scholarly journals RNA Binding Protein Syncrip Is Required for the Low-Output HSC By Sustaining Proteome Quality

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 296-296
Author(s):  
Florisela Herrejon Chavez ◽  
Paolo Cifani ◽  
Alli Pine ◽  
Karen L Chu ◽  
Ersilia Barin ◽  
...  

Abstract RNA binding proteins (RBPs) have been increasingly recognized as an important class of regulators of normal and malignant hematopoiesis. However, the exact function and underpinning mechanisms of the RBPs that govern hematopoietic stem cells (HSCs) remains poorly characterized. We had previously identified SYNCRIP as a critical RBP that controls leukemia stem cell program in myeloid leukemia. Here, using the novel murine genetic conditional knockout (cKO) model, we delineated the role of SYNCRIP in regulating the low-output HSC. We developed a Syncrip cKO allele and crossed Syncripf/f mice to the interferon (IFN) -a-inducible Mx-1-Cre mice to create Syncripf/f Mx-1-Cre+. We consistently obtained near complete depletion of SYNCRIP 3 weeks after two consecutive Poly(I:C) injections. We observed that SYNCRIP is dispensable for static hematopoiesis and Syncrip KO animals showed equivalent number and frequencies of stem and progenitor cells (Lin-Sca+cKit+ (LSK)- LT-HSC (CD48-CD150+); MPP1 (CD48-CD150-); MPP2 (CD48+CD150+); MPP4 (CD48-CD150-)). However, KO SyncripD/D deficient cells were outcompeted by WT Syncripf/f cells in the transplantation setting (bone marrow (BM) chimerism WT (n=9) 38% ± 7.8% vs. KO (n=9) 2.7% ± 0.8%, p<0.001 at 16 weeks post-transplant) and completely lost their ability to repopulate in secondary recipient animals (WT (n=5) 58% ± 7.4% vs. KO (n=5) 7.2 %± 2.9%, p<0.001 at 16 weeks post-transplant). These data strongly indicate that SYNCRIP is critical for maintenance of long-term self-renewal of HSCs. To decipher the effect of Syncrip deletion on the transcriptomic changes in different cell types upon Syncrip loss, we performed single cell RNA sequencing analysis (scRNA-seq) of sorted LK cells (Lin-cKit+ cells) from KO SyncripD/D (n=3) vs. WT Syncrip f/f (n=3) mice. While there is no significant change in frequencies of stem and progenitor compartments, we found defective trajectory from the HSC that is closely identified as low-output HSC based on previously performed barcoding studies. We observed a strong activation of cellular response to stress and unfolded proteins, in particular the HSF1-dependent pathways upon Syncrip depletion specifically within the HSC population. To further investigate the impacts of SYNCRIP loss in the HSC unfolded protein stress response, we evaluated unfolded proteins in cells using tetraphenylethene maleimide (TMI)-based flow cytometry. The abundance of accessible thiols in unfolded proteins, which is bound by TMI serves as a surrogate measurement for the state of the unfolded proteome. We consistently observed almost 2.5-fold increase in TMI signals specifically in LT-HSC, but not ST-HSCs or MPPs upon SYNCRIP deletion indicating that SYNCRIP is required to maintain high protein quality in HSCs. Similar results were obtained with the epichaperome probe PU-FITC, which consists of HSP90 inhibitor PU-H71 conjugated to FITC. PU-H71 selectively binds to the altered epichaperome, which reflects an accumulation of chaperon networks in an aberrant cellular stress condition. Altogether, these data further confirmed that SYNCRIP depletion tips off the proteostatic balance. To understand the molecular mechanisms underpinning the functional requirement of SYNCRIP in HSPCs, we identified 534 direct mRNA targets of SYNCRIP using hyper-TRIBE method. We performed transcriptomic and proteomic analysis of sorted LT- HSCs and LSKs respectively upon SYNCRIP deletion. We integrated these datasets and found a strong enrichment of SYNCRIP targets in control of cytoskeleton and RHO GTPase related pathways. Using immunofluorescence imaging, we confirmed that SYNCRIP deletion in HSCs resulted in 2-fold reduction in RHO GTPase CDC42 expression coupled with reduced tubulin and a loss of cellular polarity (percentage of tubulin polarized cells 56% WT vs. 40% KO). We also observed that Syncrip deficient HSCs demonstrated reduced expression of lysosomal-associated membrane protein 1 (LAMP-1) and less asymmetric distribution of LAMP1 marked lysosomes during cell division (LAMP1 asymmetric division 25% WT vs. 19% KO). Overexpression of CDC42 restored cell polarity and partly rescued ability of KO SyncripD/D to serially replate. Overall, SYNCRIP is required for maintenance of protein homeostasis and cell polarity of the reserve HSCs. Our study uncovers a new regulatory axis that controls stem cell stress responses to preserve HSC self-renewal. Disclosures No relevant conflicts of interest to declare.

