Mechanism of Synergy Between Bortezomib and Arsenic Trioxide in Acute Promyelocytic Leukemia and Clinical Efficacy in Relapsed Patients

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3607-3607
Author(s):  
Saravanan Ganesan ◽  
Ezhilarasi Chendamarai ◽  
Ansu Abu Alex ◽  
Sachin David ◽  
Giridhara R Jayandharan ◽  
...  

Abstract Abstract 3607 Bortezomib (Bo) has been shown to have direct cytotoxicity against malignant promyelocytes. There remain concerns about combining it with ATO. A major concern is that proteasomal inhibition by Bo could decrease ATO induced PML-RARA degradation and reduce its efficacy. In an in-vitro experiment we noted a significant synergistic cytotoxic effect when Bo (80ng/ml; a pharmacologically relevant concentration) was combined with ATO (1– 6uM) on NB4 cells (n = 12), P=0.03 (Fig 1a). We also noted that co-culture of NB4 or primary APL cells with stromal cells (Hs-5 cell line) protects them from ATO induced apoptosis and Bo can overcome this protective effect (Fig 1b). We undertook a series of experiments to address the mechanism of these observations. We observed an increased activation of NF-κB pathway in malignant promyelocytes (NB4 cells) co-cultured with stromal cells. There was a synergistic inhibition of NF-κB when a combination of ATO and Bo was used (Fig 1c). Considering potent inhibition of NF-κB pathway and the known effect of this on reactive oxygen species (ROS), we checked the level of ROS in the malignant promyelocytes and observed that there was an increase in ROS levels when compared to ATO or Bo alone treated cells (n=5; treatment for 6 hours) (Fig 1d). When the ROS was abrogated by pre-treating the NB4 cells with N-Acetyl Cysteine (NAC; 5mM for 2 hrs) there was a protective effect against the ATO combined with Bo induced apoptosis at 24 hours (mean increase of 73% from 65.5%; p=0.03; n=4). Degradation of PML-RARA was seen in ATO or Bo alone treated cells at 24 hours and a similar degradation was also seen when ATO and Bo were combined (Fig 1f; n=3). Bo at the concentration used (80ng/ml) was able to block 26s proteasome complex, as a result accumulation of ubiquitinated products was seen at 24 hours followed by degradation at the end of 48 hours (Fig 1e). We then looked at autophagy as a possible alternate mechanism to explain PML-RARA degradation when a combination of ATO and Bo was used in the setting of effective proteasomal inhibition. At 24 hours, there was a evidence of induction in autophagy as shown by LC3II formation using western blot technique and this was maximum at the end of 48 hours, this time point coincides with time at which maximum PML-RARA degradation happens in this experimental setting (Fig 1f). We have also observed that there is an accumulation of p62 (ubiquitinated protein binding protein) at 24 hours and degraded by 48 hours suggests that accumulated ubiquitinated products were cleared by autophagy via p62 (Fig 1f). The induction of autophagy was further validated by real time-PCR where autophagy genes atg5 and beclin1 levels were significantly increased by 3.1 fold (p=0.01) and 2.1 fold (p=0.03) respectively. Blocking autophagy by 3-methyl adenine or chloroquine did not enhance survival but paradoxically appeared to further enhance cell death (n= 3; p=0.02) though there was partial inhibition in the degradation of PML-RARA (data not shown). We have also observed that there was a synergistic and more rapid increase in apoptosis when ATO and Bo were combined as evidenced by an enhanced degradation of caspase3 (Fig 1f). Preliminary phase I data in 4 patients is summarized in table 1. Patients received Bo in induction and in consolidation once a week for 4 weeks along with ATO. Two cases, due to financial constraints, could not have a stem cell transplant (SCT) in molecular remission. In summary the combination of these drugs was well tolerated and durable CR3 and CR2 was obtained even in cases that did not have a SCT in remission. None of these cases have relapsed at a median follow up of 14.5 months. In conclusion blocking 26s proteasomal complex by Bo does not alter the efficacy of ATO, instead Bo synergizes with ATO. The mechanism of this synergy is multi-factorial and appears to be predominantly due to increase in ROS activity and increased apoptosis. Additionally stromal cell mediated protection against ATO induced apoptosis is overcome by addition of Bo. Observed degradation of PML-RARA by ATO+Bo could be explained by induction of autophagy in these cells. Table 1: Summary of cases and duration of CR prior and post remission induction with arsenic trioxide and bortezomib Case Age Sex Relapse number Duration of last CR (months) Prior autologous SCT Post remission SCT Duration of current CR (months) RS 25 M R2 19 Yes No 19 BJ 31 M R1 15 No Yes (auto) 15 TK 35 M R2 24 Yes Yes (MUD) 14 SS 34 F R1 19 No No 13 Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 3-3
Author(s):  
Gustavo Milone ◽  
Samuel Sarmiento Doncel ◽  
Carol Agudelo Rico ◽  
Fabiola Vizcarra Reyes ◽  
Gina Alejandra Diaz Mosquera ◽  
...  

