scholarly journals Targeting the MAPK/ERK and PI3K/AKT Signaling Pathways Affects NRF2, Trx and GSH Antioxidant Systems in Leukemia Cells

Antioxidants ◽  
2020 ◽  
Vol 9 (7) ◽  
pp. 633
Author(s):  
Ewa Jasek-Gajda ◽  
Halina Jurkowska ◽  
Małgorzata Jasińska ◽  
Grzegorz J. Lis

The mitogen-activated protein kinase (MAPK)/extracellular signal kinase (ERK) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signal transduction pathways have been implicated in the pathogenesis of leukemia. The aim of this study was to investigate the effect of the combination of ERK1/2 inhibitor AZD0364 and PI3K inhibitor ZSTK474 on acute lymphoblastic leukemia (ALL) REH, MOLT-4, acute myeloid leukemia (AML) MOLM-14, and chronic myeloid leukemia (CML) K562 cell lines. To evaluate the interactions of the drugs, cells were treated for 48 h with AZD0364 or ZSTK474 alone and in combination at fixed ratios. The combinatorial effects of both inhibitors were synergistic over a wide range of concentrations in REH, MOLT-4, and MOLM-14 cell lines. However, in K562 cells, the effects were found to be antagonistic. Furthermore, AZD0364 and ZSTK474 significantly decreased both ERK1/2 and AKT activation in REH, MOLT-4, and MOLM-14 cells. The results showed that incubation with both AZD0364 and ZSTK474 inhibited cell viability, increased reactive oxygen species (ROS) production, and induced apoptosis in leukemia cells. We observed that combined treatment with AZD0364 and ZSTK474 affected nuclear factor-κB (NF-κB) and antioxidant protein levels: NF-E2-related factor 2 (NRF2), heme oxygenase-1 (HO-1), thioredoxin (Trx), thioredoxin reductase (TrxR), and the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio. These effects were accompanied with decreased antiapoptotic survivin protein level. However, distinct cell line dependent effects were observed. In conclusion, the combination of AZD0364 and ZSTK474 can exert a synergistic anticancer effect in ALL and AML cells, which is associated with the induction of oxidative stress and the involvement of cellular antioxidant defense mechanisms.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1274-1274 ◽  
Author(s):  
Shin-ichiro Fujiwara ◽  
Tadashi Nagai ◽  
Satoru Kikuchi ◽  
Mitsuyo Uesawa ◽  
Chihiro Sakurai ◽  
...  

