FOXC1 Is Derepressed to Functional Effect in Human Acute Myeloid Leukemia

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 889-889
Author(s):  
Tim D D Somerville ◽  
Xu Huang ◽  
James T Lynch ◽  
Gary J Spencer ◽  
Tim C P Somervaille

Abstract The identification of genes and cellular pathways that are active in acute myeloid leukemia stem cells (AML LSC) but not normal hematopoietic stem and progenitor cells (HSPC) is essential, both for the understanding of disease biology and also for their evaluation as candidate therapeutic targets. Through in silico analysis, we identified FOXC1 as expressed in approximately 15-30% of cases of human AML in both LSCs and bulk cell populations. FOXC1 is a member of the forkhead box family of transcription factors and has essential roles in mesenchymal differentiation. Reflecting murine knockout phenotypes, in patients with Axenfeld-Rieger syndrome haploinsufficiency of FOXC1due to mutation or deletion causes developmental anterior segment abnormalities of the eye. By quantitative PCR we confirmed high level expression of FOXC1 in 17% of blast cell samples from patients with AML, and medium level expression of FOXC1 in 24% of samples (cohort size n=29). Critically, FOXC1 expression was not detected in any normal human hematopoietic cell population (including prospectively FACS-purified HSC, MPP, GMP, MEP, as well as defined mature cell populations, all from normal human bone marrow). Thus while FOXC1 is not expressed in normal human hematopoiesis, it is expressed in human leukemic hematopoiesis. To investigate whether FOXC1 derepression in AML makes a functional contribution to transformation, we initially performed knockdown (KD) experiments in human THP1 AML cells (which exhibit high level FOXC1 expression). FOXC1 KD led to loss of clonogenic potential and induction of morphological and immunophenotypic differentiation and this phenotype could be rescued by forced expression of a KD-resistant version of the gene. By contrast, FOXC1 KD in normal HSPC had no effect. Thus, FOXC1 contributes to the differentiation block in human AML cells. Forced expression of FOXC1 alone in normal murine HSPC induced a transient enhancement of clonogenic potential and myeloid differentiation block in serial replating assays, and myeloid skewing in in vivotransplantation assays. It did not however result in acute leukemia. Further quantitative PCR analyses demonstrated that high level FOXC1 expression associated strongly with high level HOXA9 expression in human AML. To determine whether co-expression of HOXA9 and FOXC1 is of functional significance, murine KIT+ HSPC were retrovirally infected with either Hoxa9 alone (with empty vector, MTV) or in pairwise combinations with FOXC1 or Meis1 (hereafter referred to as Hoxa9/MTV, Hoxa9/FOXC1 and Hoxa9/Meis1 cells, respectively) and their clonogenic potential was assessed in serial replating assays. As expected, Hoxa9 overexpression strongly augmented the clonogenic potential of BM HSPC, an effect which was enhanced by co-expression of Meis1. Importantly, the co-expression of Hoxa9 and FOXC1 also significantly enhanced the clonogenic potential and myeloid differentiation block of BM HSPC versus cells overexpressing Hoxa9 alone, as determined by immunophenotyping and colony morphology. Thus, FOXC1 and HOXA9 collaborate to enhance clonogenic potential and differentiation block in HSPC. To determine whether HOXA9 and FOXC1 collaborate to initiate leukemia, Hoxa9/MTV, Hoxa9/FOXC1 and Hoxa9/Meis1 double transduced HSPC were transplanted into irradiated congenic recipients. As expected, recipients of Hoxa9/Meis1 cells developed AML more rapidly than recipients of Hoxa9 cells (median latency 57 days versus 125 days). Strikingly, despite reduced initial engraftment levels, recipients of Hoxa9/FOXC1 cells succumbed to AML significantly earlier than mice receiving Hoxa9cells (median latency 83 days versus 125 days). In every case and in each cohort, autopsy demonstrated splenomegaly and pale BM due to infiltration of donor-derived cells of myeloid immunophenotype, confirming that these animals died from AML. These data demonstrate that FOXC1 functions to accelerate and enhance the development of AML in collaboration with HOXA9. Our functional studies are consistent with a model whereby lineage-inappropriate derepression of FOXC1 in human AML contributes to oncogenic transformation. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2021 ◽  
Author(s):  
Swagata Goswami ◽  
Rajeswaran Mani ◽  
Jessica Nunes ◽  
Chi-ling Chiang ◽  
Kevan Zapolnik ◽  
...  