Acute promyelocytic leukemia (APL) is a subtype of Acute Myeloid Leukemia (AML) in which a chromosomal translocation t (15; 17) (q22; q12) is generated by fusing produces a hybrid PML / RARα gene, generating an altered signal . The combination of transretinoic acid (ATRA) plus arsenic trioxide (ATO) has been shown to be superior to ATRA plus chemotherapy in the treatment of newly diagnosed standard risk patients with acute promyelocytic leukemia (APL) in several countries. The objective of the present study is to describe the frequency of remission in patients with acute promyelocytic leukemia who were administered as a first line Arsenic Trioxide (varitrinox) during the period from November 2017 to June 2020 in Colombian patients. Methods: Retrospective observational and descriptive study of 12 patients diagnosed with acute promyelocytic leukemia treated with ATO Arsenic trioxide (Varitrinox) as first line, the source of information was provided by the treating hematologists (medical records) by filling out the technical concept format. Active pharmacovigilance scientist in Colombia, this format keeps the identification information of the patient anonymized and the confidentiality of the data is guaranteed as well as compliance with the rules of good clinical practice. Results: Twelve patients with age range between 22 and 69 years with a median age of 34.0 were analyzed. It was found in the analysis that 100% had induction hematologic remission with a median of 45 days. 75% of patients received ATO + ATRA and were at low and intermediate risk, the remaining 25% received ATRA + ATO + Chemotherapy and were at high risk, and intermediate risk. 91.7% of molecular remission in consolidation was obtained and it was measured in cycle 3 by means of PCR (undetectable), 8.3% (n = 1) was positive 3% and is finishing consolidation. Regarding the most frequent adverse events, intravascular coagulation (n = 9), neutropenia (n = 6) and thrombocytopenia (n = 6) were observed. 75% of patients are disease-free, 16.7% are on maintenance (they received ATO + ATRA + Induction chemotherapy) and 8.3% are on consolidation. So far, none of the patients under study have died. Conclusions: Our results support the use of ATO (Varitrinox) in newly diagnosed APL patients (as first line), as a care strategy for low, intermediate and high risk patients. The role of ATRA-ATO is guaranteed in other studies where they manage patients of different risks. Key words: Arsenic trioxide, leukemia promyelocytic acute, leukemia myeloid acute, remission induction, tretinoin. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3474-3474
Author(s):  
Lijuan Xia ◽  
Min Lu ◽  
Duo Chen ◽  
Samuel Waxman ◽  
Yongkui Jing

Abstract Arsenic trioxide (As2O3) is a drug used world-wide that selectively causes the death of acute promyelocytic leukemia (APL) cells by novel mechanisms and induces complete clinical remission in 90% of patients without significant toxicity. The outstanding success of As2O3 therapy in relapsed APL patients has not been replicated by an equivalent success in other types of acute myeloid leukemia (AML). We have studied As2O3 mechanism(s) of action and identified agents to use in combination to improve the use of As2O3 as a treatment for other types of AML. As2O3 produces higher levels of H2O2 and apoptosis in APL NB4 cells than in other AML cells at therapeutic concentrations of 1-2 uM. As2O3 does not induce apoptosis in HL-60 cells but is synergistic with ascorbic acid (AA) or epigallocatechin-3-gallate (EGCG). Both AA and EGCG produce H2O2 which is augmented by the addition of As2O3. Apoptosis induction by As2O3 in combination with AA or EGCG is inhibited by catalase and the antioxidant N-acetylcysteine. Myeloperoxidase (MPO), a major neutrophil enzyme, augments H2O2-induced apoptosis by converting it into more potent reactive oxygen species. HL-60, NB4, SKNO-1 and PLB985 cells which express high level of MPO, but not U937 and K562 cells without expression of MPO, are responsive to As2O3 plus AA or EGCG-induced apoptosis. HP-100 cells, a subclone of HL-60 cells without MPO expression, are resistant to both combination treatments. MPO stable transfection sensitizes K562 cells to apoptosis following treatment with As2O3 and AA or EGCG. NADPH oxidase is an enzyme complex which generates O2− and H2O2 in neutrophils. In APL NB4 cells, As2O3 induces expression of NADPH oxidase members and is thought to participate in H2O2 production required for As2O3 response. X-CGD cells, a subclone of PLB-985 with targeted disruption of the gp91phox gene (Zhen et al, PNAS90:9832, 1993) and loss of NADPH oxidase activity but with high levels of MPO activity, are responsive to treatment with As2O3 and AA or EGCG. These results suggest that AA and EGCG enhance As2O3-induced apoptosis through a synergistic production of H2O2 which is independent NADPH oxidase activation in non-APL AML cells. MPO augments the apoptotic effect of H2O2 produced by As2O3 plus AA or EGCG and it could be used as a marker to predict the sensitivity of AML cells to both combination treatments.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2151-2151
