Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Mol Med Rep ; 29(6)2024 06.
Article En | MEDLINE | ID: mdl-38695236

During hematopoiesis, megakaryocytic erythroid progenitors (MEPs) differentiate into megakaryocytic or erythroid lineages in response to specific transcriptional factors, yet the regulatory mechanism remains to be elucidated. Using the MEP­like cell line HEL western blotting, RT­qPCR, lentivirus­mediated downregulation, flow cytometry as well as chromatin immunoprecipitation (ChIp) assay demonstrated that the E26 transformation­specific (ETS) transcription factor friend leukemia integration factor 1 (Fli­1) inhibits erythroid differentiation. The present study using these methods showed that while FLI1­mediated downregulation of GATA binding protein 1 (GATA1) suppresses erythropoiesis, its direct transcriptional induction of GATA2 promotes megakaryocytic differentiation. GATA1 is also involved in megakaryocytic differentiation through regulation of GATA2. By contrast to FLI1, the ETS member erythroblast transformation­specific­related gene (ERG) negatively controls GATA2 and its overexpression through exogenous transfection blocks megakaryocytic differentiation. In addition, FLI1 regulates expression of LIM Domain Binding 1 (LDB1) during erythroid and megakaryocytic commitment, whereas shRNA­mediated depletion of LDB1 downregulates FLI1 and GATA2 but increases GATA1 expression. In agreement, LDB1 ablation using shRNA lentivirus expression blocks megakaryocytic differentiation and modestly suppresses erythroid maturation. These results suggested that a certain threshold level of LDB1 expression enables FLI1 to block erythroid differentiation. Overall, FLI1 controlled the commitment of MEP to either erythroid or megakaryocytic lineage through an intricate regulation of GATA1/GATA2, LDB1 and ERG, exposing multiple targets for cell fate commitment and therapeutic intervention.


Cell Differentiation , Erythroid Cells , Megakaryocytes , Humans , Cell Differentiation/genetics , Cell Line , Erythroid Cells/metabolism , Erythroid Cells/cytology , GATA1 Transcription Factor/metabolism , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , GATA2 Transcription Factor/genetics , Gene Expression Regulation , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Megakaryocytes/metabolism , Megakaryocytes/cytology , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Transcriptional Regulator ERG/metabolism , Transcriptional Regulator ERG/genetics
2.
BMC Cancer ; 24(1): 326, 2024 Mar 09.
Article En | MEDLINE | ID: mdl-38461240

BACKGROUND: FLI1 is an oncogenic transcription factor that promotes diverse malignancies through mechanisms that are not fully understood. Herein, FLI1 is shown to regulate the expression of Ubiquitin Associated and SH3 Domain Containing A/B (UBASH3A/B) genes. UBASH3B and UBASH3A are found to act as an oncogene and tumor suppressor, respectively, and their combined effect determines erythroleukemia progression downstream of FLI1. METHODS: Promoter analysis combined with luciferase assays and chromatin immunoprecipitation (ChIP) analysis were applied on the UBASH3A/B promoters. RNAseq analysis combined with bioinformatic was used to determine the effect of knocking-down UBASH3A and UBASH3B in leukemic cells. Downstream targets of UBASH3A/B were inhibited in leukemic cells either via lentivirus-shRNAs or small molecule inhibitors. Western blotting and RT-qPCR were used to determine transcription levels, MTT assays to assess proliferation rate, and flow cytometry to examine apoptotic index. RESULTS: Knockdown of FLI1 in erythroleukemic cells identified the UBASH3A/B genes as potential downstream targets. Herein, we show that FLI1 directly binds to the UBASH3B promoter, leading to its activation and leukemic cell proliferation. In contrast, FLI1 indirectly inhibits UBASH3A transcription via GATA2, thereby antagonizing leukemic growth. These results suggest oncogenic and tumor suppressor roles for UBASH3B and UBASH3A in erythroleukemia, respectively. Mechanistically, we show that UBASH3B indirectly inhibits AP1 (FOS and JUN) expression, and that its loss leads to inhibition of apoptosis and acceleration of proliferation. UBASH3B also positively regulates the SYK gene expression and its inhibition suppresses leukemia progression. High expression of UBASH3B in diverse tumors was associated with worse prognosis. In contrast, UBASH3A knockdown in erythroleukemic cells increased proliferation; and this was associated with a dramatic induction of the HSP70 gene, HSPA1B. Accordingly, knockdown of HSPA1B in erythroleukemia cells significantly accelerated leukemic cell proliferation. Accordingly, overexpression of UBASH3A in different cancers was predominantly associated with good prognosis. These results suggest for the first time that UBASH3A plays a tumor suppressor role in part through activation of HSPA1B. CONCLUSIONS: FLI1 promotes erythroleukemia progression in part by modulating expression of the oncogenic UBASH3B and tumor suppressor UBASH3A.


