Evidence of MicroRNA-29b and Sp1/NFκB-HDAC Regulatory Network for KIT Expression in KIT-Driven Acute Myeloid Leukemia (AML): Biologic and Therapeutic Implications.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 938-938
Author(s):  
Shujun Liu ◽  
Lai Chu Wu ◽  
Jiuxia Pang ◽  
Ramasamy Santhanam ◽  
Sebastian Schwind ◽  
...  

Abstract Abstract 938 KIT is a receptor tyrosine kinase (RTK) and its aberrant activities resulting from protein overexpression and/or activating mutations are associated with a number of malignancies including core binding factor (CBF) AML [e.g., patients with t(8;21) or inv(16) or molecular equivalent RUNX1/RUNX1T1 or CBFB/MYH11, respectively]. RTK inhibitors (e.g. PKC412) have been shown to suppress aberrant KIT activity and delay tumor growth, but they are active only on distinct types of KIT mutations (KITmut). Furthermore, resistance to these inhibitors, as a result of secondary mutations or KIT overexpression, is emerging. Thus, we hypothesize that direct inhibition of KIT gene transcription may be a valuable therapeutic approach to override aberrant KIT expression and activity. Here, we described the regulatory and functional role of Sp1/NFkB-miR29b feedback loop in KIT-driven leukemia that can be targeted pharmacologically. Applying chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assays (EMSA) to RUNX1/RUNX1T1-positive Kasumi-1 cells, we demonstrated that while, the Sp1/NFkB complex was enriched on KIT promoter and acted as gene transactivator thereby leading to KIT overexpression, Sp1/NFkB recruited HDAC1 and HDAC3 to miR29b regulatory region thereby epigenetically repressing miR29b. This microRNA, when expressed, targeted Sp1 and eventually decreased Sp1/NFkB-mediated gene transactivation, including that of KIT. In agreement with these, we showed that when Sp1, NFkB and HDAC1 were transiently overexpressed in Kasumi-1 cells, increased KIT expression and decreased miR29b transcription occurred. In contrast, siRNA knockdown of Sp1, NFkB and HDAC1 augmented miR29b level and decreased KIT transcription. Moreover, ectopic miR29b expression impaired Sp1/NFkB repressor complex on the promoter of endogenous miR29b, thereby resulting in re-expression of the endogenous microRNA and further inhibition of Sp1/NFkB-dependent KIT transcription. Importantly, the activity of Sp1/NFkB/HDACs complex can also be pharmacologically modulated leading to restored miR29b transcription and abrogated KIT expression. We showed that pharmacologic interference with Sp1/NFkB/HDACs using their respective inhibitors, such as bortezomib (0, 6, 20, 60 and 100nM for Sp1 and NFkB), mithramycin A (150 and 300ng/ml for Sp1), bay 11-7082 (3μM for NFkB) and OSU-HDAC42 (1μM for HDAC), upregulated miR29b at early time point (6 hours) and decreased Sp1 and in turn KIT expression in KIT overexpression cell lines (e.g., Kasumi-1) and AML patient blasts. EMSA and ChIP assay demonstrated that bortezomib or HDAC42-mediated KIT repression and miR29b upregulation occurred through the dissociation of Sp1/NFkB complex from the corresponding promoter. To further investigate the therapeutic potential of targeting KIT over-expression in leukemia, we stably expressed KIT wild type (KITwt) or KITmut (D816V) in FDC-P1 cell line (murine non-tumorigenic cells derived from myeloid precursors), and we evidenced that both KITwt and KITmut promoted cell proliferation that was overcome by bortezomib in clonogenic assay. In in vivo study, when NOD/SCID mice were engrafted with FDC-P1/KITmut cells (5×106/mouse), they developed significant splenomegaly and marrow blast infiltration through KIT overexpression. When leukemia-carrying mice were treated with bortezomib (1mg/kg) for 48 hours, we observed an obvious increase of endogenous miR29b expression and a significant reduction of KIT expression. Leukemic mice that received 1mg/kg of bortezomib twice/week for 3 weeks starting on day 21 after engraftment (n=5 mice/group) showed no evidence of splenomegaly and had a significantly longer median survival [59 days (twice/week) vs 28 days (vehicle-treated), p=0.0036], compared to vehicle-treated mice that instead showed massive splenomegaly. Cytospin of marrow and histopathology of spleen and liver showed that vehicle-treated mice displayed extensive blast infiltration that was instead absent in bortezomib-treated mice. Altogether, our study revealed a previously unrecognized protein-microRNA regulatory network whose imbalance contributes to KIT-driven leukemia. As the aberrant activity of this network is pharmacologically targetable, this discovery may be quickly translated into the clinic as a novel therapeutic approach for KIT-driven AML and other malignancies. Disclosures: No relevant conflicts of interest to declare.

