshimi, A.; Ki, M.; et al. Synthetic lethal and convergent biological effects of cancer-associated spliceosomal gene mutations. Cancer Cell 2018, 34, 225–241.e228.
  22. Kon, A.; Yamazaki, S.; Nannya, Y.; Kataoka, K.; Ota, Y.; Nakagawa, M.M.; Yoshida, K.; Shiozawa, Y.; Morita, M.; Yoshizato, T.; et al. Physiological Srsf2 P95H expression causes impaired hematopoietic stem cell functions and aberrant RNA splicing in mice. Blood 2018, 131, 621–635.
  23. Fang, J.; Bolanos, L.C.; Choi, K.; Liu, X.; Christie, S.; Akunuru, S.; Kumar, R.; Wang, D.; Chen, X.; Greis, K.D.; et al. Ubiquitination of hnRNPA1 by TRAF6 links chronic innate immune signaling with myelodysplasia. Nat. Immunol. 2017, 18, 236–245.
  24. Jeong, H.C.; Shukla, S.; Fok, W.C.; Huynh, T.N.; Batista, L.F.Z.; Parker, R. USB1 is a miRNA deadenylase that regulates hematopoietic development. Science 2023, 379, 901–907.
  25. Suo, M.; Rommelfanger, M.K.; Chen, Y.; Amro, E.M.; Han, B.; Chen, Z.; Szafranski, K.; Chakkarappan, S.R.; Boehm, B.O.; MacLean, A.L.; et al. Age-dependent effects of Igf2bp2 on gene regulation, function, and aging of hematopoietic stem cells in mice. Blood 2022, 139, 2653–2665.
  26. De Andrés-Aguayo, L.; Varas, F.; Kallin, E.M.; Infante, J.F.; Wurst, W.; Floss, T.; Graf, T. Musashi 2 is a regulator of the HSC compartment identified by a retroviral insertion screen and knockout mice. Blood 2011, 118, 554–564.
  27. Rentas, S.; Holzapfel, N.; Belew, M.S.; Pratt, G.; Voisin, V.; Wilhelm, B.T.; Bader, G.D.; Yeo, G.W.; Hope, K.J. Musashi-2 attenuates AHR signalling to expand human haematopoietic stem cells. Nature 2016, 532, 508–511.
  28. Stumpo, D.J.; Broxmeyer, H.E.; Ward, T.; Cooper, S.; Hangoc, G.; Chung, Y.J.; Shelley, W.C.; Richfield, E.K.; Ray, M.K.; Yoder, M.C.; et al. Targeted disruption of Zfp36l2, encoding a CCCH tandem zinc finger RNA-binding protein, results in defective hematopoiesis. Blood 2009, 114, 2401–2410.
  29. Shen, C.; Sheng, Y.; Zhu, A.C.; Robinson, S.; Jiang, X.; Dong, L.; Chen, H.; Su, R.; Yin, Z.; Li, W.; et al. RNA demethylase ALKBH5 selectively promotes tumorigenesis and cancer stem cell self-renewal in acute myeloid leukemia. Cell Stem Cell 2020, 27, 64–80.e69.
  30. Vu, L.P.; Pickering, B.F.; Cheng, Y.; Zaccara, S.; Nguyen, D.; Minuesa, G.; Chou, T.; Chow, A.; Saletore, Y.; MacKay, M.; et al. The N(6)-methyladenosine (m(6)A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat. Med. 2017, 23, 1369–1376.
  31. Paris, J.; Morgan, M.; Campos, J.; Spencer, G.J.; Shmakova, A.; Ivanova, I.; Mapperley, C.; Lawson, H.; Wotherspoon, D.A.; Sepulveda, C.; et al. Targeting the RNA m(6)A reader YTHDF2 selectively compromises cancer stem cells in acute myeloid leukemia. Cell Stem Cell 2019, 25, 137–148.e136.