Author(s):  
Kensuke Kojima ◽  
Teresa McQueen ◽  
Elizabeth J. Shpall ◽  
Rui-Yu Wang ◽  
Michael Andreeff

Abstract Abstract 2151 Fms-like tyrosine kinase-3 (FLT3) inhibitors have recently been introduced to overcome the dismal prognosis of acute myeloid leukemia (AML) with FLT3/ITD mutations. However, while ciculating blasts are rapidly eliminated, bone marrow (BM) responses are in general less impressive (Zhang et al., JCI 2009). One potential explanation for the reduced bone marrow response compared to the striking activity against circulating blast cells may be microenvironmental resistance to FLT3 inhibitors, including protection of FLT3/ITD+ blasts through the SDF-1 (CXCL12)/CXCR4 axis (Zeng et al., BLOOD 2009) and novel strategies to overcome microenvironmental resistance may further enhance the clinical benefit of FLT3 inhibitors. Here we investigated the role of p53 in bone marrow stromal cells in stromal cell-mediated resistance to FLT3 inhibition in FLT3/ITD AML. We confirmed that Mdm2-p53 interaction and mutant FLT3 signaling in leukemic blasts were inhibited by the Mdm2 inhibitor Nutlin-3a and the selective FLT3 inhibitor FI-700, respectively, as previously reported (Kojima et al., BLOOD 2004; Kojima et al., LEUKEMIA 2009). BM samples were obtained from normal individuals or from FLT3/ITD AML patients with more than 70% leukemia cells after informed consent. BM-derived stromal cells were seeded in 12-well plates in MEM-alpha medium and exposed to Nutlin-3a for 24 hours, gamma-irradiated (2 Gy), or treated with 100 nM doxorubicin. The wells were then washed three times and AML cells were added. Cell cycle distribution was determined by propidium iodide staining and apoptosis by Annexin V binding. In FLT3/ITD AML cell lines and primary cells, apoptosis was induced by FI-700, but apoptosis induction was diminished under stromal coculture conditions (43.2 ± 0.5% versus 17.0 ± 1.2% in MOLM-13; p < .01). FI-700–induced reduction of Mcl-1 protein levels and activation of Bax were essentially abrogated when FLT3/ITD AML cells were co-cultured with MSC (20.9 ± 1.6% versus 8.3 ± 0.3% cells with activated Bax levels in MOLM-13;p < .01). Separation of MOLM-13 cells from stroma by 0.4 μM culture filters did not affect the protective effect of stromal cells (21.2 ± 2.3% versus 21.6 ± 1.6%). The protection appeared to be mediated partially by SDF-1/CXCR4 signaling because the addition of recombinant SDF-1 to control medium protected FLT3/ITD AML cells from FI-700–induced apoptosis while the addition of anti-CXCR4 antibody (R&D Systems) partially abrogated the protection conferred by stromal cells. The protective effect of stromal cells on MOLM-13 was significantly reduced when MSC were exposed to p53 inducers including Nutlin-3a (17.0 ± 1.2% versus 26.0 ± 1.8%; p < .01), gamma-irradiation (13.5 ± 3.0% versus 18.6 ± 3.1%; p < .05) or doxorubicin (14.2 ± 3.0% versus 19.7 ± 0.9%; p < .05), although p53 activation was not itself cytotoxic to stromal cells. However, SDF-1 mRNA and protein levels were reduced in p53-activated stroma cells. Our findings confirm that selective FLT3 inhibition may not eradicate FLT3/ITD AML cells protected by stroma and show, for the first time, that p53 activation in stroma cells blunts stroma cell-mediated resistance to a kinase, here a FLT3 inhibitor, in part through regulation of SDF-1. Results suggest that combinations of HDM2/p53 (R7112/Nutlin-3a, Andreeff et al. ASH 2010) and/or CXCR4 inhibitors (Plerixafor, Konopleva et al. ASH 2010) with FLT3 inhibitors should be investigated in clinical trials targeting FLT3 mutant leukemias, with the dual goal of inducing apoptosis in leukemic cells and, concomittantly, reduce the protective effects exerted by the marrow microenvironment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1542-1542
Author(s):  
Yoshitaka Sunami ◽  
Marito Araki ◽  
Soji Morishita ◽  
Yumi Hironaka ◽  
Yoko Edahiro ◽  
...  