Abstract Abstract 1274 Poster Board I-296 Introduction RCAN1 (also called DSCR1, calcipressin 1, MCIP1, Adapt78) is known to be a regulator of calcineurin, a Ca2+/calmodulin-dependent protein phosphatase, and has also been shown to regulate the activity of certain transcription factors, including NF-AT and NF-kB. RCAN1 expression is widely found in various tissues, including the brain, kidney, liver and heart, but the expression level of RCAN1 is extremely low in normal hematopoiesis-related organs (bone marrow and spleen). Although RCAN1 has been shown to act as a negative regulator of FceRI-induced mast cell activation and NF-AT signaling in megakaryocytes, the cellular function of RCAN1 in myeloid cells remains unknown. Recently, we found that RCAN1 was highly expressed in acute phase of chronic myeloid leukemia (CML) by DNA microarray analysis. In this study, to explore the role of RCAN1 in leukemogenesis, we compared RCAN1 expression in leukemia cells and that in normal hematopoietic cells. We also analyzed regulatory mechanisms of RCAN1 gene promoter function and searched for novel RCAN1-interacting molecules in myeloid leukemia cells. Methods Expression of RCAN1 was examined by quantitative PCR using normal human bone marrow cells, bone marrow cells from patients with acute myeloid leukemia (AML) and patients with CML, and various human hematological tumor cell lines. Promoter activity of the human RCAN1 gene was evaluated by a luciferase assay using various deletion mutants in KCL22 cells. To identify novel RCAN1-interacting proteins, a yeast two-hybrid assay was performed using a cDNA library from K562 leukemia cells. Results Fractions of CD34+CD38-, CD34+CD38+, CD33+CD14-CD16-, CD33+CD14+ and CD33+CD16+ cells from normal human bone marrow showed extremely low levels of RCAN1, indicating that RCAN1 is scarcely expressed in normal hematopoietic cells, including stem cells and progenitor cells. In contrast, bone marrow mononuclear cells (BMMC) from 15 of 16 patients with AML and all four patients with CML in acute phase showed substantial levels of RCAN1. Furthermore, RCAN1 was expressed in all human cell lines examined in this study that were derived from AML (THP-1, U937, HL60, KY821), CML in acute phase (KCL22,K562, KU812), acute lymphoblastic leukemia (Jarkat, Raji) and multiple myeloma (RPMI8226, U266). Interestingly, expression levels of RCAN1 were significantly decreased in BMMC from three of four AML patients when the patients were in complete remission. These results suggest that RCAN1 is extraordinarily expressed in a wide range of hematological tumor cells. To reveal the underlying mechanisms of ectopic expression of RCAN1, we next examined the promoter function of RCAN1 gene, which consists of seven exons. Although RCAN1 expresses several alternative isoforms that selectively include each of exons 1-4 and use distinct promoters, we found that only exon 1 isoform was expressed in these leukemia cells. Analysis of the function of human RCAN1 gene promoter upstream of exon 1 demonstrated that NKX2-5 binding site is important for basal activity of RCAN1 gene promoter in KCL22 cells. NKX2-5 is a homeobox-containing transcription factor and is involved in leukemogenesis of T-cell acute lymphoblastic leukemia. Our results raised the possibility that NKX2-5 plays a role in malignant transformation partly through expression of RCAN1 in myeloid leukemia cells. Finally, we searched for RCAN1-interacting proteins in leukemia cells. By screening a cDNA library from K562 cells using yeast two-hybrid assay, we found that RCAN1 interacts with HINT1, HINT2 and RINT1. HINT1 has been reported to be a tumor suppressor against hepatocellular carcinoma, and HINT2 and RINT1 have also been found to function as tumor suppressors. It would be interesting to clarify whether RCAN1 affects the activity of these molecules, and it is important to reveal the involvement of these molecules in leukemogenesis. Conclusion RCAN1 is extraordinarily expressed in a wide range of hematological malignant cells, and NKX2-5 might be important for its ectopic expression. It is possible that RCAN1 is biologically significant in leukemogenesis through affecting certain tumor suppressors. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3958-3958
Author(s):  
Chengyun Pan ◽  
Dan Ma ◽  
Qin Fang ◽  
Ping Liu ◽  
Jishi Wang

Backgroud and objective: Recurrence and resistance is still the biggest challenges for Acute lymphoblastic leukemia (ALL). In recent years, a new concept has been proposed that the interaction between bone marrow microenvironment and leukemia cells could reduce the sensitivity of leukemia cells to chemotherapy. As an important matric element of the bone marrow microenvironment, Cancerassociated fibroblasts (CAFs) can mediate changes in bone marrow microenvironment to promote tumor proliferation and infiltration, but its role in ALL has not been described. Heme Oxygenase-1 (HO-1) is a rate-limiting enzyme in the process of heme catabolism, which is highly expressed in leukemia cells and can promote chemo-resistance by regulating the preservation of hematopoietic stem progenitor cells in the bone marrow microenvironment according to the recent research. In this article, we explored that HO-1 may be a key factor for promoting cancer associated fibroblasts to mediated chemo-resistance of ALL in the bone marrow microenvironment. M ethods : For clinical sample analysis, Bone marrow nucleated cells of 16 ALL patients with complete remission (ALL-CR), 12 ALL with relapse/refractory (ALL-R/R) and 10 normal controls were btained from bone marrow puncture. The expression of HO-1 and CAFs markers which includes α-SMA, FAP and FSP-1 was examined by quantitative reverse transcription-PCR (RT-PCR) and Western blot. In vitro cell experiments, CAFs was obtained from transfusion of bone marrow-derived mesenchymal stem cells (BM-MSCs) with recombinant human TGF-β1 (rhTGF-β1) stimulating. We used ALL primary cells and ALL cell lines (Nalm-6/Super-B15) to culture alone or co-culture with CAFs to detect proliferation, apoptosis and cell cycle of leukemia cells. In addition, we transfected ALL cell lines by constructing siHO-1 lentiviral vector, and co-cultured with CAFs or cultured separately to detect the above index changes in leukemia cells. R esults : The expressions of α-SMA, FAP, FSP-1 and HO-1 were significantly higher in ALL-R/R patients than in ALL-CR group and normal control group (P<0.05), which suggested that CAFs and HO-1 are closely related to recurrence and resistance of ALL. In vitro tests, after rhTGF-β1 stimulated BM-MSCs for 48h, the mRNA and protein expression levels of α-SMA, FAP and FSP-1 were significantly higher than those in BM-MSCs alone (P<0.05), This step was used to obtain CAFs. Then proceed to CAFs and Nalm-6/Super-B15 cell lines or ALL primary cells co-cultured tests. Results showed that CAFs can enhanced the leukemia cells proliferation and decreased apoptosis, and leukemia cells arrested in the G0/G1 phase was increased. Interestingly, this effect could be decreased by silencing HO-1 expression in Nalm-6/Super-B15 cell lines, which suggested that HO-1 may be a key factor mediating the interaction between leukemia cells and CAFs in bone marrow microenvironment. Conclusion:As an important component of the bone marrow microenvironment, CAFs are closely related to the recurrence and resistance of ALL. HO-1 may be a key component mediating the interaction between CAFs and leukemia cells of ALL. This results may provide a new entry point for deepening the mechanism of ALL progression and finding more effective targeted therapies. But further in vitro and in vivo experiments are still needed for verification. Disclosures No relevant conflicts of interest to declare.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e13520-e13520 ◽  
Author(s):  
Yosuke Minami ◽  
Nobuaki Fukushima ◽  
Tomoki Naoe

