Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Bioengineered ; 13(3): 5700-5708, 2022 03.
Article in English | MEDLINE | ID: mdl-35212611

ABSTRACT

To investigate the effects of myeloid ecotropic viral integration site-1 (MEIS1) on the proliferation and apoptosis of acute myeloid leukemia (AML) cells and the anticancer effects of the drug, we screened Kasumi-6, KG-1, and Kasumi-1 cells using quantitative reverse transcription polymerase chain reaction. Kasumi-6 and Kasumi-1 cells were subjected to human antigen R (HuR)-mediated interference (IV). Hexokinase 2 (HK2) expression and phosphorylation of protein kinase B (p-AKT) and mammalian target of rapamycin (p-mTOR) were observed with Western blotting. Cell proliferation was assessed using Cell Counting Kit-8, apoptosis was examined using Hoechst 33,258 staining, and glucose uptake was detected with a colorimetric biochemical assay kit. We found that, among the three cell lines tested, MEIS1 expression was highest in Kasumi-1 cells, which were therefore selected for subsequent experiments. Kasumi-1 cells receiving IV showed significantly decreased proliferation (p < 0.05) and increased apoptosis compared to the control group. Compared with the controls, IV significantly increased the expression of HK2, p-AKT, p-mTOR, multidrug resistance-associated protein 1 and P-glycoprotein (P < 0.05), but decreased glucose uptake. Treatment with adriamycin, daunorubicin and imatinib resulted in a progressive increase in inhibition of cell proliferation, with the IV group showing the highest inhibition rate among the three groups (P < 0.05). Thus, inhibition of MEIS1 activity promoted apoptosis, inhibited the proliferation of Kasumi-1 and Kasumi-6 cells, and increaseed the anticancer effect of the drugs, suggesting that inhibition of MEIS1 may be a potential strategy for the treatment of AML.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Myeloid Ecotropic Viral Integration Site 1 Protein , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Glucose/pharmacology , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/antagonists & inhibitors , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases
2.
J Med Chem ; 64(19): 14448-14464, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34542289

ABSTRACT

The pivotal role of the myeloid ecotropic viral integration site 1 (MEIS1) transcriptional factor was reported in cardiac regeneration and hematopoietic stem-cell (HSC) regulation with our previous findings. MEIS1 as a promising target in the context of pharmacological inhibition, we identified a potent myeloid ecotropic viral integration site (MEIS) inhibitor, MEISi-1, to induce murine and human HSC expansion ex vivo and in vivo. In this work, we performed lead optimization on MEISi-1 by synthesizing 45 novel analogues. Structure-activity relationship studies revealed the significance of a para-methoxy group on ring A and a hydrophobic moiety at the meta position of ring B. Obtained biological data were supported by inhibitor docking and molecular dynamics simulation studies. Eleven compounds were depicted as potent inhibitors demonstrating a better inhibitory profile on MEIS1 and target genes Meis1, Hif-1α, and p21. Among those, 4h, 4f, and 4b were the most potent inhibitors. The predicted pharmacokinetics properties fulfill drug-likeness criteria. In addition, compounds exerted neither cytotoxicity on human dermal fibroblasts nor mutagenicity.


Subject(s)
Antiviral Agents/pharmacology , Myeloid Ecotropic Viral Integration Site 1 Protein/antagonists & inhibitors , Animals , Antiviral Agents/chemistry , Humans , Luminescence , Molecular Docking Simulation , Molecular Dynamics Simulation , Structure-Activity Relationship
3.
Int J Mol Med ; 48(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34318904

ABSTRACT

Meis homeobox 1 (Meis1) was initially discovered in 1995 as a factor involved in leukemia in an animal model. Subsequently, 2 years later, MEIS1, the human homolog, was cloned in the liver and cerebellum, and was found to be highly expressed in myeloid leukemia cells. The MEIS1 gene, located on chromosome 2p14, encodes a 390­amino acid protein with six domains. The expression of homeobox protein MEIS1 is affected by cell type, age and environmental conditions, as well as the pathological state. Certain types of modifications of MEIS1 and its protein interaction with homeobox or pre­B­cell leukemia homeobox proteins have been described. As a transcription factor, MEIS1 protein is involved in cell proliferation in leukemia and some solid tumors. The present review article discusses the molecular biology, modifications, protein­protein interactions, as well as the role of MEIS1 in cell proliferation of cancer cells and MEIS1 inhibitors. It is suggested by the available literature MEIS1 has potential to become a cancer therapeutic target.


Subject(s)
Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Discovery , Gene Expression Regulation, Neoplastic/drug effects , Humans , Molecular Targeted Therapy , Myeloid Ecotropic Viral Integration Site 1 Protein/analysis , Myeloid Ecotropic Viral Integration Site 1 Protein/antagonists & inhibitors , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Protein Interaction Maps/drug effects
4.
Sci Rep ; 10(1): 7994, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409701

ABSTRACT

Meis1, which belongs to TALE-type class of homeobox gene family, appeared as one of the key regulators of hematopoietic stem cell (HSC) self-renewal and a potential therapeutical target. However, small molecule inhibitors of MEIS1 remained unknown. This led us to develop inhibitors of MEIS1 that could modulate HSC activity. To this end, we have established a library of relevant homeobox family inhibitors and developed a high-throughput in silico screening strategy against homeodomain of MEIS proteins using the AutoDock Vina and PaDEL-ADV platform. We have screened over a million druggable small molecules in silico and selected putative MEIS inhibitors (MEISi) with no predicted cytotoxicity or cardiotoxicity. This was followed by in vitro validation of putative MEIS inhibitors using MEIS dependent luciferase reporter assays and analysis in the ex vivo HSC assays. We have shown that small molecules named MEISi-1 and MEISi-2 significantly inhibit MEIS-luciferase reporters in vitro and induce murine (LSKCD34l°w cells) and human (CD34+, CD133+, and ALDHhi cells) HSC self-renewal ex vivo. In addition, inhibition of MEIS proteins results in downregulation of Meis1 and MEIS1 target gene expression including Hif-1α, Hif-2α and HSC quiescence modulators. MEIS inhibitors are effective in vivo as evident by induced HSC content in the murine bone marrow and downregulation of expression of MEIS target genes. These studies warrant identification of first-in-class MEIS inhibitors as potential pharmaceuticals to be utilized in modulation of HSC activity and bone marrow transplantation studies.