1989 ◽  
Vol 9 (11) ◽  
pp. 4835-4845
Author(s):  
S J Anderson ◽  
S Miyake ◽  
D Y Loh

We identified a regulatory region of the murine V beta promoter by both in vivo and in vitro analyses. The results of transient transfection assays indicated that the dominant transcription-activating element within the V beta 8.3 promoter is the palindromic motif identified previously as the conserved V beta decamer. Elimination of this element, by linear deletion or specific mutation, reduced transcriptional activity from this promoter by 10-fold. DNase I footprinting, gel mobility shift, and methylation interference assays confirmed that the palindrome acts as the binding site of a specific nuclear factor. In particular, the V beta promoter motif functioned in vitro as a high-affinity site for a previously characterized transcription activator, ATF. A consensus cyclic AMP response element (CRE) but not a consensus AP-1 site, can substitute for the decamer in vivo. These data suggest that cyclic AMP response element-binding protein (ATF/CREB) or related proteins activate V beta transcription.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. SCI-30-SCI-30
Author(s):  
Pier Paolo Pandolfi

Abstract We will discuss exciting new data regarding the role of pseudogenes, long intergenic non-coding RNAs (lincRNAs) and miRNAs in the pathogenesis of leukemia and lymphoma, as also studied in vivo in the mouse, and their therapeutic implications. We will also focus on competing endogenous RNAs (ceRNAs), circular(circ)-ceRNAs and pseudo-ceRNAs and give important attention to how understanding their cross-talk and the ceRNA language will facilitate efforts to deconvolute ceRNA networks and their role in the pathogenesis of hematopoietic malignancies. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2368-2368
Author(s):  
Luise A de Albuquerque Simoes ◽  
Isabel Weinhäuser ◽  
Diego A Pereira-Martins ◽  
César Alexander Ortiz Rojas ◽  
Thiago Mantello Bianco ◽  
...  

Abstract Accumulating evidence suggest that the axon guidance molecules SLIT and ROBO are not only implicated in physiological process but also in cancer progression. Depending on the type of cancer the SLIT-ROBO axis can either act as a tumor suppressor gene, in which case the SLIT2 promoter site is frequently hypermethylated or as an oncogene, whereby high expression is often associated with poor prognosis. In the context of acute myeloid leukemia (AML), low expression of SLIT2 has been associated with low overall survival (OS) (Golos et al., 2019), while the functional role of SLIT2 remains largely unknown. Recently, we showed that the knockdown of SLIT2 increased cell proliferation of acute promyelocytic leukemia (APL) cells resulting in a more aggressive course of disease progression in vivo using the murine transgenic APL model (Weinhäuser et al., 2020). Here, we aimed to study the functional role of SLIT2 in a more heterogeneous disease, such as AML. Using different publicly available datasets. (GSE58477, normal karyotype blasts: 62, healthy CD34 +: 10; GSE63409, LSC: 14, HSC: 5) we detected increased methylation at the SLIT2 promoter site of AML leukemic cells compared to healthy CD34 + cells suggesting SLIT2 tumor suppressive functions. In addition, we measured decreased levels of SLIT2 in the bone marrow (BM) plasma of AML patients compared to healthy donors. To assess the biological role of SLIT2, we treated AML cell lines (KASUMI1, MV411, and MOLM13) with recombinant SLIT2 (50 ng/mL) in vitro. Administration of SLIT2 reduced AML cell growth, colony formation and induced cell cycle arrest in the G1 phase for all AML cell lines. Conversely, the knockdown of SLIT2 promoted increased THP-1 and OCI-AML3 cell proliferation. Next, we determined whether the treatment with SLIT2 could delay leukemogenesis in vivo using the AML cell line MV4-11. Engraftment was monitored by luciferase bioluminescent signal and NSGS mice were either treated with recombinant SLIT2 using a dose of 25 ng/g of body weight or vehicle (control group). SLIT2 therapy resulted in a lower disease burden, decreased leukemic infiltration in the BM and spleen, reduced spleen size, and increased OS compared to the control group (p<0.05). In conclusion, we showed that SLIT2 methylation is recurrent in AML patients and that the level of SLIT2 in the plasma of AML patients is reduced. Moreover, SLIT2 treatment appears to have a cytostatic effect on different AML cell lines delaying leukemogenesis in vivo. Overall, our study reveals the therapeutic potential of SLIT2 in hematological malignancies, which could be used as an adjuvant in the clinic. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Moataz Dowaidar