Abstract Abstract 1542 Tenovin-6, an inhibitor for nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylase Sirtuins, shows cytotoxicity to cancerous cells and thus recognized as a potential therapeutic compound for cancer treatment (Lain S., et al. (2008) Cancer Cell. 13,454–63). Since there is limited information for the cytotoxic property of this compound for hematopetic malignancies, we treated an APL (acute promyelocytic leukemia) cell line NB4 with Tenovin-6. As expected, Annexin V assay revealed that Tenovin-6 induced apoptosis in NB4 cells. However, to our surprise, at modest concentration, Tenovin did not induce cell death, rather inhibited NB4 cell proliferation and altered cell morphology. The fluorescence-activated cell sorting (FACS) analysis revealed that tenovin-6-treated NB4 cells are positive for CD11b and CD36 with decreased level of CD13 and CD33. Moreover, tenovin-6-treated NB4 cells presented nitroblue tetrazolium reduction capacity, suggesting that tenovin-6 induced monocytic differentiation in NB4 cells. To assess how Tenovin-6 induces cellular differentiation in NB4 cells, we investigated downstream target of Sirtuins. Although Tenovin-6 reportedly promotes acetylation of p53 by inhibiting SirT1, a founder of Sirtuin family proteins, the acetylation status of p53 is unchanged at the concentration where we observed differentiation. This suggests that Tenovin-6 induces NB4 differentiation through inhibiting other Sirtuin family proteins. These findings demonstrate a potential of Tenovin-6 as a differentiation-inducing reagent in APL cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 947-947
Author(s):  
Saravanan Ganesan ◽  
Ezhilarasi Chendamarai ◽  
Jayandharan G Rao ◽  
Sangeetha Hareendran ◽  
Ansu Abu Alex ◽  
...  

Abstract Abstract 947 About 5–10% newly diagnosed and about 20–30% of relapsed acute promyelocytic leukaemia (APL) patients will have disease recurrence after receiving currently accepted standards of care. There are a limited number of drugs in the armamentarium for treatment of APL. Preliminary work from our laboratory suggests that stromal cell adhesion mediated drug resistance (CAM-DR) is probably significant with arsenic trioxide (ATO) and is seen both in APL cell lines (n=8; figure 1A) and primary APL cells (n=26; data not shown). Preliminary data suggests that bortezomib (Bo) has cytotoxic effect against promyelocytes and is also known to interfere with stromal and malignant cell interaction in other haematological malignancies. Our in-vitro experiments suggest that Bo at pharmacologically relevant doses has a cytotoxic effect on APL (NB4) cell lines and primary APL cells (median IC50 = 9.8 ng/ml and 4.2 ng/ml, respectively) but has no effect at these doses on stromal cells (MSC) and mononuclear cells. We have also noted a significant synergistic cytotoxic effect when combined with ATO on NB4 cells (n = 4), P=0.05 (figure 1B). In co-culture with MSC, a combination of ATO at 2 μmol and Bo over a range of pharmacologically relevant concentrations (20- 80ng/ml) significantly reduced the APL cell viability [NB4 cells (n=8; figure1A) and primary APL cells (n=26; data not shown), suggesting that Bo overcomes the CAM-DR to ATO in APL. Moreover, we noted increased activation of NF-κB (p65) and over expression of Vascular cell adhesion molecule 1 (VCAM1) in NB4 cells when they were co-cultured with MSC. These effects on NF-κB and VCAM1expression were inhibited by Bo (figure 1C and 1D) and ATO. Bo treatment, in comparison to untreated cells and cells treated with ATO alone, reduced the capacity for serial colony forming units (CFU) of NB4 cells in methyl cellulose medium (n=2). A phase I clinical study was initiated combining ATO with weekly Bo at standard doses. A total of 3 patients have been treated and included a patient in relapse 1 and two in relapse 2. Median age was 31 years and there were 3 males. Two patients had previously under gone an autologous SCT for relapsed disease. Two patients were initially treated with a single agent ATO regimen while one patient had received a conventional ATRA plus chemotherapy regimen, this patient had subsequently had salvage chemotherapy, an autologous stem cell transplant and had relapsed; he had already exceeded the accepted safe cumulative dose of anthracycline at the time of his last relapse. All 3 patients achieved CR at a median of 59 days (range: 38–73) and all achieved a molecular remission at a median