GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Society of Hematology  (11)
  • 1
    In: Blood, American Society of Hematology, Vol. 134, No. 7 ( 2019-08-15), p. 614-625
    Abstract: Oncogenic mutations confer on cells the ability to propagate indefinitely, but whether oncogenes alter the cell fate of these cells is unknown. Here, we show that the transcriptional regulator PRDM16s causes oncogenic fate conversion by transforming cells fated to form platelets and erythrocytes into myeloid leukemia stem cells (LSCs). Prdm16s expression in megakaryocyte-erythroid progenitors (MEPs), which normally lack the potential to generate granulomonocytic cells, caused AML by converting MEPs into LSCs. Prdm16s blocked megakaryocytic/erythroid potential by interacting with super enhancers and activating myeloid master regulators, including PU.1. A CRISPR dropout screen confirmed that PU.1 is required for Prdm16s-induced leukemia. Ablating PU.1 attenuated leukemogenesis and reinstated the megakaryocytic/erythroid potential of leukemic MEPs in mouse models and human AML with PRDM16 rearrangement. Thus, oncogenic PRDM16s expression gives MEPs an LSC fate by activating myeloid gene regulatory networks.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Hematology ; 2018
    In:  Blood Vol. 131, No. 6 ( 2018-02-08), p. 591-591
    In: Blood, American Society of Hematology, Vol. 131, No. 6 ( 2018-02-08), p. 591-591
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 2473-2473
    Abstract: The adult hematopoietic system is sustained by a balance of self-renewal and differentiation in a small pool of stem and progenitor cells. This balance must be maintained to ensure a continuous supply of blood cells throughout life and prevent malignancy from arising. There are many facets of epigenetic regulation that are well known to be key components of healthy and diseased hematopoiesis, such as DNA methylation and histone post-translational modifications. However, the role of histone variant incorporation in hematopoiesis remains relatively unexplored. In this study, we explore the role of histone variant H3.3 regulation in the hematopoietic system by assessing the function of the histone H3.3 chaperone, Hira. Toward this goal, we use inducible and early developmental conditional knockout (cKO) mouse models to assess the role of Hira within the hematopoietic system. Following Hira cKO early in hematopoietic development (Vav-iCre; Hirafl/fl), we find that HSPCs are unaffected in the fetal liver but deplete quickly after homing to the bone marrow. Using polyinosinic-polycytidylic (pIpC) inducible Hira cKO mice (Mx1-Cre; Hirafl/fl), we find a similarly severe depletion of HSPCs in adult mice within 1 month after Hira loss. In contrast, differentiated cells remain largely unaffected following Hira cKO, demonstrating that Hira is especially important in the hematopoietic stem and progenitor compartment. Since Hira is known to incorporate H3.3 throughout the cell cycle and not just during S-phase like H3.1/2, we hypothesized that adult HSPCs are more dependent upon Hira to regulate histone H3 dynamics since they are slowly dividing. The loss of Hira-mediated H3.3 deposition would also be particularly detrimental to the function of these cells given its association with actively transcribed and bivalent genes. To test the role of Hira in maintaining gene expression patterns, we performed bulk RNA-seq on adult HSPCs and found that hematopoietic differentiation genes are dysregulated after Hira cKO with increased erythroid lineage and decreased lymphoid lineage gene expression. We then assessed gene expression changes in Hira cKO HSPCs in a doxycycline-inducible H2B-GFP background (Mx1-Cre; Hirafl/fl; R26-M2rtTa; TetOP-H2B-GFP) to distinguish between the gene expression changes caused by Hira loss before and after cell division. In the absence of Hira-mediated H3.3 incorporation, we expect some highly expressed genes in slowly dividing adult HSPCs to be affected by Hira loss prior to cell division due to nucleosome turnover in the wake of RNA Polymerase II. At other loci, like bivalent promoters, H3.3 would be diluted after cell division by H3.1/2 during S-phase in Hira cKO HSPCs. In support of this hypothesis, we found that increased expression of the erythroid differentiation gene Klf1 in Hira cKO MPPs after cell division (H2B-GFPLow) relative to Hira cKO MPPs before division (H2B-GFPHigh) and WT MPPs that have divided (H2B-GFPLow). The findings from both of these transcriptome analyses point toward a role of Hira in regulating HSPC differentiation genes and are supported by our in vitro and in vivo data showing increased differentiation of Hira cKO HSPCs and decreased self-renewal. In order to more fully understand the H3.3-dependent gene expression changes after Hira cKO in HSPCs, we correlated H3.3 enrichment patterns from chromatin-immunoprecipitation and sequencing (ChIP-seq) with our data from assay for transposase-accessible chromatin and sequencing (ATAC-seq). Our results demonstrate that Hira cKO HSPCs have more open chromatin and fewer H3.3 peaks, suggesting that loss of Hira-mediated H3.3 deposition increases DNA accessibility. This study identifies a novel epigenetic mechanism required for adult HSPC maintenance and elucidates a previously unappreciated regulator of normal hematopoietic homeostasis. Further understanding how Hira-mediated H3.3 regulation maintains adult HSPCs will provide greater depth to our current understanding of the epigenetic regulators essential for hematopoiesis. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 8590-8591