Subject(s)
Drug Development , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/antagonists & inhibitors , Amino Acid Sequence , Animals , Biomarkers , Bone Marrow Cells , Cell Proliferation , Drug Evaluation, Preclinical , Flow Cytometry , Genes, Reporter , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Models, Molecular , Myeloid Ecotropic Viral Integration Site 1 Protein/chemistry , Protein Conformation , Small Molecule Libraries , Structure-Activity Relationship
5.
J Mol Endocrinol ; 64(4): 249-258, 2020 05.
Article in English | MEDLINE | ID: mdl-32197234

ABSTRACT

Decidualization is a critical process for embryo implantation and pregnancy maintenance in humans. The homeobox gene HOXA10 has been widely studied in endometrial receptivity establishment and decidualization. MEIS1, a three-amino-acid loop extension (TALE) family homeobox gene, has been proven to be a co-factor for HOXA10 in mouse uterus. However, the interaction between MEIS1 and HOXA10 in the human decidual cells remains to be elucidated. siRNA and CRISPR-Cas9 were employed to knockdown and knockout MEIS1 in the cultured human endometrial stromal cells, and it was found that MEIS1 deficiency leads to impaired decidualization. The physical interaction between the MEIS1 and HOXA10 in human endometrial stromal cell was confirmed by immunoprecipitation. Moreover, KAT2B and ETA were proved to be downregulated in the absence of MEIS1, and luciferase reporter and ChIP assays demonstrated that MEIS1-HOXA10 complex binds to the promoters of KAT2B and ETA and regulates their activity. Overexpression of KAT2B and ETA can partially rescue the decidualization defects in MEIS1-knockout HESCs. Taken together, these data suggest that MEIS1 plays an indispensable role in decidualization in human endometrial stromal cells, and MEIS1 interacts with HOXA10 to regulate the downstream genes, such as KAT2B and ETA. These findings will contribute to our understanding about the regulatory network in the process of decidualization in humans.


Subject(s)
Decidua/physiology , Endometrium/physiology , Myeloid Ecotropic Viral Integration Site 1 Protein/physiology , CRISPR-Cas Systems/genetics , Cells, Cultured , Decidua/metabolism , Embryo Implantation/genetics , Endometrium/cytology , Female , Gene Knockout Techniques , Gene Regulatory Networks/physiology , HEK293 Cells , Homeobox A10 Proteins/metabolism , Humans , Myeloid Ecotropic Viral Integration Site 1 Protein/antagonists & inhibitors , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Protein Binding , RNA, Small Interfering/pharmacology , Stromal Cells/physiology
6.
FASEB J ; 33(6): 7417-7426, 2019 06.
Article in English | MEDLINE | ID: mdl-30884246

ABSTRACT

Fetal cardiomyocytes shift from glycolysis to oxidative phosphorylation around the time of birth. Myeloid ecotropic viral integration site 1 (MEIS1) is a transcription factor that promotes glycolysis in hematopoietic stem cells. We reasoned that MEIS1 could have a similar role in the developing heart. We hypothesized that suppression of MEIS1 expression in fetal sheep cardiomyocytes leads to a metabolic switch as found at birth. Expression of MEIS1 was assayed in left ventricular cardiac tissue and primary cultures of cardiomyocytes from fetal (100- and 135-d gestation, term = 145 d), neonatal, and adult sheep. Cultured cells were treated with short interfering RNA (siRNA) to suppress MEIS1. Oxygen consumption rate was assessed with the Seahorse metabolic flux analyzer, and mitochondrial activity was assessed by staining cells with MitoTracker Orange. Cardiomyocyte respiratory capacity increased with advancing age concurrently with decreased expression of MEIS1. MEIS1 suppression with siRNA increased maximal oxygen consumption in fetal cells but not in postnatal cells. Mitochondrial activity was increased and expression of glycolytic genes decreased when MEIS1 expression was suppressed. Thus, we conclude that MEIS1 is a key regulator of cardiomyocyte metabolism and that the normal down-regulation of MEIS1 with age underlies a gradual switch to oxidative metabolism.-Lindgren, I. M., Drake, R. R., Chattergoon, N. N., Thornburg, K. L. Down-regulation of MEIS1 promotes the maturation of oxidative phosphorylation in perinatal cardiomyocytes.


Subject(s)
Aging/metabolism , Fetal Heart/cytology , Gene Expression Regulation, Developmental , Mitochondria, Heart/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/physiology , Myocytes, Cardiac/metabolism , Oxidative Phosphorylation , Adenosine Triphosphate/biosynthesis , Aging/genetics , Animals , Cells, Cultured , Female , Fetal Heart/metabolism , Gestational Age , Glycolysis , Heart/growth & development , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein/antagonists & inhibitors , Myeloid Ecotropic Viral Integration Site 1 Protein/biosynthesis , Myeloid Ecotropic Viral Integration Site 1 Protein/genetics , Myocardium/cytology , Oxygen/blood , Oxygen Consumption , Partial Pressure , Pregnancy , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...