The human aortic endothelium undergoes dose-dependent DNA methylation in response to transient hyperglycemia

2021 ◽  
Vol 400 (2) ◽  
pp. 112485
Author(s):  
Mark E. Pepin ◽  
Concetta Schiano ◽  
Marco Miceli ◽  
Giuditta Benincasa ◽  
Gelsomina Mansueto ◽  
...  
2021 ◽  
Vol 36 (Supplement_1) ◽  
Author(s):  
M C Carbajo-García ◽  
A Corachán ◽  
M Segura ◽  
J Monleón ◽  
J Escrig ◽  
...  

Abstract Study question Is DNA methylation reversion through DNA methyltransferases (DNMT) inhibitors, such as 5-aza–2’-deoxycitidine, a potential therapeutic option for treatment of patients with uterine leiomyomas (UL)? Summary answer 5-aza–2’-deoxycitidine reduces proliferation and extracellular matrix (ECM) formation by inhibition of Wnt/ β-catenin pathway on UL cells, suggesting DNMT inhibitors as an option to treat UL. What is known already: UL is a multifactorial disease with an unclear pathogenesis and inaccurate treatment. Aberrant DNA methylation have been found in UL compared to myometrium (MM) tissue, showing hypermethylation of tumor suppressor genes, which contributes to the development of this tumor. The use of DNMT inhibitors, such as 5-aza–2’-deoxycytidine (5-aza-CdR), has been suggested to treat tumors in which altered methylation pattern is related to tumor progression, as occurs in UL. Based on this, we aimed to evaluate whether DNA methylation reversion through 5-aza-CdR reduces cell proliferation and ECM formation in UL cells, being a potential option for UL medical treatment. Study design, size, duration Prospective study comparing UL versus MM tissue and human uterine leiomyoma primary (HULP) cells treated with/without 5-aza-CdR at 0 µM (control), 2 µM, 5 µM and 10 µM for 72 hours. UL and MM tissue were collected from women without any hormonal treatment for the last 3 months (n = 16) undergoing myomectomy or hysterectomy due to symptomatic leiomyoma pathology. Participants were recruited between January 2019 and February 2020 at Hospital Universitario y Politecnico La Fe (Spain). Participants/materials, setting, methods Samples were collected from Caucasian premenopausal women aged 31–48 years, with a body mass index of < 30 and without hormonal treatment. DNMT1 gene expression was analysed in UL vs MM tissue by qRT-PCR and activity of DNMT was measured in UL and MM tissue and cells by ELISA. 5-aza-CdR effect on proliferation was assessed by CellTiter test and Western blot (WB), apoptosis and ECM analyzed by WB and Wnt/ β-catenin pathway by qRT-PCR and WB. Main results and the role of chance: DNMT1 gene expression was increased in UL compared to MM tissue (fold change [FC]=2.49, p-value [p]=0.0295). Similarly, DNMT activity was increased in both UL compared to MM tissue and HULP cells versus MM cells (6.50 vs 3.76 OD/h/mg, p = 0.026; 211.30 vs 63.67 OD/h/mg, p = 0.284, respectively). After 5-aza-CdR treatment, cell viability of HULP cells was reduced in a dose dependent manner, being statistically significant at 10 µM (85.25%, p = 0.0001). Accordantly, PCNA protein expression was significantly decreased at 10 µM in HULP cells (FC = 0.695, p = 0.034), demonstrating cell proliferation inhibition. Additionally, 5-aza-CdR inhibited ECM protein expression in HULP cells in a dose-dependent manner being statistically significant at 10 µM for COLLAGEN I (FC = 0.654, p = 0.023) and PAI–1 (FC = 0.654, p = 0.023), and at 2 µM and 10 µM for FIBRONECTIN (FC = 0.812, p = 0.020; FC = 0.733, p = 0.035; respectively). Final targets of Wnt/ β-catenin pathway were decreased after 5-aza-CdR treatment, protein expression of WISP1 was significantly inhibited at 10 µM (FC = 0.699, p = 0.026), while expression levels of Wnt/ β-catenin target genes C-MYC (FC = 0.745, p = 0.028 at 2 µM; FC = 0.728, p = 0.019 at 10 µM) and MMP7 (FC = 0.520, p = 0.003 at 5 µM, FC = 0.577, p = 0.007 at 10 µM) were also significantly downregulated in HULP-treated cells vs untreated cells. Limitations, reasons for caution: This study has strict inclusion criteria to diminish epigenetic variability, thereby we should be cautious extrapolating our results to general population. Besides, this is a proof of concept with the inherent cell culture limitations. Further studies are necessary to determine 5-aza-CdR dose and adverse effects on UL in vivo. Wider implications of the findings: 5-aza-CdR treatment reduces cell proliferation and ECM formation through Wnt/ β-catenin pathway inhibition, suggesting that inhibition of DNA methylation could be a promising new therapeutic approach to treat UL. Trial registration number Not applicable