Tissue hypoxia has been found as a master regulator of alternative splicing, which can have significant clinical implications. Hypoxia-elicited AS is more common in the setting of various cancer hallmarks than other illnesses, owing to the fact that hypoxia and AS are intensively explored in cancer. However, an increasing number of hypoxia-induced AS episodes have been linked to a variety of clinical conditions, including neurological and cardiovascular disorders. Hypoxia-induced AS, of course, has its own set of markers with prognostic and therapeutic implications. Targeted regulation of hypoxia signaling with the objective of modulating hypoxia-driven AS is of great interest in some cancers. In order to design acceptable therapeutic paradigms, future research will be necessary to unravel the proper molecular pathways. Although some of the discovered molecular targets appear to have therapeutic potential, more in-vivo research is required.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 31-32
Author(s):  
Thao Trinh ◽  
James Ropa ◽  
Arafat Aljoufi ◽  
Scott Cooper ◽  
Edward F. Srour ◽  
...  

The hematopoietic system is maintained by the hematopoetic stem and progenitor cells (HSCs/HPCs), a group of rare cells that reside in a hypoxic bone marrow (BM) microenvironment. Leptin (Lep) is well-known for its neuroendocrine and immunological functions, and its receptor (Lepr) has been studied extensively in the BM niche cells. Yet, its biological implications in HSC/HPC biology remained largely unknown. In this study, we hypothesized that Lepr-expressing HSCs/HPCs are functionally and transcriptomically distinct from their negative counterparts. To test our hypothesis, we utilized both in vitro and in vivo approaches. We first employed Fluorescence-activated cell sorting (FACS) analysis to confirm expression of Lepr on HSCs/HPCs in adult mouse BM. We then isolated equal numbers of Lepr+Lineage-Sca1+cKit+ (LSK cells - a heterogenous population of long-term, short-term HSCs and multipotent HPCs) and Lepr-LSK cells from C57BL/6 (CD45.2+) mouse BM to perform colony-forming unit (CFU) assay and competitive transplantation assay, which also included using competitor cells from BoyJ (CD45.1+) unseparated BM and lethally-irradiated F1 (CD45.1+CD45.2+) as hosts. To determine whether Lepr can further hierarchize HSCs into two distinct populations, we repeated the competitive transplants using freshly isolated C57BL/6 Lepr+HSCs or Lepr-HSCs cells instead. At the end of primary transplants, whole BM were analyzed for donor chimerisms in the peripheral blood (PB) and BM as well as transplanted in a non-competitive fashion into lethally-irradiated secondary recipients. To gain mechanistic insights, we assessed homing potential as homing plays a role in increased engraftment. We also performed bulk RNA-seq using freshly sorted BM Lepr+HSCs or Lepr-HSCs to elucidate potential molecular pathways that are responsible for the differences in their functional capacity. By phenotypic studies, our FACS analyses showed that Lepr+ cells represented a smaller population within the hematopoietic compartment in the BM. However, HSCs contained a higher percentage of Lepr+ cells than other HPC populations. By functional assessments, Lepr+LSK cells were more highly enriched for colony-forming progenitor cells in CFU assay as compared to Lepr-LSK cells. Interestingly, Lepr+LSK cells exhibited more robust engraftment capability in primary transplants and substantial self-renewal capacity in secondary transplants throughout different time points in both PB and BM. In addition, Lepr+HSCs showed significantly higher donor chimerisms in PB month 1, 2, 4 and BM month 4 with similar lineage output compared to Lepr-HSCs. Higher engraftment could be due to increased homing of HSCs to the BM; however, Lepr+HSCs and Lepr-HSCs showed similar homing capacity as well as levels of surface CXCR4 expression. Molecularly, Fast Preranked Gene Set Enrichment Analysis (FGSEA) showed that Lepr+HSCs were enriched for Type-I Interferon and Interferon-gamma response pathways with Normalized Enrichment Scores of 2 or higher. Lepr+HSC transcriptomic study also revealed that these cells as compared to Lepr-HSCs expressed significantly higher levels of genes involved in megakaryopoiesis and proinflammatory immune responses including the NF-κB subunits (Rel and Relb). Interestingly, both IFN-γ and NF-κB signalings have been demonstrated to be critical for the emergence of HSCs from the hemogentic endothelium during embryonic development. In summary, although Lepr+LSK cells occupied a minor fraction compared to their negative counterparts in the BM, they possessed higher colony-forming capacity and were more highly enriched for long-term functional HSCs. In line with this, Lepr+HSCs engrafted significantly higher and self-renewed more extensively than Lepr-HSCs, suggesting that Lepr not only can be used as a marker for functional HSCs but also further differentiate HSCs into two functionally distinguishable populations. Intriguingly, Lepr+HSCs were characterized with a proinflammatory transcriptomic profile that was previously suggested to be critical for the development of HSCs in the embryo. All together, our work demonstrated that Lepr+HSCs represent a subset of highly engrafting adult BM HSCs with an embryonic-like transcriptomic signature. This can have potential therapeutic implications in the field of hematopoietic transplantation as Lepr is highly conserved between mice and human. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 338-338 ◽  
Author(s):  
Barbara Ghinassi ◽  
Leda Ferro ◽  
Stefan Kachala ◽  
Isabelle Riviere ◽  
Michel Sadelain ◽  
...  

Abstract Abstract 338 Ex vivo expanded erythroblasts (EBs) are red blood cell precursors with proliferative capacity that have the potential to serve as alternative transfusion product. In the present study, we investigated the biodistribution and persistence of human EBs expanded ex vivo from cord blood following intravenous administration to NOD/SCID/IL2Rγnull mice. In the first experiment, 107 EBs generated ex vivo from cord blood under Human Erythroid Massive Amplification (HEMA) culture conditions (Migliaccio G et al. Blood Cells Mol Dis. 2002;28:169) were labeled with CFSE and transfused via the tail vein into NOD/SCID/IL2Rγnull mice which had been bled (200 μL) 24 hrs earlier to increase erythropoietin (EPO) levels. The presence of human EBs in bone marrow (BM), spleen and blood of the transfused recipient mice was analyzed by flow cytometry for CSFE and human CD235a. At day 4, 1.5 – 5% of cells in BM and spleen of the animals were CD235apos but no human cells were detectable in blood. To clarify failure of human EBs to generate red blood cells in mice, a second cohort of mice was given 25×106 expanded EBs and sacrificed 4 days thereafter. Their tissues, including BM, liver and spleen, were examined by immunohistochemistry for expression of human markers. Human CD235apos cells were found in the spleen, representing up to 18% of total cells spleen cells of transfused recipients, but the cells were trapped inside larger CD235aneg cells, probably of murine origin. These results indicate that lodging of human EBs in the spleen, where they are probably destroyed by the macrophages, may represent a barrier to using mouse models as a surrogate assay for investigating transfused human EBs. To test this hypothesis, we analyzed the fate and biodistribution of human EBs into normal vs. splenectomized NOD/SCID/IL2Rγnull mice. Cell biodistribution was analyzed using bioluminescence imaging (BLI), following retroviral-mediated transfer of eGFP and the external Gaussia luciferase genes (Santos et al Nat Med 15: 338, 2009) into expanding cord blood-derived EBs. Cord blood-derived CD34pos cells were either expanded in HEMA culture (as control) or cultured for 3 days with TPO, SCF and FLT3L before retroviral transduction. After 3 days, the cells were cultured under HEMA conditions to induce EBs expansion. Mature EBs were detectable after 11 days of culture in the untransduced, control group and the cells expanded 67-fold. By contrast, transduced cord blood cells matured by day 5–7 and amplified only 24-fold (see Figure). Transduction efficiency, as reflected by GFP expression, was on the order of 32–50% in expanded EBs. All the recipient mice were bled 10 hrs before injection (200 μL). Half of them were splenectomized 24 hrs earlier. Mice were given 15×106 each expanded EBs together with 20 units of EPO and the cell biodistribution analyzed by imaging 24 hrs later (see Figure). In intact mice, BLI signal was virtually undetectable (other than in the tail). In contrast, in the splenectomized mice, significant signal levels were also observed in the body of the animals (see Figure). Four days after injection, mice were sacrificed and the presence of human CD235apos cells in the marrow, liver, spleen (intact animals only) and blood analyzed. CD235apos cells were detectable in the marrow (20%) and liver (6%) of the splenectomized mice while in intact animals they were mainly detected in spleen (15%). Detection of human red cells in blood, however, remained low in all the cases. Overall, we have established a model for the tracking and quantification of human EBs transfused into NOD/SCID/IL2Rγnull mice. This model will be very valuable to investigate the in vivo function and persistence of human EBs expanded under different conditions and thereby define the therapeutic potential of ex vivo generated human EBs derived from different stem cell sources. Disclosures: No relevant conflicts of interest to declare.