2019 ◽  
Vol 116 (27) ◽  
pp. 13651-13660 ◽  
Author(s):  
Yi-Ting Su ◽  
Shun-Fat Lau ◽  
Jacque P. K. Ip ◽  
Kit Cheung ◽  
Tom H. T. Cheung ◽  
...  

Adult hippocampal neurogenesis involves the lifelong generation of neurons. The process depends on the homeostasis of the production of neurons and maintenance of the adult neural stem cell (NSC) pool. Here, we report that α2-chimaerin, a Rho GTPase-activating protein, is essential for NSC homeostasis in adult hippocampal neurogenesis. Conditional deletion of α2-chimaerin in adult NSCs resulted in the premature differentiation of NSCs into intermediate progenitor cells (IPCs), which ultimately depleted the NSC pool and impaired neuron generation. Single-cell RNA sequencing and pseudotime analyses revealed that α2-chimaerin–conditional knockout (α2-CKO) mice lacked a unique NSC subpopulation, termed Klotho-expressing NSCs, during the transition of NSCs to IPCs. Furthermore, α2-CKO led to defects in hippocampal synaptic plasticity and anxiety/depression-like behaviors in mice. Our findings collectively demonstrate that α2-chimaerin plays an essential role in adult hippocampal NSC homeostasis to maintain proper brain function.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. SCI-46-SCI-46
Author(s):  
Kristin Hope

Abstract The balance between hematopoietic stem cell (HSC) differentiation and self-renewal is central to clinical regenerative paradigms. Unravelling the precise molecular mechanisms that govern HSC fate choices will thus have far reaching consequences for the development of effective therapies for hematopoietic and immunological disorders. There is an emerging recognition that beyond transcription, HSC homeostasis is subject to post-transcriptional control by RNA-binding proteins (RBPs) that ensure precise control of gene expression by modulating mRNA splicing, polyadenylation, localization, degradation or translation. RBPs can synchronously regulate the fates of operationally similar RNAs, in what have been termed RNA regulons. We have used a variety of functional approaches, in combination with unbiased genome- and proteome-scale, methods to define the tenets that govern this regulation and to determine key downstream circuitries of stem cell-regulating RBPs whose targeting could provide the basis for novel regenerative treatments. Through loss-of-function efforts, we have identified the RBP, MSI2, as a required factor for human HSC maintenance. By contrast, at supraphysiological levels, MSI2 has a profound positive effect on human HSC self-renewal decisions. Using a combination of global profiling, including mapping MSI2's targets through cross-linking immunoprecipitation (CLIP)-seq, we show that MSI2 achieves its ex vivo self-renewal-promoting effects by directing a co-ordinated post-transcriptional repression of key targets within the aryl hydrocarbon receptor (AHR) pathway. We are currently exploring the "rules" by which MSI2 influences its downstream effects on target RNAs and how it functions, in combination with other protein interactors, to instill a putative RBP regulatory code in HSCs. HSCs exhibit highly unique epigenomes, transcriptomes and proteomes and it is this distinctive molecular landscape that provides the canvas upon which MSI2, and indeed any other HSC-specific RBP exert their post-transcriptional influence over stem cell function. As such, to decipher the bona fide RNA networks that RBPs function upon in HSCs and to understand how they influence this network to enforce self-renewal, we are working towards performing systematic studies of RBP regulons in these cells specifically. In turn these approaches are elucidating a host of RBPs and post-transcriptional control mechanisms previously unappreciated for their role in HSC control. When modulated appropriately, we can successfully tailor these post-transcriptional regulons to enforce desired HSC outputs ex vivo. In summary, approaches to elucidate key HSC-regulatory RBPs and their protein and RNA interactomes provide valuable insights into a layer of HSC control previously not well understood, and one that can be capitalized on to achieve successful HSC expansion. Disclosures No relevant conflicts of interest to declare.


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 ◽  
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 ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 440-440
Author(s):  
Sun Mi Park ◽  
Angela Thornton ◽  
Ly P. Vu ◽  
Sagar Chhangawala ◽  
Gerard Minuesa ◽  
...  