2015 ◽  
Vol 97 (2) ◽  
pp. 216-227 ◽  
Author(s):  
Emma Strong ◽  
Darci T. Butcher ◽  
Rajat Singhania ◽  
Carolyn B. Mervis ◽  
Colleen A. Morris ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3763-3763
Author(s):  
Nadja Blagitko-Dorfs ◽  
Tobias Bauer ◽  
Maren Prinz ◽  
Wolfram Brugger ◽  
Gesine Bug ◽  
...  

Abstract Introduction Epigenetic therapies with azanucleoside DNA hypomethylating agents, alone or in combination with histone deacetylase inhibitors (HDACi), show clinical activity in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML), particularly when given at non-cytotoxic doses. They are able to reactivate epigenetically silenced genes including, among others, a number of highly immunogenic proteins dubbed Cancer/testis antigens (CTAs), predominantly the CTAs located on the X chromosome. We have previously shown that decitabine can induce expression of several CTAs, including MAGEB2 and NY-ESO-1, in myeloid cells in vitro and thereby trigger an immune response (Almstedt et al., Leuk. Res. 2010). Induction of a CTA-specific cytotoxic T cell response in vivo was reported also in AML patients treated with azacitidine and sodium valproate (VPA) and correlated with clinical response (Goodyear et al., Blood 2010). To the best of our knowledge, no data have yet been reported on the effect of combination treatment with decitabine and panobinostat or sodium valproate (VPA) on CTA reactivation in myeloid leukemia. Aim We hypothesized that by combining decitabine with HDACi we could further enhance expression of CTAs in myeloid leukemia cells and thereby boost recognition of the malignant cells by the cytotoxic T lymphocytes. Methods The myeloid cell lines U937 and Kasumi-1 were treated with decitabine alone or in combination with the HDACi VPA or panobinostat applied at non-toxic concentrations (>80% cell viability). Expression of CTAs was analyzed by RT-qPCR and Western blot after 48 hours of HDACi treatment. DNA methylation of NY-ESO-1 and MAGEB2 promoter regions was quantified by pyrosequencing. Bone marrow mononuclear cells from 19 AML patients (treated with or without VPA as add-on to decitabine in the ongoing randomized phase II DECIDER clinical trial, NCT00867672) were collected before and on day 15 of treatment, in some patients also after 2 treatment cycles. CTA mRNA expression and promoter DNA methylation were quantified as described above. Results VPA or panobinostat alone did not induce MAGEB2 or NY-ESO-1 expression in vitro. However the pretreatment of cells with decitabine prior to addition of either HDACi resulted in a synergistic dose-dependent reactivation of MAGEB2 and NY-ESO-1 on the mRNA level (confirmed for the latter on the protein level). Pyrosequencing analysis of the heavily methylated NY-ESO-1 and MAGEB2 promoters revealed, as expected, no methylation changes upon HDACi treatment, but a dose-dependent hypomethylation upon decitabine. In recently initiated in vivo studies (DECIDER trial), until now cells from 19 AML patients receiving epigenetic treatment were sequentially analyzed. Induction of MAGEB2 mRNA was observed in 9 patients (from absent to a median of 0.002 relative to GAPDH, range 0.0004-0.043), with concomitant DNA hypomethylation of the MAGEB2 promoter from median 83% pretreatment methylation (range 63%-90%) to 63% posttreatment (range 44%-74%). In 5 patients modest hypomethylation without changes in MAGEB2 expression was observed (from median pretreatment values of 89% [72%-92%] to 82% [58%-87%] posttreatment). Another 5 patients disclosed neither hypomethylation nor reexpression of MAGEB2 (results as yet blinded to treatment arm and clinical response). Conclusions Combined epigenetic treatment with the hypomethylating agent decitabine and the HDACi VPA or panobinostat synergistically induced a dose-dependent reactivation of the CTAs MAGEB2 and NY-ESO-1 in vitro, accompanied by promoter hypomethylation. First translational results of the DECIDER AML trial also indicate in vivo effects of the epigenetic treatment on CTA induction. The unmasking of CTAs to the immune system by epigenetically active drugs can increase anti-tumor immune responses, and thus has clear implications for future clinical trials combining epigenetic therapy and specific immunotherapy in myeloid neoplasia. Disclosures: No relevant conflicts of interest to declare.