Dysregulated cellular differentiation is a hallmark of acute leukemogenesis. Phosphatases are widely suppressed in cancers but have not been traditionally associated with differentiation. Herein, we identified that the silencing of Protein Phosphatase 2A (PP2A) directly contributes to differentiation block in acute myeloid leukemia (AML). Gene expression and mass cytometric profiling reveal that PP2A activation modulates cell cycle and transcriptional regulators that program terminal myeloid differentiation. Using a novel pharmacological agent OSU-2S in parallel with genetic approaches, we discovered that PP2A enforces c-Myc and p21 dependent terminal differentiation, proliferation arrest and apoptosis in AML. Finally, we demonstrate that PP2A activation decreases leukemia initiating stem cells, increases leukemic blast maturation, and improves overall survival in murine Tet2-/-Flt3ITD/WT and human AML models in-vivo. Our findings identify the PP2A/c-Myc/p21 axis as a critical regulator of the differentiation/proliferation switch in AML that can be therapeutically targeted in malignancies with dysregulated maturation fate.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2669-2669
Author(s):  
Nunki Hassan ◽  
Basit Salik ◽  
Alastair Duly ◽  
Jenny Yingzi Wang

Acute myeloid leukemia (AML) is associated with high relapse rates and poor survival, with limited response to conventional cancer therapy and lacking effective targeting of highly self-renewing leukemic stem cells (LSCs). The mechanism underlying the high self-renewal activity of LSCs that determines the aggressiveness of disease remains poorly understood. Although we and others have previously demonstrated the clinical significance of aberrant WNT/β-catenin signaling in AML (Science, 327:1650-1653, 2010; Cancer Cell, 18:606-618, 2010), its pharmacologically tractable components essential for the regulation of LSC self-renewal have not yet been determined. Our studies discover, for the first time, a critical link between R-spondin (RSPO)-LGR4/HOXA9 and WNT/β-catenin pathways in AML LSCs. Microarray data analysis of 183 AML patient samples showed a significant positive correlation between expression of LGR4 and HOXA9 (r=0.546, P<0.0001). LGR4 exerted a cell-of-origin-specific function in promoting aberrant self-renewal and AML progression in vivo through cooperating with HOXA9, a poor prognostic predictor. We observed that LGR4 itself was not able to fully transform normal hematopoietic stem/progenitor cells (HSPCs), but instead cooperated with HOXA9 in HSPCs to accelerate disease onset producing a highly aggressive short latency AML in vivo. LGR4 and HOXA9 were epigenetically upregulated and their coexpression was an essential determinant of RSPO-LGR4 oncogenic activity. RSPO/WNT3 ligands could serve as stem cell growth factors to sustain myeloid differentiation block and to promote proliferation of CD34+ LSC-enriched subpopulations in primary AML patient specimens co-expressing LGR4 and HOXA9. Conversely, CRISPR/Cas9-mediated knockout of LGR4 not only suppressed RSPO/WNT3 signals and markedly decreased nuclear active β-catenin, but also reduced tumor burden in a patient-derived xenograft (PDX) mouse model of relapsed AML. Importantly, this study is the first to demonstrate that pharmacological inhibition of RSPO3-LGR4 signaling by a clinical-grade anti-RSPO3 monoclonal antibody induced LSC differentiation and consequently prevented tumor growth in AML PDX mice but did not affect normal human stem cell compartment in NSG mice. Together, our findings support a critical role for RSPO-LGR4 in the Wnt/β-catenin signaling pathway to promote AML leukemogenesis. Aberrant activation of RSPO-LGR4 is crucial for enhancing the self-renewal potential and myeloid differentiation block, which contribute to an aggressive leukemia phenotype through cooperating with HOXA9. Genetic and pharmacological targeting of this pathway impairs LSC self-renewal and survival and impedes AML development in murine models and patient-derived xenografts, highlighting the therapeutic value of targeting RSPO-LGR4 signaling in AML. References: Wang Y, et al. The Wnt/beta-catenin pathway is required for the development of leukemia stem cells in AML. Science. 2010;327:1650-1653. Yeung J, et al. Beta-catenin mediates the establishment and drug resistance of MLL leukemic stem cells. Cancer Cell. 2010;18:606-618. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Author(s):  
Saumya E Samaraweera ◽  
Paul Po-Shen Wang ◽  
Ka Leung Li ◽  
Debora A. Casolari ◽  
Jinghua Feng ◽  
...  

Blood ◽  
2021 ◽  
Author(s):  
Amanda G Davis ◽  
Daniel T. Johnson ◽  
Dinghai Zheng ◽  
Ruijia Wang ◽  
Nathan D. Jayne ◽  
...  