Leukemia, Erythroblastic, Acute , Proto-Oncogene Protein c-fli-1 , Humans , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/metabolism , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , RNA, Small Interfering/genetics , RNA-Binding Protein EWS/genetics , Protein Tyrosine Phosphatases/metabolism
3.
J Inflamm Res ; 16: 2007-2020, 2023.
Article En | MEDLINE | ID: mdl-37193069

Aim: Histamine decarboxylase (HDC) catalyzes decarboxylation of histidine to generate histamine. This enzyme affects several biological processes including inflammation, allergy, asthma, and cancer, although the underlying mechanism is not fully understood. The present study provides a novel insight into the relationship between the transcription factor FLI1 and its downstream target HDC, and their effects on inflammation and leukemia progression. Methods: Promoter analysis combined with chromatin immunoprecipitation (ChIp) was used to demonstrate binding of FLI1 to the promoter of HDC in leukemic cells. Western blotting and RT-qPCR were used to determine expression of HDC and allergy response genes, and lentivirus shRNA was used to knock-down target genes. Proliferation, cell cycle, apoptosis assays and molecular docking were used to determine the effect of HDC inhibitors in culture. An animal model of leukemia was employed to test the effect of HDC inhibitory compounds in vivo. Results: Results presented herein demonstrate that FLI1 transcriptionally regulates HDC by direct binding to its promoter. Using genetic and pharmacological inhibition of HDC, or the addition of histamine, the enzymatic product of HDC, we show neither have a discernable effect on leukemic cell proliferation in culture. However, HDC controls several inflammatory genes including IL1B and CXCR2 that may influence leukemia progression in vivo through the tumor microenvironment. Indeed, diacerein, an IL1B inhibitor, strongly blocked Fli-1-induced leukemia in mice. In addition to allergy, FLI1 is shown to regulate genes associated with asthma such as IL1B, CPA3 and CXCR2. Toward treatment of these inflammatory conditions, epigallocatechin (EGC), a tea polyphenolic compound, is found strongly inhibit HDC independently of FLI1 and its downstream effector GATA2. Moreover, the HDC inhibitor, tetrandrine, suppressed HDC transcription by directly binding to and inhibiting the FLI1 DNA binding domain, and like other FLI1 inhibitors, tetrandrine strongly suppressed cell proliferation in culture and leukemia progression in vivo. Conclusion: These results suggest a role for the transcription factor FLI1 in inflammation signaling and leukemia progression through HDC and point to the HDC pathway as potential therapeutics for FLI1-driven leukemia.