2006 ◽  
Vol 188 (2) ◽  
pp. 305-310 ◽  
Author(s):  
Pauliina Utriainen ◽  
Jianqi Liu ◽  
Tiina Kuulasmaa ◽  
Raimo Voutilainen

Activin affects adrenocortical steroidogenesis and increases apoptosis, while follistatin (FS) acts as an activin antagonist by binding to activin, preventing attachment to its receptors. The regulation of FS expression in the adrenal cortex is poorly understood. Adrenocortical tumors often display aberrant methylation. In the present study, we investigated the effect of DNA methylation on FS mRNA expression and peptide secretion in adrenocortical cells. We treated human NCI-H295R adrenocortical cells with the methylation inhibitor 5-Aza-2′deoxycytidine (Azad; 0.1–100 μM for 1, 4 or 7 days) and measured FS mRNA expression by Northern blot and quantitative real time RT-PCR analyses as well as FS secretion by specific ELISA. Methylation-specific PCR showed decreased methylation in the FS promoter region after Azad treatment. A significant (P < 0.05) time- and dose-dependent increase in FS mRNA expression (up to 4.6-fold) and peptide secretion (up to 17.1-fold) was detected after Azad treatment. We conclude that FS gene expression and peptide secretion in NCI-H295R adrenocortical cells are regulated by DNA methylation. Thus, variable methylation in different adrenocortical tumors may influence activin bioactivity and its consequences in steroidogenesis and cell proliferation/apoptosis.


Author(s):  
Gerhard P. Aigner ◽  
Verena Pittl ◽  
Birgit Fiechtner ◽  
Bernhard Egger ◽  
Maja Šrut ◽  
...  

2014 ◽  
Vol 46 (7) ◽  
pp. 245-255 ◽  
Author(s):  
Pankaj Chaturvedi ◽  
Anuradha Kalani ◽  
Srikanth Givvimani ◽  
Pradip Kumar Kamat ◽  
Anastasia Familtseva ◽  
...  

The mechanisms of homocysteine-mediated cardiac threats are poorly understood. Homocysteine, being the precursor to S-adenosyl methionine (a methyl donor) through methionine, is indirectly involved in methylation phenomena for DNA, RNA, and protein. We reported previously that cardiac-specific deletion of N-methyl-d-aspartate receptor-1 (NMDAR1) ameliorates homocysteine-posed cardiac threats, and in this study, we aim to explore the role of NMDAR1 in epigenetic mechanisms of heart failure, using cardiomyocytes during hyperhomocysteinemia (HHcy). High homocysteine levels activate NMDAR1, which consequently leads to abnormal DNA methylation vs. histone acetylation through modulation of DNA methyltransferase 1 (DNMT1), HDAC1, miRNAs, and MMP9 in cardiomyocytes. HL-1 cardiomyocytes cultured in Claycomb media were treated with 100 μM homocysteine in a dose-dependent manner. NMDAR1 antagonist (MK801) was added in the absence and presence of homocysteine at 10 μM in a dose-dependent manner. The expression of DNMT1, histone deacetylase 1 (HDAC1), NMDAR1, microRNA (miR)-133a, and miR-499 was assessed by real-time PCR as well as Western blotting. Methylation and acetylation levels were determined by checking 5′-methylcytosine DNA methylation and chromatin immunoprecipitation. Hyperhomocysteinemic mouse models (CBS+/−) were used to confirm the results in vivo. In HHcy, the expression of NMDAR1, DNMT1, and matrix metalloproteinase 9 increased with increase in H3K9 acetylation, while HDAC1, miR-133a, and miR-499 decreased in cardiomyocytes. Similar results were obtained in heart tissue of CBS+/− mouse. High homocysteine levels instigate cardiovascular remodeling through NMDAR1, miR-133a, miR-499, and DNMT1. A decrease in HDAC1 and an increase in H3K9 acetylation and DNA methylation are suggestive of chromatin remodeling in HHcy.


2014 ◽  
Vol 133 (1) ◽  
pp. 18-25 ◽  
Author(s):  
Hongli Li ◽  
Yan Wang ◽  
Wenwei Xu ◽  
Lin Dong ◽  
Yan Guo ◽  
...  

