A Novel Histone Deacetylase Inhibitor MCT-3 Augments Inhibition of Leukemia Cell Growth and Suppresses Induction of MMP-9 Expression by Azacitidine in HL-60 Cells.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4151-4151
Author(s):  
Hongbin Liu ◽  
Penelope A. Mayes ◽  
Patrick Perlmutter ◽  
Joseph J. Mckendrick ◽  
Anthony E. Dear

Abstract Azacitidine (AZA), a demethylating agent, has recently been demonstrated to have efficacy in the treatment of myelodysplasia and acute myeloid leukemia. A potential concern when considering the use of this agent is a recent report demonstrating AZA-mediated re-activation of matrix metalloproteinase-9 (MMP-9) expression facilitating the invasive metastatic phenotype (Sato NM et al J Natl Cancer Inst. 2003 Feb 19;95(4):327–30). Histone deacetylase inhibitors (HDACi) are a recently identified class of agents with considerable in vitro and in vivo activity and early phase clinical efficacy in the treatment of haematological malignancies (Bolden JE Nat Rev Drug Discov. 2006 Sep;5(9):769–84). Pre-clinical in vitro studies suggest that addition of HDACi to 5-aza-2′-deoxycytidine, a derivative of AZA, enhances the efficacy of this agent (Yang H et al Leuk Res. 2005 Jul; 29(7):739–48) whilst early phase clinical trials identify therapeutic activity using a combination of demethylating agents and HDACi (Garcia-Manero G, Blood. 2006 Nov 15;108(10):3271–9). Our current study aimed to determine the in vitro activity and molecular mechanisms of action of the novel HDACi MCT-3, a derivative of Oxamflatin, a hydroxamate analogue, (Dear AE, et al, Org Biomol Chem, 2006, 4, 3778–3784) in the HL-60 cell line alone and combination with AZA. AZA (1.0 microM) and MCT-3 (2.5 microM) alone inhibited HL-60 cell growth over 24hrs by 40%, 30% respectively. The combination of AZA with MCT-3 inhibited HL-60 cell growth up to 50%. Real-time PCR demonstrated that AZA and MCT-3 alone increased p15INK4b and Caspase 3 mRNA expression 2 fold. A Combination of AZA with MCT-3 increased p15INK4b and Caspase 3 mRNA expression up to 2.5 and increased p21WAF1/CIP1 and the orphan nuclear receptor Nur77 expression 2 fold. A combination of AZA and MCT-3 significantly attenuated AZA-induced MMP-9 mRNA expression and proteolytic activity. AZA and MCT-3 alone reduce HL-60 cell growth in vitro. Addition of MCT-3 to AZA increased inhibition of cell growth, suggesting that this HDACi may have the potential for additive activity with demethylating agents. AZA and MCT-3 have similar effects on expression of genes implicated in cell cycle arrest and apoptosis. Increased expression of p21WAF1/CIP1 and the orphan nuclear receptor Nur77 via inhibition of cell cycle progression and enhanced apoptosis may in part be responsible for the enhanced anti-leukaemia activity of the combination of AZA and MCT-3. Importantly MCT-3 is able to inhibit AZA-mediated induction of MMP-9 expression.

2005 ◽  
Vol 19 (1) ◽  
pp. 12-24 ◽  
Author(s):  
Ki Cheol Park ◽  
Kwang-Hoon Song ◽  
Hyo Kyun Chung ◽  
Ho Kim ◽  
Dong Wook Kim ◽  
...  

Abstract CR6-interacting factor 1 (CRIF1) was recently identified as a nuclear protein that interacts with the Gadd45 (growth arrest and DNA damage inducible 45) family of proteins and participates in the regulation of the G1/S phase of the cell cycle. However, the nuclear action of CRIF1 is largely unknown. In this study, we demonstrate that CRIF1 acts as a novel coregulator of transactivation of the orphan nuclear receptor Nur77. Both in vitro and in vivo studies show that CRIF1 interacts with Nur77 via the Nur77 AB domain and that it dramatically inhibits the AB domain-mediated transactivation of Nur77. Transient transfection assays demonstrate that CRIF1 inhibits steroid receptor coactivator-2-mediated Nur77 transactivation, and silencing of endogenous CRIF1 by small interfering RNA relieves this repression. CRIF1 possesses intrinsic repressor activities that are not affected by the histone deacetylase inhibitor Trichostatin A. In addition, overexpression of CRIF1 inhibits TSH/protein kinase A-induced Nur-responsive element promoter activity. CRIF1 inhibited Nur77-dependent induction of E2F1 promoter activity, mRNA expression, and Nur77-mediated G1/S progression in cell cycle. These results suggest that CRIF1 acts as a repressor of the orphan nuclear receptor Nur77 by inhibiting AB domain-mediated transcriptional activity.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4382-4382
Author(s):  
Maria J. Carnicer ◽  
Adriana Lasa ◽  
Elena Serrano ◽  
Jorge Pena ◽  
Angel F. Remacha ◽  
...  