Abstract Deregulated epigenetic program is found in many cancers, and genetic aberrations of histone methyltransferases contribute to transformation in myeloid leukemias. Post-transcriptional regulation in leukemia has recently been highlighted as a novel way for maintaining the leukemia stem cell (LSC) program. We have recently demonstrated that Msi2 is required for LSC function in a murine MLL-AF9 leukemia model. We determined that MSI2 maintains the mixed-lineage leukemia (MLL) self-renewal program by interacting and retaining efficient translation of critical MLL regulated transcription factors including Hoxa9, Myc and Ikzf2. Despite extensive studies implicating Myc and Hoxa9 in leukemia, the role for Ikzf2 in myeloid leukemia is not known. Ikzf2 is a member of the Ikaros transcription factor family and regulates lymphocyte development by controlling regulatory T-cell function. Ikzf2 is highly expressed in Hematopoietic Stem Cells (HSC) and to investigate if Ikzf2 is involved in HSC function, we utilized mice that have a specific deletion of Ikzf2 in the hematopoietic system through the Vav-cre system. We found similar frequencies for different populations in the stem, progenitor and mature cells in the bone marrow of Ikzf2f/f and Ikzf2D/D mice. Colony assays of isolated Lin- Sca1+ c-Kit+ (LSK) cells from Ikzf2f/f and Ikzf2D/D mice resulted in a comparable number of myeloid progenitor colonies. Furthermore, noncompetitive transplant of Ikzf2f/f and Ikzf2D/D bone marrow cells showed similar chimerism after 34 months indicating that I kzf2D/D mice have normal HSC function and hematopoiesis. To interrogate the role of Ikzf2 in acute myeloid leukemia we utilized the MLL-AF9 retroviral transduction model. Intracellular flow cytometry showed that IKZF2 is highly expressed in the LSC population compared to the non-LSCs. We then transduced Ikzf2f/f and Ikzf2D/D LSK cells with MLL-AF9 and found that Ikzf2 deletion results in a ten-fold reduction in colony formation compared to Ikzf2f/f cells. Transplantation of transduced cells results in delayed leukemia progression with reduced disease burden. Secondary transplantation of the initiation experiment exhibited a significant delay in leukemogenesis in the Ikzf2D/D compared to the Ikzf2f/f mice (median survival of 32 and 19.5 days, respectively). The role for Ikzf2 in maintenance was assessed with an inducible puro-creER system, which resulted in 80% decrease in viable cell number within 24hrs of 4-hydroxytamoxifen (4-OHT) treatment. Flow cytometric analysis showed that the Ikzf2-deficient cells had increased apoptosis and differentiation, shown by AnnexinV/7-AAD and Mac1 expression respectively. Furthermore, inducible deletion of Ikzf2 using puro-creER system in vivo revealed that Ikzf2 deletion leads to a delay in leukemia after tamoxifen administration in mice. These results indicate that Ikzf2 is required for both leukemia initiation and maintenance. To determine a role for IKZF2 in human leukemia cells, we performed Ikzf2 knockdown experiments with shRNAs in Kasumi-1, KG1, KCL22 and MOLM13 cells. Ikzf2 depletion resulted in decreased cell growth and increased apoptosis compared to cells infected with scramble shRNA. To determine the mechanism for how IKZF2 controls leukemia cell survival and self-renewal, we performed gene expression profiling of the Ikzf2-deficient Vav-cre LSCs and demonstrated enrichment in signatures for self-renewal loss, increased differentiation, loss of Myc-regulated genes and loss for targets of Hoxa9 and Meis1. Further analysis overlapping our MSI2 HITS-CLIP data and our differentially regulated genes revealed a strong enrichment suggesting that the MSI2 bound targets are transcriptionally regulated by IKZF2. Lastly, ATAC-sequencing of Ikzf2f/f and Ikzf2D/D LSCs revealed alterations in chromatin accessibility that correlated closely with differentially expressed genes. Utilizing the ATAC-seq data we predicted that HOXA9 and MYC sites were significantly altered. We validated that MYC RNA and protein levels were reduced in both murine and human AML cell lines. In contrast to its known tumor suppressor role in hypodiploid B-ALL and T-ALL, these results suggest that Ikzf2 contributes to MLL leukemia cell initiation and maintenance. Thus, we provide evidence that Ikzf2 can regulate c-MYC expression helping in maintaining the stem cell self-renewal program in LSCs. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 714-714
Author(s):  
Tzu-Chieh Ho ◽  
James Taggart ◽  
Elianna Amin ◽  
Haiming Xu ◽  
Trevor Barlowe ◽  
...  