5.
Cancer Gene Ther ; 28(3-4): 265-278, 2021 04.
Article En | MEDLINE | ID: mdl-32873871

Multiple myeloma (MM) is still an incurable hematological malignancy, with even poorer prognosis in MM patients with distant invasion. The present study was designed to explore the effects of C3a and C5a on the migration, invasion, and adhesion of MM tumor cells and to investigate the underlying mechanisms. As a result, the levels of C3a and C5a in plasma of MM patients were significantly higher than those of healthy donors. Consistently, the expression of C3a and C5a receptors on myeloma cells of MM patients was also significantly higher than that on sorted plasma cells of normal donors. C3a and C5a have been confirmed to increase the migration, invasion and adhesion of MM cell lines by activating the MEK/ERK pathway and increasing the nuclear transfer of Nrf2 in vitro. Moreover, the MM cell line U266 with Nrf2 downregulation was incubated with C3a and C5a, followed by injection into the tail vein of NOD-SCID mice. We found that Nrf2 downregulation attenuated the migration of anaphylatoxin C3a and C5a to MM tumor cells in bone marrow, liver and lung in vivo. In conclusion, our results indicate that activation of the complement cascade in MM patients may contribute to the migration, invasion and adhesion of MM cells, and this type of tumor cells dissemination in MM is, at least partially, regulated by Nrf2. Thereby, complement suppression or Nrf2 downregulation might offer a novel therapeutic opportunity for MM.


Complement C3a/metabolism , Complement C5a/metabolism , Multiple Myeloma/metabolism , NF-E2-Related Factor 2/metabolism , Adult , Aged , Animals , Case-Control Studies , Female , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Multiple Myeloma/pathology , Neoplasm Metastasis , Survival Analysis , Transfection
6.
Int Immunopharmacol ; 80: 106008, 2020 Mar.
Article En | MEDLINE | ID: mdl-31978797

B-cell acute lymphoblastic leukemia (B-ALL) is immune to the chemotherapy-induced apoptosis as a result of the protection of bone marrow mesenchymal stromal cells (BMSCs). However, the precise underlying mechanism of such protection remains unclear so far. In this experiment, protein tyrosine phosphatase 2 (Shp2), which was encoded by the PTPN11 gene, was highly expressed in BMSCs of the newly diagnosed and the recurrent B-ALL patients. The plasmid-induced (including Shp2 E76K) Shp2 activation in BMSCs (Shp2-activated BMSCs) markedly increased the BMSCs-mediated resistance of leukemia cells both in vitro and in vivo. Additionally, studies in vitro suggested that, the expression of vascular cell adhesion molecule 1 (VCAM-1) was markedly up-regulated in Shp2-activated BMSCs, and VCAM-1 expression in BMSCs of B-ALL patients was negatively correlated with Shp2 expression. Down-regulation of VCAM-1 in BMSCs using siRNA reversed the resistance of CCRF-SB cells mediated by the Shp2-activated BMSCs. As for the molecular mechanism, the PI3K/AKT pathway mediated the regulation of VCAM-1 by Shp2. Blocking the very late antigen-4 (VLA-4) by antibodies in CCRF-SB cells dramatically reversed the resistance of CCRF-SB cells mediated by the Shp2-activated BMSCs, and decreased the adhesion effects of both CCRF-SB cells and BMSCs. In conclusion, Shp2 activation in BMSCs up-regulates VCAM-1 expression through increasing the PI3K/AKT phosphorylation level, and targeting the VCAM-1/VLA-4 signaling may serve as a clinically relevant mechanism to overcome the BMSCs-mediated chemoresistance of B-ALL cells.


Drug Resistance, Neoplasm/genetics , Mesenchymal Stem Cells/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Vincristine/pharmacology , Adolescent , Adult , Animals , Bone Marrow/pathology , Cell Line, Tumor , Child , Child, Preschool , Humans , Integrin alpha4beta1/metabolism , Mice , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Primary Cell Culture , RNA, Small Interfering/metabolism , Tumor Microenvironment , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Vincristine/therapeutic use , Xenograft Model Antitumor Assays , Young Adult
7.
Ann N Y Acad Sci ; 1467(1): 77-93, 2020 05.
Article En | MEDLINE | ID: mdl-31930541