Abstract Pharmacological treatment of cancer cells with demethylating agents and histone deacetylase inhibitors synergistically reactivates the transcription of previously silenced genes. The aim of this study was to investigate the antileukemic properties of a DNA mehtyltransferase inhibitor 5-aza-2′-deoxycytidine (5-Aza-dC),an histone deacetylase inhibitor Trichostatin A (TSA) and trans-retinoc acid (ATRA), alone or in combination. The effects of these drugs on apoptosis, cell cycle progression, cell-survival pathways and restoration of proliferation-associated genes silenced by aberrant epigenetic mechanisms were assessed. Four human leukemic cell lines were used: lymphoid cell-lines REH (Tel-Aml1+) and DAUDI (c-myc) and myeloid cell-lines Kasumi-1(Aml1-Eto+) and NB4(Pml-Rara+). The cells were cultured for three days. Cell viability, percentage of apoptosis, cell cycle and apoptosis-controlling proteins were examined by multiparametric flow cytometry, western blotting and a RT-PCR low-density array (LDA)containing 48 probes. Combined 5-Aza-dC and TSA treatment induced a high degree of apoptosis and affected the cell-cycle in all the cell lines analyzed. Increased levels of cleaved PARP were detected after the double treatment and paralleled those of caspase-3 except for DAUDI cell-line where there was no cleavage of procaspase 3 suggesting that this treatment induced a death-pathway independent of caspase-3 activation. Furthermore, ATRA alone has a limited capacity to induce apoptosis in all the tested cell-lines. The genes upregulated following the combined epigenetic-active treatment in DAUDI were; PRKCG, TNFSF10, TNFRSF10B, MAPK8, CASP9, APAF1, PER1, CDKN1A, CCND1. In the REH cell line were; PRKCG, TNFRSF10A, TNFRSF10B, MAPK8, CASP8, CASP9, APAF1, CASP3, AKT1, PIK3CG, BIK, PRKCG, MCL1, CCND1, CCND2, EZH2, PER1, GCLC, GSTP1. In the KASUMI-1 cell line were; PRKCABP, TNFSF10, TNFRSF10A, TNFRSF10B, FAS, CFLAR, CASP8, APAF1, CEBPA, CCND1, CDC25C, CDKN1A, EZH2.For the NB4 cell-line were: PRKCG, PRKCABP, AKT1, TNFSF10, CFLAR, CASP8 and CASP9. We can conclude that combined treatment with demethylating agents and histone deacetylase inhibitors may be active in a wide range of human leukemias. The potential use of this combination in the commonest form of pediatric ALL warrants further investigation.


2020 ◽  
Vol 16 (3) ◽  
pp. 340-349
Author(s):  
Ebrahim S. Moghadam ◽  
Farhad Saravani ◽  
Ernest Hamel ◽  
Zahra Shahsavari ◽  
Mohsen Alipour ◽  
...  

Objective: Several anti-tubulin agents were introduced for the cancer treatment so far. Despite successes in the treatment of cancer, these agents cause toxic side effects, including peripheral neuropathy. Comparing anti-tubulin agents, indibulin seemed to cause minimal peripheral neuropathy, but its poor aqueous solubility and other potential clinical problems have led to its remaining in a preclinical stage. Methods: Herein, indibulin analogues were synthesized and evaluated for their in vitro anti-cancer activity using MTT assay (on the MCF-7, T47-D, MDA-MB231 and NIH-3T3 cell lines), annexin V/PI staining assay, cell cycle analysis, anti-tubulin assay and caspase 3/7 activation assay. Results: One of the compounds, 4a, showed good anti-proliferative activity against MCF-7 cells (IC50: 7.5 μM) and low toxicity on a normal cell line (IC50 > 100 μM). All of the tested compounds showed lower cytotoxicity on normal cell line in comparison to reference compound, indibulin. In the annexin V/PI staining assay, induction of apoptosis in the MCF-7 cell line was observed. Cell cycle analysis illustrated an increasing proportion of cells in the sub-G-1 phase, consistent with an increasing proportion of apoptotic cells. No increase in G2/M cells was observed, consistent with the absence of anti-tubulin activity. A caspase 3/7 assay protocol showed that apoptosis induction by more potent compounds was due to activation of caspase 3. Conclusion: Newly synthesized compounds exerted acceptable anticancer activity and further investigation of current scaffold would be beneficial.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Shuiyan Wu ◽  
You Jiang ◽  
Yi Hong ◽  
Xinran Chu ◽  
Zimu Zhang ◽  
...  