  32. Li, Z.; Weng, H.; Su, R.; Weng, X.; Zuo, Z.; Li, C.; Huang, H.; Nachtergaele, S.; Dong, L.; Hu, C.; et al. FTO plays an oncogenic role in acute myeloid leukemia as a N(6)-methyladenosine RNA demethylase. Cancer Cell 2017, 31, 127–141.
  33. Wang, X.; Cooper, S.; Broxmeyer, H.E.; Kapur, R. Transient regulation of RNA methylation in human hematopoietic stem cells promotes their homing and engraftment. Leukemia 2023, 37, 453–464.
  34. Mapperley, C.; van de Lagemaat, L.N.; Lawson, H.; Tavosanis, A.; Paris, J.; Campos, J.; Wotherspoon, D.; Durko, J.; Sarapuu, A.; Choe, J.; et al. The mRNA m6A reader YTHDF2 suppresses proinflammatory pathways and sustains hematopoietic stem cell function. J. Exp. Med. 2021, 218, e20200829.
  35. Li, Z.; Qian, P.; Shao, W.; Shi, H.; He, X.C.; Gogol, M.; Yu, Z.; Wang, Y.; Qi, M.; Zhu, Y.; et al. Suppression of m(6)A reader Ythdf2 promotes hematopoietic stem cell expansion. Cell Res. 2018, 28, 904–917.
  36. Ciesla, M.; Ngoc, P.C.T.; Muthukumar, S.; Todisco, G.; Madej, M.; Fritz, H.; Dimitriou, M.; Incarnato, D.; Hellstrom-Lindberg, E.; Bellodi, C. m(6)A-driven SF3B1 translation control steers splicing to direct genome integrity and leukemogenesis. Mol. Cell 2023, 83, 1165–1179.e11.
  37. Chen, B.Y.; Song, J.; Hu, C.L.; Chen, S.B.; Zhang, Q.; Xu, C.H.; Wu, J.C.; Hou, D.; Sun, M.; Zhang, Y.L.; et al. SETD2 deficiency accelerates MDS-associated leukemogenesis via S100a9 in NHD13 mice and predicts poor prognosis in MDS. Blood 2020, 135, 2271–2285.
  38. Guzzi, N.; Cieśla, M.; Ngoc, P.C.T.; Lang, S.; Arora, S.; Dimitriou, M.; Pimková, K.; Sommarin, M.N.E.; Munita, R.; Lubas, M.; et al. Pseudouridylation of tRNA-derived fragments steers translational control in stem cells. Cell 2018, 173, 1204–1216.e1226.
  39. Calvanese, V.; Nguyen, A.T.; Bolan, T.J.; Vavilina, A.; Su, T.; Lee, L.K.; Wang, Y.; Lay, F.D.; Magnusson, M.; Crooks, G.M.; et al. MLLT3 governs human haematopoietic stem-cell self-renewal and engraftment. Nature 2019, 576, 281–286.
  40. Mohrin, M.; Shin, J.; Liu, Y.; Brown, K.; Luo, H.; Xi, Y.; Haynes, C.M.; Chen, D. Stem cell aging. A mitochondrial UPR-mediated metabolic checkpoint regulates hematopoietic stem cell aging. Science 2015, 347, 1374–1377.
  41. Hu, M.; Zeng, H.; Chen, S.; Xu, Y.; Wang, S.; Tang, Y.; Wang, X.; Du, C.; Shen, M.; Chen, F.; et al. SRC-3 is involved in maintaining hematopoietic stem cell quiescence by regulation of mitochondrial metabolism in mice. Blood 2018, 132, 911–923.
  42. Wang, F.; He, J.; Liu, S.; Gao, A.; Yang, L.; Sun, G.; Ding, W.; Li, C.Y.; Gou, F.; He, M.; et al. A comprehensive RNA editome reveals that edited Azin1 partners with DDX1 to enable hematopoietic stem cell differentiation. Blood 2021, 138, 1939–1952.