of 42 days (range: 39 – 56 days). Combination therapy was well tolerated; one second relapse patient had prolonged cytopenia while non hematological toxicity was mild and reversible and included one patient developing a transient grade II hepatotoxicity. None of the cases had evidence of significant neuropathy, worsening of coagulopathy or a differentiation syndrome. In summary Bo has direct cytotoxicity on malignant promyelocytes, synergizes with ATO. It is also capable of overcoming CAM-DR to ATO, which is probably mediated by down regulation of the NF-κB pathway and its downstream effects, such as expression of VCAM-1. In a phase I clinical study ATO and Bo combination was well tolerated and the preliminary outcome data points towards significant efficacy. Disclosures: Off Label Use: Bortezomib.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4763-4763
Author(s):  
Xin-Yan Pei ◽  
Michael W Sanderson ◽  
Leena E Youssefian ◽  
Jessica Felthousen ◽  
Lora B Kramer ◽  
...  

Abstract Introduction: Multiple myeloma (MM) is characterized by deregulation of members of the Bcl-2 family of apoptotic regulatory proteins. This has led to the development of BH3-mimetics such as ABT-737 which inhibits Bcl-2/xL but not Mcl-1. Previously, we reported that simultaneous inhibition of Chk1 and MEK1/2 dramatically induced apoptosis in cultured and primary MM cells, including cells resistant to conventional agents, while sparing their normal counterparts (Pei et al., Blood 2007, 2011). Recently, we reported that this strategy circumvented MM cell resistance conferred by overexpression of Mcl-1, an important survival factor in this disease (Pei et al., PLoS One 2014). However, Bcl-2 overexpression confers significant resistance to the Chk1/MEK1/2 inhibition strategy. This raised the possibility that BH3-mimetics targeting Bcl-2 might circumvent this resistance mechanism. The purpose of the present studies was to determine whether BH3-mimetics could overcome such resistance while preserving anti-myeloma selectivity. An additional goal was to employ a new mathematical model to characterize interactions combining three novel agents that coordinately inhibit multiple survival signaling pathways. Methods: Various parental and Bcl-2 or Bcl-xL-over-expressing MM cell lines, as well as primary CD138+ MM cells were employed. ABT-737, the MEK1/2 inhibitor PD184352 (PD), and the Chk1/Wee1 inhibitor (Chk1i) were obtained from Abbott, Millipore and Calbiochem, respectively. Cells were exposed to agents alone or in various combinations for 4 -72 h, after which effects on apoptosis and signaling pathways were determined. Results: Co-administration of ABT-737 potentiated PD/Chk1i-mediated lethality in multiple parental MM cell lines, in association with Mcl-1 down-regulation, Bim up-regulation, and increased DNA damage (ΥH2A.X). Consistent with earlier findings, ectopic expression of Bcl-2 or Bcl-xL protected MM cells from the PD/Chk1i regimen. However, co-administration of ABT-737 significantly restored sensitivity towards PD/Chk1i lethality. Mathematical modeling indicated 3-agent synergistic interactions, particularly in Bcl-2 overexpressing cells. PD/Chk1i exposure inhibited phosphorylation (T705 and S727) of Stat3, another important survival factor for MM cells, while cells expressing constitutively active Stat3 (CA-STAT3) exhibited resistance to this regimen. However, the latter event was reversed by co-exposure to ABT-737. Moreover, combining ABT-737 with PD/Chk1i resulted in release of Bim from anti-apoptotic proteins including Bcl-2, Bcl-xL, and Mcl-1, accompanied by Bak and Bax conformational change (activation). Knock-down of Bim by shRNA significantly protected cells from apoptosis induced by the 3-agent combination, indicating a functional role for Bim in anti-MM activity of this regimen. Furthermore, similar interactions, together with down-regulation of pStat3, were also observed in bortezomib-resistant MM cells, as well as in patient-derived CD138+ MM cells. In contrast, the regimen was minimally toxic to normal cord blood CD34+ cells or CD138- bone marrow cells. Finally, co-culture of parental or bortezomib-resistant MM cells with HS-5 stromal cells induced up-regulation of pStat3, while treatment with ABT-737 in combination with PD/Chk1i prevented Stat3 activation and robustly induced apoptosis despite the presence of stromal cells. Conclusion: ABT-737 co-administration synergistically potentiates the