1989 ◽  
Vol 9 (11) ◽  
pp. 4835-4845 ◽  
Author(s):  
S J Anderson ◽  
S Miyake ◽  
D Y Loh

We identified a regulatory region of the murine V beta promoter by both in vivo and in vitro analyses. The results of transient transfection assays indicated that the dominant transcription-activating element within the V beta 8.3 promoter is the palindromic motif identified previously as the conserved V beta decamer. Elimination of this element, by linear deletion or specific mutation, reduced transcriptional activity from this promoter by 10-fold. DNase I footprinting, gel mobility shift, and methylation interference assays confirmed that the palindrome acts as the binding site of a specific nuclear factor. In particular, the V beta promoter motif functioned in vitro as a high-affinity site for a previously characterized transcription activator, ATF. A consensus cyclic AMP response element (CRE) but not a consensus AP-1 site, can substitute for the decamer in vivo. These data suggest that cyclic AMP response element-binding protein (ATF/CREB) or related proteins activate V beta transcription.


2006 ◽  
Vol 80 (21) ◽  
pp. 10506-10513 ◽  
Author(s):  
Veerasamy Ravichandran ◽  
Bruce F. Sabath ◽  
Peter N. Jensen ◽  
Sidney A. Houff ◽  
Eugene O. Major

ABSTRACT The infectious cycle of the human polyomavirus JC (JCV) is ultimately regulated in cellular nuclei at the level of viral protein expression and genomic replication. Such activity is prompted by interactions between variant nucleotide sequences within the JCV regulatory region (promoter) and cellular transcription factors that bind specific DNA consensus sites. In previous work we identified an NF-1 class member, NF-1X, as a critical transcription factor affecting the JCV cellular host range. Within variant JCV promoters, as well as other viral and cellular promoters, adjacently located NF-1 and AP-1 consensus sites are often found. The close proximity of these two binding sites suggests the opportunity for interaction between NF-1 and AP-1 proteins. Here, by electrophoretic mobility shift assays, we show temporal and dose-dependent interference by an AP-1 family member, c-Jun, upon NF-1 proteins binding an NF-1 consensus site derived from JCV promoter sequence. Moreover, as demonstrated by protein-protein interaction assays, we identify specific binding affinity independent of DNA binding between NF-1X and c-Jun. Finally, to compare the binding profiles of NF-1X and c-Jun on JCV promoter sequence in parallel with in vivo detection of viral activity levels, we developed an anchored transcriptional promoter (ATP) assay. With use of extracts from JCV-infected cells transfected to overexpress either NF-1X or c-Jun, ATP assays showed concurrent increases in NF-1X binding and viral protein expression. Conversely, increased c-Jun binding accompanied decreases in both NF-1X binding and viral protein expression. Therefore, inhibition of NF-1X binding by c-Jun appears to play a role in regulating levels of JCV activity.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 159-159
Author(s):  
Margaret F. Bennewitz ◽  
Ravi Vats ◽  
Egemen Tutuncuoglu ◽  
Mark T. Gladwin ◽  
Prithu Sundd