Post-transcriptional regulation has emerged as a driver for leukemia development and an avenue for therapeutic targeting. Among post-transcriptional processes, alternative polyadenylation (APA) is globally dysregulated across cancer types. However, limited studies have focused on the prevalence and role of APA in myeloid leukemia. Furthermore, it is poorly understood how altered poly(A) site (PAS) usage of individual genes contributes to malignancy or whether targeting global APA patterns might alter oncogenic potential. In this study, we examined global APA dysregulation in acute myeloid leukemia (AML) patients by performing 3' Region Extraction And Deep Sequencing (3'READS) on a subset of AML patient samples along with healthy hematopoietic stem and progenitor cells (HSPCs) and by analyzing publicly available data from a broad AML patient cohort. We show that patient cells exhibit global 3' untranslated region (UTR) shortening and coding sequence (CDS) lengthening due to differences in PAS usage. Among APA regulators, expression of FIP1L1, one of the core cleavage and polyadenylation factors, correlated with the degree of APA dysregulation in our 3'READS dataset. Targeting global APA by FIP1L1 knockdown reversed the global trends seen in patients. Importantly, FIP1L1 knockdown induced differentiation of t(8;21) cells by promoting 3'UTR lengthening and downregulation of the fusion oncoprotein AML1-ETO. In non-t(8;21) cells, FIP1L1 knockdown also promoted differentiation by attenuating mTORC1 signaling and reducing MYC protein levels. Our study provides mechanistic insights into the role of APA in AML pathogenesis and indicates that targeting global APA patterns can overcome the differentiation block of AML patients.


Cancers ◽  
2020 ◽  
Vol 12 (8) ◽  
pp. 2163
Author(s):  
Pei-Ching Hsiao ◽  
Jer-Hwa Chang ◽  
Wei-Jiunn Lee ◽  
Chia-Chi Ku ◽  
Meng-Ying Tsai ◽  
...  

Curcumin (CUR) has a range of therapeutic benefits against cancers, but its poor solubility and low bioavailability limit its clinical use. Demethoxycurcumin (DMC) and diphenyl difluoroketone (EF-24) are natural and synthetic curcumin analogues, respectively, with better solubilities and higher anti-carcinogenic activities in various solid tumors than CUR. However, the efficacy of these analogues against non-solid tumors, particularly in acute myeloid leukemia (AML), has not been fully investigated. Herein, we observed that both DMC and EF-24 significantly decrease the proportion of viable AML cells including HL-60, U937, and MV4-11, harboring different NRAS and Fms-like tyrosine kinase 3 (FLT3) statuses, and that EF-24 has a lower half maximal inhibitory concentration (IC50) than DMC. We found that EF-24 treatment induces several features of apoptosis, including an increase in the sub-G1 population, phosphatidylserine (PS) externalization, and significant activation of extrinsic proapoptotic signaling such as caspase-8 and -3 activation. Mechanistically, p38 mitogen-activated protein kinase (MAPK) activation is critical for EF-24-triggered apoptosis via activating protein phosphatase 2A (PP2A) to attenuate extracellular-regulated protein kinase (ERK) activities in HL-60 AML cells. In the clinic, patients with AML expressing high level of PP2A have the most favorable prognoses compared to various solid tumors. Taken together, our results indicate that EF-24 is a potential therapeutic agent for treating AML, especially for cancer types that lose the function of the PP2A tumor suppressor.


Blood ◽  
2000 ◽  
Vol 95 (12) ◽  
pp. 4011-4013 ◽  
Author(s):  
Robert Hromas ◽  
Rinah Shopnick ◽  
Hani George Jumean ◽  
Charles Bowers ◽  
Marileila Varella-Garcia ◽  
...  

Abstract AML1 is a transcriptional activator that is essential for normal hematopoietic development. It is the most frequent target for translocations in acute leukemia. We recently identified 3 patients in whom pancytopenia developed almost 50 years after high-level radiation exposure from nuclear explosions during or after World War II. In all 3 patients, acute myeloid leukemia (AML) eventually developed that had similar characteristics and clinical courses. Cytogenetics from the 3 patients revealed a t(1;21)(p36;q22), a t(18;21)(q21;q22), and a t(19;21)(q13.4;q22). By fluorescent in situ hybridization (FISH), all 3 translocations disrupted the AML1 gene. Two of theseAML1 translocations, the t(18;21) and the t(19;21), have not been reported previously. It is possible that the AML1 gene is a target for radiation-induced AML.


Sign in / Sign up

Export Citation Format

Share Document