Although imatinib (IM) has been demonstrated to be an efficient treatment in chronic myeloid leukemia (CML), some patients still experience IM resistance and disease relapse. Through in vitro studies, we observed that HDAC3 levels were elevated in BCR-ABL1 kinase-independent, IM-resistant primary cells from CML patients and in IM-resistant K562 (K562R) cells and that downregulation of HDAC3 could enhance IM efficacy in K562R cells. Furthermore, betulinic acid (BA), a lupane-type pentacyclic triterpenoid saponin isolated from birch trees, restored IM sensitivity in the BCR-ABL1 kinase-independent, IM-resistant primary cells and in K562R cells, as well as in primary CD34+ bone marrow cells from CML patients. We found that BA restored IM sensitivity through inhibition of HDAC3 accumulation in cells, and that this was mediated by BA-dependent ubiquitination and degradation of HDAC3. BA at low dosage significantly increased IM antitumor effects on murine xenografts bearing K562R cells and inhibited HDAC3 expression in tumor tissue. Our findings demonstrated that HDAC3 is an essential factor in BCR-ABL1 kinase-independent IM resistance, and that BA in combination with IM may be a novel treatment strategy for overcoming IM resistance in CML.


Bone Marrow Cells/drug effects , Drug Resistance, Neoplasm/drug effects , Histone Deacetylases/metabolism , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Triterpenes/pharmacology , Adult , Apoptosis/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/physiology , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , Pentacyclic Triterpenes , Signal Transduction/drug effects , Ubiquitination/drug effects , Young Adult , Betulinic Acid
8.
RSC Adv ; 10(2): 828-844, 2020 Jan 02.
Article En | MEDLINE | ID: mdl-35494464

Imatinib (IM) is utilized for targeting the BCR-ABL fusion protein and as such, chronic myeloid leukemia (CML) is considered to be a curable disorder for which patients can achieve a long survival. However, 15-20% CML cases end up with IM resistance that will develop into the accelerated stage and eventually the blast crisis, thereby restricting the treatment choices and giving rise to a dismal survival rate. Histone deacetylases (HDACs) have been identified to modulate the oncogene as well as tumor suppressor gene activities, and they play crucial parts in tumorigenesis. It is found recently that IM combined with HDAC inhibitors (HDACi) can serve as a promising means of overcoming IM resistance in CML cases. Santacruzamate A (CAY10683) has been developed as one of the selective and powerful HDACi to resist HDAC2. Therefore, in this study, we aimed to examine whether CAY10683 combined with IM could serve as the candidate antitumor treatment for CML cases with IM resistance. The influences of CAY10683 combined with IM on the cell cycle arrest, apoptosis, and viability of CML cells with IM resistance were investigated, and it was discovered that the combined treatment exerted synergistic effects on managing the IM resistance. Moreover, further studies indicated that CAY10683 combined with IM mainly exerted synergistic effects through inhibiting HDAC2 in K562-R and LAMA84-R cells with IM resistance. Besides, the PI3K/Akt signal transduction pathway was found to mediate the HDAC2 regulation of CML cells with IM resistance. Eventually, it was also discovered, based on the xenograft mouse model, that the combined treatment dramatically suppressed CML proliferation in vivo. To sum up, findings in the current study indicate that CAY10683 combined with IM can be potentially used as the candidate treatment for CML with IM resistance.

9.
Anticancer Drugs ; 31(2): 158-168, 2020 02.
Article En | MEDLINE | ID: mdl-31584454

Although arsenic trioxide (ATO) treatment has transformed acute promyelocytic leukemia (APL) from the most fatal to the most curable hematological cancer, many high-risk APL patients who fail to achieve a complete molecular remission or relapse become resistant to ATO. Herein, we report that 7-(4-(3-ethynylphenylamino)-7-methoxyquinazolin-6-yloxy)-N-hydroxyheptanamide (CUDC-101) exhibits specific anticancer effects on APL and ATO-resistant APL in vitro and in vivo, while showing negligible cytotoxic effect on the noncancerous cells including normal CD34 cells and bone marrow mesenchymal stem cells from APL patients. Further mechanistic studies show that CUDC-101 triggers caspase-dependent degradation of the promyelocytic leukemia-retinoic acid receptor alpha fusion protein. As a result, APL and ATO-resistant APL cells undergo apoptosis upon CUDC-101 treatment and this apoptosis-inducing effect is even stronger than that of ATO. Finally, using a xenograft mouse model, we demonstrated that CUDC-101 significantly represses leukemia development in vivo. In conclusion, these results suggested that CUDC-101 can serve as a potential candidate drug for APL, particularly for ATO-resistant APL.