Abstract Background: Vaso-occlusive crisis (VOC) is the primary reason for emergency medical care by sickle cell disease (SCD) patients. SCD patients hospitalized with VOC often develop acute chest syndrome (ACS), a form of acute lung injury, suggesting a role for pulmonary vaso-occlusion in the onset of ACS. However, the cellular, molecular and biophysical mechanism of pulmonary vaso-occlusion is unknown. Methods: SCD transgenic or non-sickle control mice were intravenously (IV) challenged with 2 to 3 ng of bacterial lipopolysaccharide (LPS). Fluorescent anti-mouse Ly-6G and CD49b mAbs were administered IV for in vivo staining of circulating neutrophils and platelets, respectively. Multiphoton excitation enabled quantitative fluorescence intravital lung microscopy (qFILM) was used to determine the molecular mechanism of pulmonary vaso-occlusion in live mice. Function-blocking anti-mouse P-selectin mAb (Fab fragments) was administered IV to assess the role of platelet P-selectin in promoting pulmonary vaso-occlusion. Results: A nanogram dose of IV LPS selectively triggered pulmonary vaso-occlusion in SCD but not control mice. Remarkably, pulmonary vaso-occlusion involved occlusion of the pre-capillary pulmonary arteriole bottle-necks (junction of an arteriole and capillaries) by large neutrophil-platelet embolic aggregates. IV administration of Fab fragments of function blocking anti-P-selectin mAb led to the resolution of pulmonary vaso-occlusion, which was primarily mediated by the attenuation of large neutrophil-platelet aggregates into smaller aggregates that are not stopped by the arteriolar bottle-necks. Conclusion: These results establish the relevance of neutrophil-platelet aggregation in pulmonary arterioles in promoting pulmonary vaso-occlusion in SCD and also highlight the therapeutic potential of inhibiting platelet P-selectin to prevent ACS in SCD patients hospitalized with VOC. Acknowledgments: This study was supported by 1R01HL128297-01 (P.S.), AHA 11SDG7340005 (P.S.), VMI startup funds (P.S.). M.F.B. was supported by NIH-NHLBI training grant T32HL110849 and NIH-NHLBI F32 NRSA 1F32HL131216-01. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 67 (2) ◽  
pp. 161-165
Author(s):  
Yun Dai ◽  
Guangming Yang ◽  
Lie Yang ◽  
Li Jiang ◽  
Guohua Zheng ◽  
...  

Forkhead box (FOX) transcription factors regulate the development of several human cancers. However, the role and therapeutic potential of FOXA1 in gastric cancer is still largely unexplored. The results showed a significant (P < 0.05) upregulation of FOXA1 in gastric cancer tissues and cell lines. Silencing of FOXA1 in gastric cells significantly (P < 0.05) decreased their viability through induction of apoptosis. The induction of apoptosis was associated with upregulation of Bax and downregulation of Bcl-2. Additionally, FOXA1 silencing caused activation of caspase-3 and 9 with no apparent effects on the expression of caspase-8 suggestive of intrinsic apoptosis. The transwell cell invasion revealed significant (P < 0.05) decline of cell invasion of gastric cancer cells upon FOXA1 silencing. The FOXA1 knockdown further inhibited the in vivo tumor growth suggestive of its therapeutic potential. Taken together, the findings of the present revealed that FOXA1 regulates the proliferation and development of gastric cancer and may exhibit therapeutic implications in gastric cancer treatment.


Sign in / Sign up

Export Citation Format

Share Document