lethality of the PD/Chk1i regimen in MM cells, including bortezomib-resistant and primary MM cells, but not in normal hematopoietic progenitors. It also overcomes PD/Chk1i resistance conferred by overexpression of Bcl-2 or Bcl-xL, as well as by microenvironmental factors. Mechanisms responsible for these interactions are likely to be multi-factorial, including inactivation of Stat3, up-regulation of Bim, release of Bim from Bcl-2, Bcl-xL, and Mcl-1, and activation of Bak and Bax. Collectively, these findings demonstrate that combining BH3-mimetics with Chk1/MEK1/2 inhibition circumvents multiple forms of drug resistance in MM cells while exhibiting minimal toxicit toward normal hematopoietic cells. They also argue that a strategy targeting three coordinate survival signaling pathways may be highly effective in killing MM cells, particularly those resistant to current anti-MM therapies. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 4 (4) ◽  
Author(s):  
Sabatina Windyaningrum ◽  
◽  
Tri Yudani Mardining Raras ◽  
Bambang Rahardjo ◽  
Rose Khasana Dewi

Background: kefir is a fermented milk product that demonstrates numerous health benefits including antioxidant and immunomodulatory. Aim: to study the protective effect kefir on the expression of estrogen receptor alpha (ERα) in endometrial stromal cells and endometrial thickness on female rats that were exposed to arsenic. Methods: twenty-five female Wistar rats (Rattus norvegicus) were divided into five groups (CRL, As, T1, T2, T3). Control group (given a normal diet), As group (given the normal diet and exposed to arsenic trioxide 2 mg/kgBW/day). The T1; T2; T3 were exposed to arsenic trioxide 2 mg/kgBW/day and treated with different doses of kefir (1.25; 2.5; and 5 mL/kgBW/day, respectively) for 35 days. The rats of group As treated with arsenic trioxide only and group CRL served as control with normal feed in water. Cytological samples were taken after 35 days of treatment and examined every day to see the rat oestrus phase, and the proestrus phase of the oestrous cycle was chosen for termination. Uterine tissue fixed in 10% neutral buffered formalin for tissue preparation. ERα expression in endometrial stromal cells was analized using immunohistochemistry method, endometrial thickness was observed using histopathological methods. Results: significant reduction of ERα expression in endometrial stromal cells and endometrial thickness in female rats exposed to arsenic were observed in groups on treated rats (p ≤ 0.000; 0.009, respectively). Conclusion: the administration of kefir in female Wistar rats exposed to arsenic had shown significantly differences on ERα expressions and endometrial thickness. The smallest dose of kefir (1.25 mL/kgBW/day) could increase ERα expression and endometrial thickness in female Wistar rats with arsenic exposure. Therefore kefir has protective effect related to female reproductive system.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2836-2836
Author(s):  
Bulent Ozpolat ◽  
Ugur Akar ◽  
Isabel Zorilla-Calancha ◽  
Pablo Vivas-Mejia ◽  
Gabriel Lopez-Berestein

Abstract All-trans Retinoic Acid (ATRA) is a naturally occurring metabolite of retinol (vitamin A)and acts as a potent inducer of cellular differentiation and growth arrest in acute promyelocytic leukemia (APL), a type of acute myeloid leukemia (M3-AML). APL is characterized by translocation t(15;17), fusing PML (promyelocytic leukemia) and RARα (retinoic acid receptor) genes, leding to expression of PML/RARα receptor protein and differentiation block. Arsenic trioxide (ATO) induces (<0.5 μM) differentiation at low doses and apoptosis at high doses (>1 μM) in APL cells. Currently, both ATRA and ATO are successfully used in the treatment of APL in the clinic. However, the molecular mechanisms of myeloid differentiation and apoptosis induced by these agents are not fully understood. We previously reported that ATRA inhibits the translation initiation through multiple mechanisms, including upregulation of translation initiation inhibitors, DAP5/p97 and PDCD4 tumor suppressor protein. Here we investigated the role and regulation of death associated protein-5 (DAP5/p97/NAT1), a novel inhibitor of translational initiation, in myeloid (granulocytic and monocytic) cell differentiation and apoptosis. We found that ATRA (1 μM) induced a marked DAP5/p97 protein and mRNA expression during granulocytic differentiation of NB4 and HL60 cells but not in