Abstract Background T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease with a high risk of induction failure and poor outcomes, with relapse due to drug resistance. Recent studies show that bromodomains and extra-terminal (BET) protein inhibitors are promising anti-cancer agents. ARV-825, comprising a BET inhibitor conjugated with cereblon ligand, was recently developed to attenuate the growth of multiple tumors in vitro and in vivo. However, the functional and molecular mechanisms of ARV-825 in T-ALL remain unclear. This study aimed to investigate the therapeutic efficacy and potential mechanism of ARV-825 in T-ALL. Methods Expression of the BRD4 were determined in pediatric T-ALL samples and differential gene expression after ARV-825 treatment was explored by RNA-seq and quantitative reverse transcription-polymerase chain reaction. T-ALL cell viability was measured by CCK8 assay after ARV-825 administration. Cell cycle was analyzed by propidium iodide (PI) staining and apoptosis was assessed by Annexin V/PI staining. BRD4, BRD3 and BRD2 proteins were detected by western blot in cells treated with ARV-825. The effect of ARV-825 on T-ALL cells was analyzed in vivo. The functional and molecular pathways involved in ARV-825 treatment of T-ALL were verified by western blot and chromatin immunoprecipitation (ChIP). Results BRD4 expression was higher in pediatric T-ALL samples compared with T-cells from healthy donors. High BRD4 expression indicated a poor outcome. ARV-825 suppressed cell proliferation in vitro by arresting the cell cycle and inducing apoptosis, with elevated poly-ADP ribose polymerase and cleaved caspase 3. BRD4, BRD3, and BRD2 were degraded in line with reduced cereblon expression in T-ALL cells. ARV-825 had a lower IC50 in T-ALL cells compared with JQ1, dBET1 and OTX015. ARV-825 perturbed the H3K27Ac-Myc pathway and reduced c-Myc protein levels in T-ALL cells according to RNA-seq and ChIP. In the T-ALL xenograft model, ARV-825 significantly reduced tumor growth and led to the dysregulation of Ki67 and cleaved caspase 3. Moreover, ARV-825 inhibited cell proliferation by depleting BET and c-Myc proteins in vitro and in vivo. Conclusions BRD4 indicates a poor prognosis in T-ALL. The BRD4 degrader ARV-825 can effectively suppress the proliferation and promote apoptosis of T-ALL cells via BET protein depletion and c-Myc inhibition, thus providing a new strategy for the treatment of T-ALL.


2021 ◽  
Vol 18 (1) ◽  
Author(s):  
Xiaohong Zhou ◽  
Christina Monnie ◽  
Maria DeLucia ◽  
Jinwoo Ahn

Abstract Background Vpr is a virion-associated protein that is encoded by lentiviruses and serves to counteract intrinsic immunity factors that restrict infection. HIV-1 Vpr mediates proteasome-dependent degradation of several DNA repair/modification proteins. Mechanistically, Vpr directly recruits cellular targets onto DCAF1, a substrate receptor of Cullin 4 RING E3 ubiquitin ligase (CRL4) for poly-ubiquitination. Further, Vpr can mediate poly-ubiquitination of DCAF1-interacting proteins by the CRL4. Because Vpr-mediated degradation of its known targets can not explain the primary cell-cycle arrest phenotype that Vpr expression induces, we surveyed the literature for DNA-repair-associated proteins that interact with the CRL4-DCAF1. One such protein is SIRT7, a deacetylase of histone 3 that belongs to the Sirtuin family and regulates a wide range of cellular processes. We wondered whether Vpr can mediate degradation of SIRT7 via the CRL4-DCAF1. Methods HEK293T cells were transfected with cocktails of plasmids expressing DCAF1, DDB1, SIRT7 and Vpr. Ectopic and endogeneous levels of SIRT7 were monitered by immunoblotting and protein–protein interactions were assessed by immunoprecipitation. For in vitro reconstitution assays, recombinant CRL4-DCAF1-Vpr complexes and SIRT7 were prepared and poly-ubiqutination of SIRT7 was monitored with immunoblotting. Results We demonstrate SIRT7 polyubiquitination and degradation upon Vpr expression. Specifically, SIRT7 is shown to interact with the CRL4-DCAF1 complex, and expression of Vpr in HEK293T cells results in SIRT7 degradation, which is partially rescued by CRL inhibitor MNL4924 and proteasome inhibitor MG132. Further, in vitro reconstitution assays show that Vpr induces poly-ubiquitination of SIRT7 by the CRL4-DCAF1. Importantly, we find that Vpr from several different HIV-1 strains, but not HIV-2 strains, mediates SIRT7 poly-ubiquitination in the reconstitution assay and degradation in cells. Finally, we show that SIRT7 degradation by Vpr is independent of the known, distinctive phenotype of Vpr-induced cell cycle arrest at the G2 phase, Conclusions Targeting histone deacetylase SIRT7 for degradation is a conserved feature of HIV-1 Vpr. Altogether, our findings reveal that HIV-1 Vpr mediates down-regulation of SIRT7 by a mechanism that does not involve novel target recruitment to the CRL4-DCAF1 but instead involves regulation of the E3 ligase activity.