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2019
    In:  Blood Vol. 134, No. Supplement_1 ( 2019-11-13), p. 879-879
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 879-879
    Abstract: Acute myeloid leukemia (AML) is primarily a disease of older adults with poor treatment outcomes. Despite years of intensive research, the standard induction therapy for AML has remained largely unchanged for decades. Thus, the development of new and efficacious therapeutic targets for AML is urgently needed. Leukemia cells exhibit multiple metabolic aberrations that may be therapeutically targeted. Here, we show that nicotinamide adenine dinucleotide (NAD+) promotes leukemogenesis and causes chemotherapy treatment resistance through fueling energetic metabolism, and pinpoints nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) is a novel therapeutic target for AML. To identify novel genes essential for AML, we performed a whole genome CRISPR dropout screen by using MOLM13 cell line and identified 1,951 essential genes (Fig. A). By searching druggable targets among these genes, we narrowed down to 345 genes, among which we found two genes, NMNAT1 (nicotinamide nucleotide adenylyltransferase 1) and NAMPT (nicotinamide phosphoribosyltransferase), both involved in key steps in NAD+ biosynthesis. We comprehensively analyzed dependency scores for all genes involved in the NAD+ biosynthetic pathways (de novo synthesis pathway, the Preiss-Handler pathway and the salvage pathway) across a broad panel of cancer cell lines from the Dependency Map database (https://depmap.org/portal/). The results showed that NMNAT1 and NAMPT are both strongly selective and uniquely required for hematological malignancies compared to other cancers (Fig. B). Since little success has been achieved for NAMPT inhibitors in clinical trials, our attention was drawn to NMNAT1, which encodes a nuclear localized enzyme catalyzing the final step in NAD+ biosynthesis. We confirmed that deletion of NMNAT1 in AML cells significantly reduced nuclear NAD+ level and cell viability over time while sparing normal hematopoietic progenitor cells, suggesting that NMNAT1 is targetable to AML. Overexpression of wild-type Nmnat1 but not the enzymatically inactive forms rescued NMNAT1-KO AML, indicating that the catalytic activity of NMNAT1 is required for AML. To study the role of NAD+ in AML, we first measured NAD+ levels in leukemic and normal cells, and found higher NAD+ levels in leukemia-initiating cells from a murine MLL-AF9-induced AML model compared to normal cells. Supplementation of NAD+ metabolites (NMN, NAM and NR) increased AML proliferation, enhanced glycolysis (lactate production) and oxidative phosphorylation (ATP production), resulting in chemotherapy resistance (Fig. C). Deletion of NMNAT1 sensitized AML cell to chemotherapy treatment. To study the role of NMNAT1 in leukemogenesis in vivo, we genetically deleted NMNAT1 in murine or human leukemia cells, transplanted them into recipient mice, and found that deletion of NMNAT1 reduced leukemic burden and extended leukemia-free survival (Fig. D). Finally, to reveal the molecular mechanisms underlying NMNAT1 KO-mediated cell death (increased levels of gamma-H2AX), RNA-seq and functional assay of NAD+ dependent enzymes were performed. We found that the reduction of nuclear NAD+ resulting from NMNAT1 deletion upregulated genes involved in DNA repair pathway, which may be linked to impaired PARPs and Sirtuins activity. Our findings reveal the important function of NAD+ in leukemogenesis and chemoresistance, and identify NMANT1 as a novel therapeutic target for AML. Figure Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society of Hematology ; 2019
    In:  Blood Vol. 134, No. Supplement_1 ( 2019-11-13), p. 1181-1181
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 1181-1181