e13520 Background: Aberrant activation of the Hedgehog (Hh) signaling pathway is involved in a variety of cancers, and required for maintenance of the leukemic stem cell population in several experimental systems. PF-04449913 (PF) is a novel oral small molecule inhibitor that targets Smoothened (SMO) in the Hh pathway. Treatment with PF has shown promising results regarding safety, tolerability, and early signs of efficacy in a phase 1 study of hematologic malignancies including acute myeloid leukemia (AML) (Jamieson C, et al. ASH, 2011). Methods: We used AML cell lines and primary AML cells in order to elucidate mechanisms and biomarkers in PF treatment. We also used a co-culturing system with HS-5 stromal cells, and an immunodeficient NOD/SCID/IL2rγnull (NOG) mouse model serially xenotransplanted with primary AML cells to evaluate effects of PF on AML propagation. Results: In vivo-treatment with PF attenuated leukemia-initiation potential in acute myeloid leukemia cells through the serial transplantation system, while limiting reduction of tumor burden in the primary leukemia system. Ex vivo-treatment with PF inhibited proliferation and minimally induced cell death in leukemia cell lines and primary AML cells increased expression of the myeloid differentiation marker, CD11b. In addition, PF treatment down-regulated mRNAs encoding downstream effector GLIs in the canonical Hh pathway using RQ-PCR assays and decreased nuclear expression of GLI-2 using immunofluorescence assays. Moreover, combined treatment with PF abrogated resistance to Ara-C in MOLM-14 cells co-cultured with HS-5 stromal cells. We are also investigating biomarkers in these models including CD markers (such as CD44) as well as the toxicity for normal cord blood cells with PF treatment. Conclusions: These results imply that PF treatment can attenuate leukemia-initiation potential in acute myeloid leukemia cells and improve AML therapy through overcoming the resistance to chemotherapy in the bone marrow microenvironment.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Federica Palombarini ◽  
Silvia Masciarelli ◽  
Alessio Incocciati ◽  
Francesca Liccardo ◽  
Elisa Di Fabio ◽  
...  

Abstract Background In recent years, the use of ferritins as nano-vehicles for drug delivery is taking center stage. Compared to other similar nanocarriers, Archaeoglobus fulgidus ferritin is particularly interesting due to its unique ability to assemble-disassemble under very mild conditions. Recently this ferritin was engineered to get a chimeric protein targeted to human CD71 receptor, typically overexpressed in cancer cells. Results Archaeoglobus fulgidus chimeric ferritin was used to generate a self-assembling hybrid nanoparticle hosting an aminic dendrimer together with a small nucleic acid. The positively charged dendrimer can indeed establish electrostatic interactions with the chimeric ferritin internal surface, allowing the formation of a protein-dendrimer binary system. The 4 large triangular openings on the ferritin shell represent a gate for negatively charged small RNAs, which access the internal cavity attracted by the dense positive charge of the dendrimer. This ternary protein-dendrimer-RNA system is efficiently uptaken by acute myeloid leukemia cells, typically difficult to transfect. As a proof of concept, we used a microRNA whose cellular delivery and induced phenotypic effects can be easily detected. In this article we have demonstrated that this hybrid nanoparticle successfully delivers a pre-miRNA to leukemia cells. Once delivered, the nucleic acid is released into the cytosol and processed to mature miRNA, thus eliciting phenotypic effects and morphological changes similar to the initial stages of granulocyte differentiation. Conclusion The results here presented pave the way for the design of a new family of protein-based transfecting agents that can specifically target a wide range of diseased cells. Graphic abstract