Abstract + The first four authors contributed equally to this project. Myelodysplastic syndromes (MDS) are a group of blood cell disorders, characterized by ineffective hematopoiesis and severe cytopenias, which often transform to acute leukemia. MDS is also considered to be a clonal stem cell disease driven by alterations that are both genetic and epigenetic. However, it remains unclear how stem cell function is dysregulated and what factors drive these alterations in MDS HSCs. MSI2 is an important RNA-binding protein in normal HSC maintenance and can promote aggressive myeloid leukemia. Our preliminary data indicate that MSI2 expression is increased in high-risk MDS compared to low-risk MDS and correlates with poor survival. In order to model the role of MSI2 in MDS, we utilized the NUP98-HOXD13 transgenic (NHD13) model, which recapitulates many salient features of MDS including, leukopenia, severe anemia, erythroid dysplasia and leukemic transformation. Despite the lethal MDS or AML disease found in primary NHD13 animals, bone marrow cells transplanted into congenic mice generate a non-lethal MDS that rarely transform. Depletion of Msi2 utilizing a conditional knockout (NHD13-Msi2f/f -MX1-Cre) reversed the MDS phenotype and after one month the diseased HSPCs were eliminated. Conversely, we found that tetracycline inducible MSI2 overexpression in the context of the NHD13 transgene (NHD13/MSI2 mice) resulted in a worse MDS disease and a fully penetrant and lethal transformation to an AML, which was further accelerated during serial transplantation. AML arising in NHD13/MSI2 mice remained dependent on sustained MSI2 overexpression as mice removed from doxycycline demonstrated improved survival. Most interestingly, MSI2 overexpression expanded and maintained a more activated (G1) MDS hematopoietic stem and progenitor compartment (HSPC) in NHD13 cells. Gene expression profiling of the LSKs (Lineagelo, c-Kit+, Sca1+) before disease progression identified 891 significant genes, of which 137 genes were up-regulated (log2 fold change > 0) and 754 genes were down-regulated (log2 fold change <= 0). Furthermore, Gene Set Enrichment Analysis (GSEA) demonstrated a more progenitor like gene expression signature, enrichment in an NRAS activated signature, and a reduced quiescent phenotype. Unsupervised hierarchical clustering of the NHD13/MSI2 LSK gene signature in MDS patients resulted in four distinct clusters. Clusters segregated MSI2 high expressing MDS patients and this "MSI2 high cluster" predicted poor survival. In summary, our findings suggest that MSI2 plays a critical functional role in the maintenance of the hematopoietic stem and progenitor compartment in MDS and highlights it as a novel therapeutic target in this disease. Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 2013 ◽  
pp. 1-12 ◽  
Author(s):  
Qishan Chen ◽  
Min Jin ◽  
Jianhua Zhu ◽  
Qingzhong Xiao ◽  
Li Zhang

Stem cells possess huge importance in developmental biology, disease modelling, cell replacement therapy, and tissue engineering in regenerative medicine because they have the remarkable potential for self-renewal and to differentiate into almost all the cell types in the human body. Elucidation of molecular mechanisms regulating stem cell potency and differentiation is essential and critical for extensive application. Heterogeneous nuclear ribonucleoproteins (hnRNPs) are modular proteins consisting of RNA-binding motifs and auxiliary domains characterized by extensive and divergent functions in nucleic acid metabolism. Multiple roles of hnRNPs in transcriptional and posttranscriptional regulation enable them to be effective gene expression regulators. More recent findings show that hnRNP proteins are crucial factors implicated in maintenance of stem cell self-renewal and pluripotency and cell differentiation. The hnRNPs interact with certain sequences in target gene promoter regions to initiate transcription. In addition, they recognize 3′UTR or 5′UTR of specific gene mRNA forming mRNP complex to regulate mRNA stability and translation. Both of these regulatory pathways lead to modulation of gene expression that is associated with stem cell proliferation, cell cycle control, pluripotency, and committed differentiation.


Stem Cells ◽  
2012 ◽  
Vol 30 (3) ◽  
pp. 452-460 ◽  
Author(s):  
Elisa Närvä ◽  
Nelly Rahkonen ◽  
Maheswara Reddy Emani ◽  
Riikka Lund ◽  
Juha-Pekka Pursiheimo ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document