differentiation-resistant cells, which express very low levels of DAP5/p97. DAP5/p97 was translocated into nucleus during the differentiation of NB4 cells induced ATRA. At differentiation inducing doses, ATO, dimethysulfoxide, 1,25-dihydroxy-vitamin-D3, and phorbol-12-myristate-13-acetate also induced a significant DAP5/p97 expression in NB4 cells. However, ATO at apoptotic doses, but not ATRA, induced DAP5/p86, a proapoptotic form of DAP5/p97. ATRA and ATO -induced expression of DAP5/p97 was associated with inhibition of phosphaditylinositol 3-kinase (PI3K)/Akt pathway, which is known to stimulate cap-dependent translation of mRNAs. To show direct link between PI3K/Akt/mTOR pathway and DAP5 expression, we treated cell with PI3K and mTOR inhibitors LY294002 and by rapamycin, respectively. We found that inhibition of PI3K/Akt/mTOR pathway upregulated DAP5/p97 expression in NB4 cells. Finally, knockdown of DAP5/p97 expression by small interfering RNA significantly inhibited ATRA-induced granulocytic differentiation detected by expression of CD11b and ATO-induced apoptosis in NB4 cells detected by Annexin V assay (p<0.05). In conclusion, our data suggest that DAP5/p97 plays a role in ATRA-induced differentiation and ATO-induced apoptosis in APL cells. Our data demonstrated for the first time that DAP5/p97 is constitutively suppressed by of PI3K/Akt/mTOR pathway, and ATRA and ATO-induced expression of DAP5 is mediated by the inhibition of this survival pathway, suggesting a novel mechanism of DAP5 regulation and a role of translational control in induction of differentiation and apoptosis. Figure Figure


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3858-3858
Author(s):  
Yeung-Chul Mun ◽  
Jee-Young Ahn ◽  
Eun-Sun Yoo ◽  
Jungwon Huh ◽  
Kyoung Eun Lee ◽  
...  

Abstract Backgrounds The Arsenic trioxide (ATO) is an effective cancer therapeutic drug for acute promyelocytic leukemia (APL). ATO exerts its effect mainly raising oxidative stress. However, not only the mechanisms of reactive oxygen species (ROS) generation by ATO but involvement of redox enzymes including peroxiredoxin (PRX) and thioredoxin (TRX) remains elusive. Aim of current study is to elucidate the mechanism of redox enzymes to elevate ROS during ATO-induced apoptosis in APL-derived NB4 cells. Methods NB4 cell line, which is one of the human acute promyelocytic leukemia cell lines, was cultured in RPMI-1640 medium supplemented with 10% FBS in CO2 humidified atmosphere at 37°C. NB4 cells were cultured with 2 μM arsenic trioxide to induce apoptosis for 16-48 hours. Apoptosis was measured by staining with 7-amino-actinomycin D (7-AAD) with flow cytometry. 2, 7-dichlrodihydro-fluorescein-diacetate (H2DCF-DA) and MitoSOX Red were used to detect cellular and mitochondrial ROS. SO2 form for PRX I, PRX II, and PRX III was detected by western blot assay using PRX SO2 form-specific antibody. Monomer/Dimer assay for PRX I, PRX II, PRX III, and TRX I was performed by western blot using non-reducing gel. Results Intracellular ROS of NB4 cells was increased significantly after 16 hour of ATO but decreased after 24 hour of ATO. Mitochondrial ROS of NB4 cells was increased significantly after 39 hour of ATO. Apoptosis of NB4 cell after ATO treatment was increased as time elapsed (24% on 16hr, 26% on 24hr, 48% on 39hr, and 60% on 48hr). Monomer, indicated active and reduced form, of peroxiredoxins was decreased and cysteine sulfinic acid (CP–SO2H) peroxiredoxins, indicated inactive and oxidized peroxiredoxins, was increased in NB4 cells after ATO treatment as time goes by. Similarily, monomer of thioredoxin-1 (active thioredoxin) was decreased and multimer of thioredoxin-1 (inactive thioredoxin) was increased in NB4 cells after ATO treatment as time elapsed. Conclusions Our data showed inactivation of peroxiredoxins by oxidation was developed during ATO-induced ROS generation and APL cell apoptosis. These peroxiredoxins oxidation was probably due to increment of reduced thioredoxin in NB4 cells after ATO treatment. These findings suggest ATO-induced anti-leukemic activity is more likely due to a TRX system-mediated cellular redox changes. Our study may provide the insights for finding novel targets in the development of new therapies, which potentiate ATO-induced apoptosis in APL cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5217-5217
Author(s):  
Yeung-Chul Mun ◽  
Jee-Young Ahn ◽  
Eun-Sun Yoo ◽  
Kyoung Min Cho ◽  
Kyoung Eun Lee ◽  
...  