Arsenic Trioxide/pharmacology , Caspases/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Hydroxamic Acids/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Quinazolines/pharmacology , Retinoic Acid Receptor alpha/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor , Cell Proliferation , Female , Humans , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
J Cell Biochem ; 120(10): 17791-17810, 2019 10.
Article En | MEDLINE | ID: mdl-31264739

Chemoresistance often causes treatment failure of B-cell acute lymphoblastic leukemia (B-ALL). However, the mechanism remains unclear at present. Herein, overexpression of heme oxygenase-1 (HO-1) was found in the bone marrow stromal cells (BMSCs) from B-ALL patients developing resistance to vincristine (VCR), a chemotherapeutic agent. Two B-ALL cell lines Super B15 and CCRF-SB were cocultured with BMSCs transfected with lentivirus to regulate the expression of HO-1. Silencing HO-1 expression in BMSCs increased the apoptotic rates of B-ALL cell lines induced by VCR, whereas upregulating HO-1 expression reduced the rate. Cell cycle can be arrested in the G2/M phase by VCR. In contrast, B-ALL cells were arrested in the G0/G1 phase due to HO-1 overexpression in BMSCs, which avoided damage from the G2/M phase. Vascular endothelial growth factor (VEGF) in BMSCs, as a key factor in the microenvironment-associated chemoresistance, was also positively coexpressed with HO-1. VEGF secretion was markedly increased in BMSCs with HO-1 upregulation but decreased in BMSCs with HO-1 silencing. B-ALL cell lines became resistant to VCR when cultured with VEGF recombinant protein, so VEGF secretion induced by HO-1 expression may promote the VCR resistance of B-ALL cells. As to the molecular mechanism, the PI3K/AKT pathway mediated regulation of VEGF by HO-1. In conclusion, this study clarifies a mechanism by which B-ALL is induced to resist VCR through HO-1 overexpression in BMSCs, and provides a novel strategy for overcoming VCR resistance in clinical practice.


Drug Resistance, Neoplasm , Heme Oxygenase-1/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism , Vincristine/pharmacology , Adolescent , Adult , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Child, Preschool , Drug Resistance, Neoplasm/drug effects , Humans , Inhibitory Concentration 50 , Lentivirus/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred NOD , Mice, SCID , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Up-Regulation/drug effects , Young Adult
11.
J Cell Physiol ; 234(4): 5252-5263, 2019 04.
Article En | MEDLINE | ID: mdl-30256411

Resistance towards imatinib (IM) remains troublesome in treating many chronic myeloid leukemia (CML) patients. Heme oxygenase-1 (HO-1) is a key enzyme of antioxidative metabolism in association with cell resistance to apoptosis. Our previous studies have shown that overexpression of HO-1 resulted in resistance development to IM in CML cells, while the mechanism remains unclear. In the current study, the IM-resistant CML cells K562R indicated upregulation of some of the histone deacetylases (HDACs) compared with K562 cells. Therefore, we herein postulated HO-1 was associated with HDACs. Silencing HO-1 expression in K562R cells inhibited the expression of some HDACs, and the sensitivity to IM was increased. K562 cells transfected with HO-1 resisted IM and underwent obvious some HDACs. These findings related to the inhibitory effects of high HO-1 expression on the reactive oxygen species (ROS) signaling pathway that negatively regulated HDACs. Increased expression of HO-1 activated HDACs by inhibiting ROS production. In summary, HO-1, which is involved in the development of drug resistance in CML cells by regulating the expression of HDACs, is probably a novel target for improving CML therapy.