Cells ◽  
2021 ◽  
Vol 10 (3) ◽  
pp. 665
Author(s):  
Margot S.F. Roeten ◽  
Johan van Meerloo ◽  
Zinia J. Kwidama ◽  
Giovanna ter Huizen ◽  
Wouter H. Segerink ◽  
...  

At present, 20–30% of children with acute leukemia still relapse from current chemotherapy protocols, underscoring the unmet need for new treatment options, such as proteasome inhibition. Ixazomib (IXA) is an orally available proteasome inhibitor, with an improved safety profile compared to Bortezomib (BTZ). The mechanism of action (proteasome subunit inhibition, apoptosis induction) and growth inhibitory potential of IXA vs. BTZ were tested in vitro in human (BTZ-resistant) leukemia cell lines. Ex vivo activity of IXA vs. BTZ was analyzed in 15 acute lymphoblastic leukemia (ALL) and 9 acute myeloid leukemia (AML) primary pediatric patient samples. BTZ demonstrated more potent inhibitory effects on constitutive β5 and immunoproteasome β5i proteasome subunit activity; however, IXA more potently inhibited β1i subunit than BTZ (70% vs. 29% at 2.5 nM). In ALL/AML cell lines, IXA conveyed 50% growth inhibition at low nanomolar concentrations, but was ~10-fold less potent than BTZ. BTZ-resistant cells (150–160 fold) displayed similar (100-fold) cross-resistance to IXA. Finally, IXA and BTZ exhibited anti-leukemic effects for primary ex vivo ALL and AML cells; mean LC50 (nM) for IXA: 24 ± 11 and 30 ± 8, respectively, and mean LC50 for BTZ: 4.5 ± 1 and 11 ± 4, respectively. IXA has overlapping mechanisms of action with BTZ and showed anti-leukemic activity in primary leukemic cells, encouraging further pre-clinical in vivo evaluation.


Antioxidants ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 897
Author(s):  
Wen-Ping Jiang ◽  
Jeng-Shyan Deng ◽  
Shyh-Shyun Huang ◽  
Sheng-Hua Wu ◽  
Chin-Chu Chen ◽  
...  

Liver damage induced by paracetamol overdose is the main cause of acute liver failure worldwide. In order to study the hepatoprotective effect of Sanghuangporus sanghuang mycelium (SS) on paracetamol-induced liver injury, SS was administered orally every day for 6 days in mice before paracetamol treatment. SS decreased serum aminotransferase activities and the lipid profiles, protecting against paracetamol hepatotoxicity in mice. Furthermore, SS inhibited the lipid peroxidation marker malondialdehyde (MDA), hepatic cytochrome P450 2E1 (CYP2E1), and the histopathological changes in the liver and decreased inflammatory activity by inhibiting the production of proinflammatory cytokines in paracetamol-induced acute liver failure. Moreover, SS improved the levels of glutathione (GSH), superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase in the liver. Significantly, SS diminished mitogen-activated protein kinase (MAPK), Toll-like receptor 4 (TLR4), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt), and the nuclear factor-kappa B (NF-κB) axis, as well as upregulated the Kelch-like ECH-associated protein 1 (Keap1)/erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway, in paracetamol-induced mice. SS mainly inhibited the phosphorylation of the liver kinase B1 (LKB1), Ca2+/calmodulin-dependent kinase kinase β (CaMKKβ), and AMP-activated protein kinase (AMPK) protein expression. Furthermore, the protective effects of SS on paracetamol-induced hepatotoxicity were abolished by compound C, an AMPK inhibitor. In summary, we provide novel molecular evidence that SS protects liver cells from paracetamol-induced hepatotoxicity by inhibiting oxidative stress and inflammation.