Abstract Backgrounds: The Arsenic trioxide (ATO) is an effective cancer therapeutic drug for acute promyelocytic leukemia (APL), but in some cases, APL cells are resistant to ATO treatment. ATO exerts its effect mainly raising oxidative stress. However, not only the mechanisms of reactive oxygen species (ROS) generation by ATO but involvement of redox enzymes including peroxiredoxin (PRX) during ATO-induced apoptosis and its resistance remain elusive. Recently, Rhee et al had reported that PRX III and sufiredoxin together protect mice from pyrazole-induced oxidative liver injury was found (Antioxid & Redox Signal, 2012:17:1351-1361). Aims of current study are to elucidate that the changes of redox enzyme could be a mechanism of anti-leukemia effect in APL-derived NB4 cells during ATO treatment and to find ways to potentiate the anti-leukemic effects of ATO on APL cells. Methods: NB4, one of the human acute promyelocytic leukemia cell lines, was treated with 0~10 μM arsenic trioxide to induce apoptosis for 16-48 hours in RPMI-1640 medium supplemented with 10% FBS in CO2humidified atmosphere at 37°C. Apoptosis was measured by staining with 7-amino-actinomycin D (7-AAD) with flow cytometry. 2, 7-dichlrodihydro-fluorescein-diacetate (H2DCF-DA) and MitoSOX Red was used to detect cellular and mitochondrial ROS. SO2 form for PRX I, PRX II, and PRX III was detected by western blot assay using PRX SO2 form-specific antibody. Sulfiredoxin (SRX) and caspase 3, 9 were also detected by western blot analysis. To evaluate the effect of SRX depletion, NB4 cells were transfected with small interfering RNA (siRNA). Results: Intracellular ROS of NB4 cells was increased significantly after 16 hour of ATO treatment but decreased after 24 hour of ATO treatment. Mitochondrial ROS of NB4 cells was increased significantly after 39 hour of ATO treatment. Apoptosis of NB4 cell after ATO treatment was increased as time elapsed (24% on 16hr, 26% on 24hr, 48% on 39hr, and 60% on 48hr). Increased cysteine sulfinic acid (Cys–SO2H) PRX III, inactive and oxidized form, was observed as a hyperoxidation reaction in NB4 cells after ATO treatment in concordance with mitochondrial ROS increment of NB4 cells. Increased expressions of cleaved caspase-9 and cleaved caspase-3 were also observed during NB4 cell apoptosis by ATO treatment. Meanwhile, SRX expression was increased in NB4 cells after ATO treatment. Down regulation of SRX by siRNA promoted ROS generation and apoptosis in ATO-treated NB4 cells. Conclusions: Our data showed inactivation of PRX III by Cys–SO2H formation as hyperoxidation is developed during ATO-induced mitochondrial ROS generation and apoptosis process in APL cells. In addition, ATO promotes expression of SRX, which is known as reducing enzyme of Cys–SO2H PRX and which leads to down regulation of ROS accumulation in APL cells. These findings might be due to protective effect of SRX from ATO on mitochondrial oxidative stress. These findings suggest ATO-induced anti-leukemic activity could be down regulated by an enhancing PRX III reduction after ATO-induced SRX activation. Currently, the effect of down regulation of SRX by siRNA are being investigated to amplify the apoptosis in ATO-treated NB4 cells. Our study may provide the insights for finding novel targets in the development of new therapies, which potentiate ATO-induced apoptosis in APL cells. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document