  43. Liu, T.; Kong, W.X.; Tang, X.Y.; Xu, M.; Wang, Q.H.; Zhang, B.; Hu, L.D.; Chen, H. The transcription factor Zfp90 regulates the self-renewal and differentiation of hematopoietic stem cells. Cell Death Dis. 2018, 9, 677.
  44. Ho, T.T.; Warr, M.R.; Adelman, E.R.; Lansinger, O.M.; Flach, J.; Verovskaya, E.V.; Figueroa, M.E.; Passegué, E. Autophagy maintains the metabolism and function of young and old stem cells. Nature 2017, 543, 205–210.
  45. Arai, F.; Hirao, A.; Ohmura, M.; Sato, H.; Matsuoka, S.; Takubo, K.; Ito, K.; Koh, G.Y.; Suda, T. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell 2004, 118, 149–161.
  46. Ito, K.; Turcotte, R.; Cui, J.; Zimmerman, S.E.; Pinho, S.; Mizoguchi, T.; Arai, F.; Runnels, J.M.; Alt, C.; Teruya-Feldstein, J.; et al. Self-renewal of a purified Tie2+ hematopoietic stem cell population relies on mitochondrial clearance. Science 2016, 354, 1156–1160.
  47. Dong, S.; Wang, Q.; Kao, Y.R.; Diaz, A.; Tasset, I.; Kaushik, S.; Thiruthuvanathan, V.; Zintiridou, A.; Nieves, E.; Dzieciatkowska, M.; et al. Chaperone-mediated autophagy sustains haematopoietic stem-cell function. Nature 2021, 591, 117–123.
  48. Banani, S.F.; Lee, H.O.; Hyman, A.A.; Rosen, M.K. Biomolecular condensates: Organizers of cellular biochemistry. Nat. Rev. Mol. Cell Biol. 2017, 18, 285–298.
  49. Fang, Q.; Tian, G.G.; Wang, Q.; Liu, M.; He, L.; Li, S.; Wu, J. YTHDF1 phase separation triggers the fate transition of spermatogonial stem cells by activating the IkappaB-NF-kappaB-CCND1 axis. Cell Rep. 2023, 42, 112403.
  50. Shao, W.; Bi, X.; Pan, Y.; Gao, B.; Wu, J.; Yin, Y.; Liu, Z.; Peng, M.; Zhang, W.; Jiang, X.; et al. Phase separation of RNA-binding protein promotes polymerase binding and transcription. Nat. Chem. Biol. 2022, 18, 70–80.
  51. Shao, X.; Chen, Y.; Xu, A.; Xiang, D.; Wang, W.; Du, W.; Huang, Y.; Zhang, X.; Cai, M.; Xia, Z.; et al. Deneddylation of PML/RARalpha reconstructs functional PML nuclear bodies via orchestrating phase separation to eradicate APL. Cell Death Differ. 2022, 29, 1654–1668.
  52. Xiao, C.; Wu, G.; Chen, P.; Gao, L.; Chen, G.; Zhang, H. Phase separation in epigenetics and cancer stem cells. Front. Oncol. 2022, 12, 922604.
  53. Wang, J.; Yu, H.; Ma, Q.; Zeng, P.; Wu, D.; Hou, Y.; Liu, X.; Jia, L.; Sun, J.; Chen, Y.; et al. Phase separation of OCT4 controls TAD reorganization to promote cell fate transitions. Cell Stem Cell 2021, 28, 1868–1883.e1811.
  54. Maneix, L.; Iakova, P.; Moree, S.; Sweeney, M.; Moka, N.; Yellapragada, S.V.; Catic, A. Protein phase separation in hematopoietic stem cell aging. Blood 2019, 134, 5003.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 205
Revisions: 2 times (View History)
Update Date: 09 Jun 2023
1000/1000
Video Production Service