2019 ◽  
Author(s):  
Katerina Hlozkova ◽  
Alena Pecinova ◽  
David Pajuelo Reguera ◽  
Marketa Simcikova ◽  
Lenka Hovorkova ◽  
...  

Abstract Background Effectiveness of L-asparaginase administration in acute lymphoblastic leukemia treatment is mirrored in overall outcome of patients. Generally, leukemia patients differ in their sensitivity to L-asparaginase; however, the mechanism underlying their inter-individual differences is still not fully understood. We have previously shown that L-asparaginase rewires the biosynthetic and bioenergetic pathways of leukemia cells to activate both anti-leukemic and pro-survival processes. Herein, we investigated the relationship between the metabolic profile of leukemia cells and their sensitivity to currently used cytostatic drugs.Methods Altogether, 19 leukemia cell lines and primary leukemia cells from 11 patients were used. Glycolytic function and mitochondrial respiration were measured using Seahorse bioanalyzer. Sensitivity to cytostatics was measured using MTS assay and/or absolute count and flow cytometry. Mitochondrial membrane potential was determined as TMRE fluorescence.Results We characterized the basal metabolic state of the cells derived from different leukemia subtypes using cell lines and primary samples and assessed their sensitivity to cytostatic drugs. We found that leukemia cells cluster into distinct groups according to their metabolic profile, which is mainly driven by their hematopoietic lineage of origin from which they derived. However, majority of lymphoid leukemia cell lines and patients with lower sensitivity to L-asparaginase clustered regardless their hematopoietic phenotype together with myeloid leukemias. Furthermore, we observed a correlation of specific metabolic parameters with sensitivity to L-asparaginase. Greater ATP-linked respiration and lower basal mitochondrial membrane potential in cells significantly correlated with higher sensitivity to L-asparaginase. No such correlation was found in other tested cytostatic drugs.Conclusions These data support the prominent role of the cell metabolism in the treatment effect of L-asparaginase. Based on these findings metabolic profile could identify leukemia patients with lower sensitivity to L-asparaginase with no specific genetic characterization.


Antioxidants ◽  
2020 ◽  
Vol 9 (10) ◽  
pp. 924
Author(s):  
Ni-Chun Kuo ◽  
Shieh-Yang Huang ◽  
Chien-Yi Yang ◽  
Hsin-Hsueh Shen ◽  
Yen-Mei Lee

Magnolol (MG) is the main active compound of Magnolia officinalis and exerts a wide range of biological activities. In this study, we investigated the effects of MG using tyloxapol (Tylo)-induced (200 mg/kg, i.p.) hyperlipidemia in rats and palmitic acid (PA)-stimulated (0.3 mM) HepG2 cells. Our results showed that Tylo injection significantly increased plasma levels of triglyceride and cholesterol as well as superoxide anion in the livers, whereas MG pretreatment reversed these changes. MG reduced hepatic lipogenesis by attenuating sterol regulatory element-binding protein-1c (SREBP-1c) and fatty acid synthase (FAS) proteins and Srebp-1, Fas, Acc, and Cd36 mRNA expression as well as upregulated the lipolysis-associated genes Hsl, Mgl, and Atgl. Furthermore, MG reduced plasma interleukin-1β (IL-1β) and protein expression of NLR family pyrin domain-containing 3 (NLRP3), apoptosis-associated speck-like protein (ASC), and caspase 1 as well as upregulated nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and induction of heme oxygenase-1 (HO-1) in hepatocytes of Tylo-treated rats. Enhanced autophagic flux by elevation of autophagy related protein 5-12 (ATG5-12), ATG7, Beclin1, and microtubule-associated protein light chain 3 B II (LC3BII)/LC3BI ratio, and reduction of sequestosome-1 (SQSTM1/p62) and phosphorylation of mTOR was observed by MG administration. However, autophagy inhibition with 3-methyladenine (3-MA) in HepG2 cells drastically abrogated the MG-mediated suppression of inflammation and lipid metabolism. In conclusion, MG inhibited hepatic steatosis-induced NLRP3 inflammasome activation through the restoration of autophagy to promote HO-1 signaling capable of ameliorating oxidative stress and inflammatory responses.


Sign in / Sign up

Export Citation Format

Share Document