    Abstract: The maintenance of the hematopoietic system by hematopoietic stem cells (HSCs) is an important topic in both clinical and basic hematology study due to their enormous therapeutic potential. The hematopoietic hierarchy has recently garnered renewed interest following the development of single-cell assays and improved strategies for genetic labeling, resulting in new hierarchical models that challenge the classical view of hematopoiesis. However, the kinetic of hematopoiesis under steady-state and stress condition and the contribution of HSCs toward steady-state hematopoiesis remain controversial and unclear. Using two tamoxifen-induced lineage tracing models, namely Fgd5-CreERT2/ROSA26-tdTomato and Krt18-CreERT2/ROSA26-tdTomato, we traced the contribution of HSCs to downstream blood cells. The initial labeling will occurred mostly within HSCs due to the largely restricted expression of Fgd5 and Krt18 in HSCs. Our preliminary data suggest that adult HSCs contribute robustly to steady-state hematopoiesis, exhibiting faster efflux toward platelets and myeloid lineages compared with lymphoid lineages. Interestingly, we found that myeloid cells and platelets were labeled rapidly after the initial HSC labeling without the appearance of the previously identified intermediate populations, suggesting the potential bypassing differentiation from HSCs to these lineages. In addition to steady-state hematopoiesis, we also investigated the response of the hematopoietic system to stress. Using the lineage tracing models mentioned above, we observed that HSCs are able to respond to stress to compensate the loss of a specific cell type, without affecting their contribution toward other lineages. For example, antibody-mediated platelet depletion resulted in a compensatory output from HSCs toward platelets. On the other hand, PHZ-induced hemolytic anemia led to an accelerated production of red blood cells. In both cases, other blood lineages remained unaffected compared to controls. In order to acquire a more comprehensive view of steady-state hematopoiesis, we combined Fgd5-CreERT2/ROSA26-tdTomato lineage tracing model with single-cell RNA-seq. To be inclusive of known and unknown HSPC populations, we sorted Lin-tdTomato+ cells at 1 week, 2 weeks and 1 month after the first tamoxifen treatment. HSCs, MPPs and HPC1/2 sorted from wild-type animals served as the reference groups. We observed the appearance of myeloid and megakaryocytic progeny as early as 1 week after the initial HSC labeling, suggesting the rapid contribution of HSCs toward these lineages. Erythroid-biased population emerged at the 2-week time point, with the expansion of myeloid progeny. Intriguingly, pseudo-time analysis revealed a relatively upstream position of erythroid-biased cluster, indicating the early emergence of erythroid progeny. The clear appearance of lymphoid progeny was absent until 1 month after the initial labeling, corresponding to the later onset of lymphoid differentiation. In summary, our results support the active role of HSCs in steady-state hematopoiesis and rapid output toward megakaryocytic, erythroid, and myeloid lineages from HSCs. Moreover, our data also provide insight into the responses of HSCs toward different stimuli, which can potentially contribute to our understanding of hematopoietic-related diseases and uncover novel treatment avenues. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood Advances, American Society of Hematology, Vol. 2, No. 11 ( 2018-06-12), p. 1220-1228
    Abstract: HSCs contribute robustly to steady-state hematopoiesis. Platelets receive extensive influx from HSCs compared with other myeloid or lymphoid cells.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 2876449-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 134, No. 24 ( 2019-12-12), p. 2183-2194
    Abstract: There is increasing evidence that the metabolic regulation of acute myeloid leukemia (AML) cell growth interacts with epigenetic pathways of gene expression and differentiation. Jiang et al link inhibition of glucose metabolism to epigenetic changes and altered transcriptional pathways in leukemic cells and demonstrate synergy between simultaneously targeting metabolism and chromatin modifiers in suppression of AML.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Society of Hematology ; 2016
    In:  Blood Vol. 128, No. 22 ( 2016-12-02), p. 2701-2701
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 2701-2701
    Abstract: Maintaining metabolic homeostasis is a fundamental requirement for cells to survive. One critical requirement is to accomplish a balance between anabolism and catabolism. AMPK regulates this balance by directly sensing AMP-to-ATP ratio and its activation during cell energy deficit promotes ATP production and inhibits ATP usage. Several pieces of evidence point AMPK as a tumor suppressor: the upstream protein, LKB1, is a well-established tumor suppressor; AMPK negatively interacts with the tumor promoting mTOR pathway; anti-diabetic drugs such as metformin with tumor preventive potential are shown to activate AMPK. However, emerging evidence accumulates to support an opposite role of AMPK in promoting tumor growth. AMPK is found to protect tumor cells from metabolic crisis through different mechanisms: autophagy induction; maintaining proper ATP levels and redox environment; histone H2B tail phosphorylation. These two contrasting findings suggested multiple facets of AMPK, thus pointing to the urge to understand mechanisms of AMPK in specific contexts. We examined the role of