Antineoplastic Agents/pharmacology , Heme Oxygenase-1/metabolism , Histone Deacetylases/metabolism , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/pharmacology , Adult , Drug Resistance, Neoplasm , Enzyme Activation , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Middle Aged , Reactive Oxygen Species/metabolism , Signal Transduction , Young Adult
12.
Life Sci ; 211: 224-237, 2018 Oct 15.
Article En | MEDLINE | ID: mdl-30248347

AIMS: The aim of this study was to investigate the combinatorial effects of IM and a novel HDAC inhibitor AR-42 on the proliferation, apoptosis, cell cycle arrest, migration and invasion of CML cells, and to explore the underlying mechanisms. MAIN METHODS: We assessed the ability of the pan-HDAC inhibitor AR-42 and IM, to synergistically kill CML cells by survival, apoptosis, cell cycle, migration and invasion assays in vitro. We also assessed the HDAC1 expression by Western blot and real-time PCR. Synergy was calculated using combinatorial indices as determined by CalcuSyn. KEY FINDINGS: We found that Combining AR-42 with IM synergistically inhibited CML cell proliferation, enhanced cell apoptosis, induced cell cycle arrest, and decreased migration and invasion. The expression of HDAC1 in K562R cells was higher than that in K562 cells. AR-42 enhanced IM-induced HDAC1 expression inhibition in K562 and K562R cells. Importantly, HDAC1 overexpression partly reversed the apoptosis, G2/M phase arrest, migration and invasion of K562 cells induced by the combination of IM with AR-42. Moreover, HDAC1 knockdown partly promoted K562R cell apoptosis and G2/M phase arrest, migration and invasion induced by IM in combination with AR-42. SIGNIFICANCE: In conclusion, AR-42 may increase the sensitivity of CML cells to IM and reverse IM resistance by regulating HDAC1 expression. This study provides new insights into the effects of combined therapy using IM and pan-HDAC inhibitor AR-42, paving the way for overcoming IM resistance in clinical practice.


Apoptosis/drug effects , Drug Synergism , Gene Expression Regulation, Enzymologic/drug effects , Histone Deacetylase 1/antagonists & inhibitors , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Phenylbutyrates/pharmacology , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Drug Combinations , Histone Deacetylase Inhibitors/pharmacology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Tumor Cells, Cultured
13.
Biomed Pharmacother ; 100: 509-520, 2018 Apr.
Article En | MEDLINE | ID: mdl-29494986

To improve the treatment outcomes of acute myeloid leukemia (AML), epigenetic modification has been widely tested and used in recent years. However, drug-resistance is still a choke point to cure the malignancy. The growth factor independent 1 transcriptional repressor (GFI-1), as a zinc-finger transcriptional repressor, can bind histone deacetylases to allow the transcriptional repression. According to the finding of our study, AML patients with low level of GFI-1 not only implicated poor prognosis but also caused Panobinostat-resistance. In our prevent study revealed that heme oxygenase-1(HO-1) was one of the main factors leading to chemotherapy sensitivity to AML. Thus, this study tried to test the correlation between GFI-1 and HO-1. Our study discovered that AML patients with lower expression of GFI-1 had higher level of HO-1, HDAC1, HDAC2 and HDAC3, which resulted in poor prognosis in AML. The results of the in vitro study were the same. Panobinostat is a promising new class of anti-cancer drugs in AML. However, knocking down GFI-1 by siRNA could eliminate the Panobinostat-induced cell apoptosis. Subsequently, we utilized ZnPP to down regulate the level of HO-1, finding that the Panobinostat-resistance between the low level of GFI-1 and empty vector had eased. After further exploring the mechanism, it could be found that with knock down GFI-1, the phosphorylation of Akt and PI3K could be activated. Subsequently, Akt pathway and HO-1 inhibitor were utilized respectively and the resistance was reversed. It suggested that the resistance of Panobinostat to AML cells at low level of GFI-1 was mainly due to up-regulated level of HO-1 through the PI3K-Akt pathway.


Antineoplastic Agents/pharmacology , DNA-Binding Proteins/biosynthesis , Drug Resistance, Neoplasm/drug effects , Heme Oxygenase-1/biosynthesis , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Leukemia, Myeloid, Acute/metabolism , Transcription Factors/biosynthesis , Adolescent , Adult , Aged , Antineoplastic Agents/therapeutic use , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/physiology , Female , Gene Expression Regulation, Neoplastic , HL-60 Cells , Heme Oxygenase-1/genetics , Humans , Hydroxamic Acids/therapeutic use , Indoles/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Panobinostat , Transcription Factors/genetics , Young Adult
...