Background: Gene silencing associated with aberrant methylation of promoter region CpG islands is an acquired epigenetic alteration that serves as an alternative to genetic defects in the inactivation of tumor suppressor genes in human cancers. The demethylating, dose-dependent effect of arsenic trioxide (As2O3) on several tumor-related genes has already been postulated. However, whether such a demethylating effect also applies to the TMS1 gene in chronic myeloid leukemia cell line K562 cells has not been studied so far. The aim of the present study was to detect the methylation status of the TMS1 gene in K562 cells and the demethylation effect of As2O3 on TMS1 as well as TMS1 apoptosis-associated protein Bcl-2/Bax and DNA methyltransferase (DNMT) expression. Methods: TMS1 mRNA expression in K562 cells and normal bone marrow was determined by reverse transcription (RT) polymerase chain reaction (PCR), and the DNA methylation status of the TMS1 promoter in K562 cells treated with different concentrations of As2O3 for 48 h was determined by methylation-specific PCR. RT-PCR and Western blot were used to detect TMS1 and DNMT expression. We also assessed TMS1-associated apoptosis protein Bcl-2/Bax expression by Western blot and apoptosis rates by flow cytometry using annexin V/propidium iodide double staining. Results: In K562 cells, TMS1 was completely methylated and both TMS1 mRNA and protein showed a low expression, but 2 μmol/l As2O3 could significantly restore the expression of the TMS1 gene both at mRNA and protein level (p < 0.01) by fully reversing DNA methylation. As2O3 decreased mRNA and protein expression of DNMT1 (p < 0.05) in a dose-dependent manner. Flow cytometry showed that in the experimental group (2 μmol/l As2O3), cell apoptosis was significantly increased compared with the control group (no As2O3; p < 0.05). In the experimental group, Western blot showed that the expression of the anti-apoptotic protein Bcl-2 was significantly decreased; however, the proapoptotic protein Bax was markedly increased and the Bcl-2/Bax ratio was markedly reduced (p < 0.01). Conclusions: As2O3 could restore the expression of TMS1 by inhibiting DNMT to reverse the hypermethylation and induced apoptosis of K562 cells by downregulation of Bcl-2/Bax expression.


2017 ◽  
Vol 7 (1) ◽  
pp. e994-e994 ◽  
Author(s):  
R Cervera-Juanes ◽  
L J Wilhelm ◽  
B Park ◽  
K A Grant ◽  
B Ferguson

1987 ◽  
Author(s):  
H Bult ◽  
F Jordaens ◽  
A G Herman ◽  
C Van Hove ◽  
T J Verbeuren

Endothelium may release EDRF and PGI2; the former causes relaxation of arterial smooth muscle cells, whereas PGI2 suppresses phagocyte and platelet activation and may relax some arteries. The aim of the present experiments was to investigate whether EDRF from rabbit aorta affects platelet reactivity. To this end the isolated thoracic aorta was perfused with a Krebs' solution (3 ml/min), andthe perfusate was continuously super fused over a de-endothe-lialized ring of the abdominal aorta, contracted withnoradrenaline in the presence of atropine, to monitor EDRF. Aliquots (200 μl) ofeffluent were also added to 200 (il prewarmedautologous citrate platelet rich plasma, which was immediately stimulated with ADP or U46619, a thromboxane (TX) A2 mimetic. Both agonists induce aggregation independently of TXA2 formation. Finally, the PGI2 content of the effluent was assessed by specific radioimmunoassay of 6-oxo-PGF1alpha(ir-6oxo). Infusion of 0.3,1 and 3 μM acetylcholine (Ach) through the aorta for 7.5 min caused a dose-dependent relaxation of the abdominal ring (33 to 100 % at 3 (μM Ach), indicating intraluminal EDRF release. Maximum relaxation was reached in 3 min and maintained. Release of platelet inhibitory activity was variable. At 3 μM Ach, anti-platelet activity(more than 10 % suppression of aggregation)was present in the effluent of 5 (of 7) aorta's for ADP-, and in effluent of 6 (of 7) aorta's for U46617-induced aggregation. U46619 was more sensitive to the anti-platelet activity, a finding consistent withauthentic PGI2. When a clear anti-platelet activity was monitored, its release was abolished by indo-methacin, which affected neither EDRF release nor platelet aggregation. Baseline release of ir-6oxo was low and variable, but dose-dependently stimulated by Ach, reaching a maximum for each concentration in the 2nd to 3rd minute. In contrast to EDRF, ir-6oxo release was not maintained during the subsequent 4 min and tapered off. Formation of ir-6oxo and anti-aggregatingeffect were positively correlated, but therewas no association between EDRF and ir-6oxo or anti-platelet activity. In conclusion, rabbit aortic endothelium could release EDRF and PGI2, but the present set-up did not reveal an anti-platelet effect of EDRF.


Sign in / Sign up

Export Citation Format

Share Document