AMPK by deleting both α1 and α2 subunit in a mouse model of acute myeloid leukemia with t(9;11) translocation. AMPK deficiency depletes the leukemia-initiating-cell population and decreases the leukemogenic potential of these cells. In order to study the metabolic regulatory effects of AMPK, we profiled the metabolites and found AMPK deficiency associates with a decreased level of glycolytic activity and reduction of acetyl-CoA, which is the major donor for histone acetylation. Therefore, we hypothesized that AMPK can affect histone acetylation through regulating the level of acetyl-CoA, and functionally alter leukemogenic potential. We first profiled histone acetylation using western blot with antibodies targeting global histone acetylation and specific histone residues. Intriguingly, we found in AMPK-deficient cells, global histone H3 and H4 acetylation levels are decreased, as well as acetylation at specific histone residues such as H3K9, H3K27 and H4K8. To build a causal link between decreased acetyl-CoA and histone acetylation, we supplemented leukemia-initiating-cells in culture with acetyl-CoA precursors such as acetate and pyruvate. The supplementation successfully increased intracellular acetyl-CoA levels as well as histone acetylation levels. Functionally restoring the intracellular acetyl-CoA and histone acetylation increased the proliferative potential of AMPK-deficient leukemia-initiating-cells and maintained cell at a more undifferentiated state. These results suggest that AMPK regulates a set of leukemogenic genes by maintaining histone acetylation levels. We hypothesize that AMPK links metabolic status to epigenetic gene regulation to promote leukemogenesis. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society of Hematology ; 2019
    In:  Blood Vol. 134, No. Supplement_1 ( 2019-11-13), p. 638-638
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 638-638
    Abstract: Deregulation of transcription is a hallmark of acute myeloid leukemia (AML) that drives oncogenic expression programs and presents opportunities for therapeutic targeting. We hypothesized that by integrating the active enhancer chromatin landscapes with pan-cancer enhancer and genetic dependency mapping, we would be able to identify targetable tumor-specific oncogenic enhancer regulation and gain mechanistic insights into the underlying basis of the regulation. Using this approach, we find that tumor-specific super enhancers statistically demarcate tumor-specific dependency (Fig. A). In addition to several well-known AML oncogenes (e.g. MYB, CDK6, FLI1…), we find SEPHS2 to have highly AML-specific enhancer regulation and genetic dependency (Fig. B). SEPHS2 is a key component of the selenoprotein production pathway and is required for the selenocysteine incorporation during protein translation (Fig. C). Across AML cell lines and primary patient samples, we observe a large AML-specific super enhancer at the SEPHS2 locus (Fig. D) that correlates with SEPHS2 genetic dependency (Fig. E) and includes a prominent binding site for AML transcription factors (Fig. D). We confirmed that the oncogenic transcription factor MYB strongly regulates SEPHS2 expression and that SEPHS2 expression correlates with poor prognosis in AML (not shown in this figure). Selenoproteins play an important role in mediating oxidative stress. We find that SEPHS2 knockout increases oxidative stress and that antioxidant treatment rescues viability effects of SEPHS2 knockout (Fig. F, G). Across murine and human in vivo AML models, genetic perturbation of selenoprotein production pathway genes strongly delays leukemogenesis (one example in Fig. H). Other cell lines (both cancerous and non-cancerous) are minimally affected by SEPHS2 knockout, confirming specificity in AML (Fig. E). As a druggable enzyme SEPHS2 merits strong consideration as a therapeutic target. In the interim, we hypothesized that selenium dietary restriction (Fig. I) may be able to phenocopy selenoprotein pathway inhibition with a lower regulatory burden. We show that mice tolerate selenium-depleted chow with no observable effects on body weight or hematopoiesis (one example in Fig. J). Confirming our hypothesis, AML transplanted into these mice exhibit a strong delay in leukemogenesis (Fig. K). Throughout the cancer biology field, there is broad interest in understanding how best to leverage pan-cancer genetic dependency data. We find that the integration of enhancer data adds another layer of specificity and helps provide mechanistic insight into the underlying basis of oncogenic deregulation and dependency. Our identification of AML-specific enhancer regulation of selenoprotein production - which has been minimally studied in this disease - validates our unbiased approach and points to selenoprotein production as a deregulated and therapeutically-actionable metabolic axis in AML. Figure Disclosures Lin: Syros Pharmaceuticals: Equity Ownership, Patents & Royalties.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...