2006 ◽  
Vol 82 (6) ◽  
pp. 877-887 ◽  
Author(s):  
J. Sehm ◽  
H. Lindermayer ◽  
H. H. D. Meyer ◽  
M. W. Pfaffl

Flavan-3-ols are a class of flavonoids that are widely distributed in fruits and beverages including red wine and apples. Consumption of flavanoid-rich food has been shown to exhibit anti-microbial, anti-oxidative, anti-inflammatory, and immune-modulating effects. To test the nutritional effects of flavanols on mRNA gene-expression of inflammatory and apoptotic marker genes, piglets were given two flavanoids-rich feeding regimens: a low flavanoid standard diet (SD) was compared with diets enriched with 3·5% apple pomace (APD) or 3·5% red-wine pomace (RWPD). The influence on mRNA expression levels was investigated in different immunological active tissues and in the gastro-intestinal tract (GIT). The investigation took place from 1 week prior weaning to 19 days post weaning in 78 piglets. The expression of expressed marker genes was determinate by one-step quantitative real-time (qRT-PCR): TNFα, NFκB as pro-inflammatory; IL10, as anti-inflammatory; caspase 3 as apoptosis; cyclin D1 as cell cycle marker; and nucleosome component histon H3 as reference gene.The feeding regimens result in tissue individual regulation of mRNA gene expression in all investigated organs. It was discovered that there were significant differences between the applied diets and significant changes during feeding time curse. Both pomace treatments caused a significant up-regulation of all investigated genes in liver. The effect on mesenterial lymph nodes and spleen was not prominent. In the GIT, the treatment groups showed a inhibitory effects on gene expression mainly in stomach and jejunum (NFκB, cyclin D1 and caspase 3). In colon the trend of caspase 3 was positive with the greatest change in the RWPD group.In jejunum and stomach the cell cycle turn over was reduced, whereas in liver the cell turn over was highly accelerate. The influence on inflammatory marker gene expression is mainly relevant in stomach. It is presume that both flavanoid rich feeding regimens have the potential to modulate the mRNA expressions of inflammatory, proliferation and apoptotic marker genes in the GIT and piglet organs.


2020 ◽  
Vol 29 ◽  
pp. 101583
Author(s):  
José Athayde Vasconcelos Morais ◽  
Mosar C. Rodrigues ◽  
Fernanda F. Ferreira ◽  
Kunal Ranjan ◽  
Ricardo Bentes Azevedo ◽  
...  

2020 ◽  
Vol 52 (2) ◽  
pp. 168-179 ◽  
Author(s):  
Huilin Gong ◽  
Shan Gao ◽  
Chenghuan Yu ◽  
Meihe Li ◽  
Ping Liu ◽  
...  

Abstract Y-box binding protein 1 (YB-1) is manifested as its involvement in cell proliferation and differentiation and malignant cell transformation. Overexpression of YB-1 is associated with glioma progression and patient survival. The aim of this study is to investigate the influence of YB-1 knockdown on glioma cell progression and reveal the mechanisms of YB-1 knockdown on glioma cell growth, migration, and apoptosis. It was found that the knockdown of YB-1 decreased the mRNA and protein levels of YB-1 in U251 glioma cells. The knockdown of YB-1 significantly inhibited cell proliferation, colony formation, and migration in vitro and tumor growth in vivo. Proteome and phosphoproteome data revealed that YB-1 is involved in glioma progression through regulating the expression and phosphorylation of major proteins involved in cell cycle, adhesion, and apoptosis. The main regulated proteins included CCNB1, CCNDBP1, CDK2, CDK3, ADGRG1, CDH-2, MMP14, AIFM1, HO-1, and BAX. Furthermore, it was also found that YB-1 knockdown is associated with the hypo-phosphorylation of ErbB, mTOR, HIF-1, cGMP-PKG, and insulin signaling pathways, and proteoglycans in cancer. Our findings indicated that YB-1 plays a key role in glioma progression in multiple ways, including regulating the expression and phosphorylation of major proteins associated with cell cycle, adhesion, and apoptosis.


2003 ◽  
Vol 23 (24) ◽  
pp. 9375-9388 ◽  
Author(s):  
Melanie J. McConnell ◽  
Nathalie Chevallier ◽  
Windy Berkofsky-Fessler ◽  
Jena M. Giltnane ◽  
Rupal B. Malani ◽  
...  

ABSTRACT The transcriptional repressor PLZF was identified by its translocation with retinoic acid receptor alpha in t(11;17) acute promyelocytic leukemia (APL). Ectopic expression of PLZF leads to cell cycle arrest and growth suppression, while disruption of normal PLZF function is implicated in the development of APL. To clarify the function of PLZF in cell growth and survival, we used an inducible PLZF cell line in a microarray analysis to identify the target genes repressed by PLZF. One prominent gene identified was c-myc. The array analysis demonstrated that repression of c-myc by PLZF led to a reduction in c-myc-activated transcripts and an increase in c-myc-repressed transcripts. Regulation of c-myc by PLZF was shown to be both direct and reversible. An interaction between PLZF and the c-myc promoter could be detected both in vitro and in vivo. PLZF repressed the wild-type c-myc promoter in a reporter assay, dependent on the integrity of the binding site identified in vitro. PLZF binding in vivo was coincident with a decrease in RNA polymerase occupation of the c-myc promoter, indicating that repression occurred via a reduction in the initiation of transcription. Finally, expression of c-myc reversed the cell cycle arrest induced by PLZF. These data suggest that PLZF expression maintains a cell in a quiescent state by repressing c-myc expression and preventing cell cycle progression. Loss of this repression through the translocation that occurs in t(11;17) would have serious consequences for cell growth control.


2005 ◽  
Vol 289 (4) ◽  
pp. C826-C835 ◽  
Author(s):  
Sharon Barone ◽  
Tomohisa Okaya ◽  
Steve Rudich ◽  
Snezana Petrovic ◽  
Kathy Tenrani ◽  
...  

Ischemia-reperfusion injury (IRI) in liver and other organs is manifested as an injury phase followed by recovery and resolution. Control of cell growth and proliferation is essential for recovery from the injury. We examined the expression of three related regulators of cell cycle progression in liver IRI: spermidine/spermine N-acetyltransferase (SSAT), p21 (a cyclin-dependent kinase inhibitor), and stathmin. Mice were subjected to hepatic IRI, and liver tissues were harvested at timed intervals. The expression of SSAT, the rate-limiting enzyme in the polyamine catabolic pathway, had increased fivefold 6 h after IRI and correlated with increased putrescine levels in the liver, consistent with increased SSAT enzymatic activity in IRI. The expression of p21, which is transactivated by p53, was undetectable in sham-operated animals but was heavily induced at 12 and 24 h of reperfusion and declined to undetectable baseline levels at 72 h of reperfusion. The interaction of the polyamine pathway with the p53-p21 pathway was shown in vitro, where activation of SSAT with polyamine analog or the addition of putrescine to cultured hepatocytes induced the expression of p53 and p21 and decreased cell viability. The expression of stathmin, which is under negative transcriptional regulation by p21 and controls cell proliferation and progression through mitosis, remained undetectable at 6, 12, and 24 h of reperfusion and was progressively and heavily induced at 48 and 72 h of reperfusion. Double-immunofluorescence labeling with antibodies against stathmin and PCNA, a marker of cell proliferation, demonstrated colocalization of stathmin and PCNA at 48 and 72 h of reperfusion in hepatocytes, indicating the initiation of cell proliferation. The distinct and sequential upregulation of SSAT, p21, and stathmin, along with biochemical activation of the polyamine catabolic pathway in IRI in vivo and the demonstration of p53-p21 upregulation by SSAT and putrescine in vitro, points to the important role of regulators of cell growth and cell cycle progression in the pathophysiology and/or recovery in liver IRI. The data further suggest that SSAT may play a role in the initiation of injury, whereas p21 and stathmin may be involved in the resolution and recovery after liver IRI.


Sign in / Sign up

Export Citation Format

Share Document