Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cells ; 11(15)2022 08 08.
Article in English | MEDLINE | ID: mdl-35954304

ABSTRACT

The prevention of metastasis is a central goal of cancer therapy. Caveolin-1 (Cav-1) is a structural membrane and scaffolding protein shown to be a key regulator of late-stage breast cancer metastasis. However, therapeutic strategies targeting Cav-1 are still lacking. Here, we demonstrate that the pharmacological activation of potassium channel Kv11.1, which is uniquely expressed in MDA-MB-231 triple negative breast cancer cells (TNBCs) but not in normal MCF-10A cells, induces the dephosphorylation of Cav-1 Tyr-14 by promoting the Ca2+-dependent stimulation of protein tyrosine phosphatase 1B (PTP1B). Consequently, the dephosphorylation of Cav-1 resulted in its disassociation from ß-catenin, which enabled the accumulation of ß-catenin at cell borders, where it facilitated the formation of cell-cell adhesion complexes via interactions with R-cadherin and desmosomal proteins. Kv11.1 activation-dependent Cav-1 dephosphorylation induced with NS1643 also reduced cell migration and invasion, consistent with its ability to regulate focal adhesion dynamics. Thus, this study sheds light on a novel pharmacological mechanism of promoting Cav-1 dephosphorylation, which may prove to be effective at reducing metastasis and promoting contact inhibition.


Subject(s)
Caveolin 1 , Triple Negative Breast Neoplasms , Caveolin 1/metabolism , Cell Movement , Cresols , Humans , Phenylurea Compounds , Potassium Channels , Triple Negative Breast Neoplasms/drug therapy , beta Catenin/metabolism
2.
Cancers (Basel) ; 13(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34885136

ABSTRACT

Control of ionic gradients is critical to maintain cellular homeostasis in both physiological and pathological conditions, but the role of ion channels in cancer cells has not been studied thoroughly. In this work we demonstrated that activity of the Kv11.1 potassium channel plays a vital role in controlling the migration of colon cancer cells by reversing the epithelial-to-mesenchymal transition (EMT) into the mesenchymal-to-epithelial transition (MET). We discovered that pharmacological stimulation of the Kv11.1 channel with the activator molecule NS1643 produces a strong inhibition of colon cancer cell motility. In agreement with the reversal of EMT, NS1643 treatment leads to a depletion of mesenchymal markers such as SNAIL1, SLUG, TWIST, ZEB, N-cadherin, and c-Myc, while the epithelial marker E-cadherin was strongly upregulated. Investigating the mechanism linking Kv11.1 activity to reversal of EMT into MET revealed that stimulation of Kv11.1 produced a strong and fast inhibition of the TGFß signaling. Application of NS1643 resulted in de-phosphorylation of the TGFß downstream effectors R-SMADs by activation of the serine/threonine phosphatase PP2B (calcineurin). Consistent with the role of TGFß in controlling cancer stemness, NS1643 also produced a strong inhibition of NANOG, SOX2, and OCT4 while arresting the cell cycle in G0/G1. Our data demonstrate that activation of the Kv11.1 channel reprograms EMT into MET by inhibiting TGFß signaling, which results in inhibition of motility in colon cancer cells.

3.
Redox Biol ; 45: 102030, 2021 09.
Article in English | MEDLINE | ID: mdl-34147842

ABSTRACT

Potassium channels are important regulators of cellular homeostasis and targeting these proteins pharmacologically is unveiling important mechanisms in cancer cell biology. Here we demonstrate that pharmacological stimulation of the Kv11.1 potassium channel activity results in mitochondrial reactive oxygen species (ROS) production and fragmentation in breast cancer cell lines and patient-derived organoids independent of breast cancer subtype. mRNA expression profiling revealed that Kv11.1 activity significantly altered expression of genes controlling the production of ROS and endoplasmic-reticulum (ER) stress. Characterization of the transcriptional signature of breast cancer cells treated with Kv11.1 potassium channel activators strikingly revealed an adaptive response to the potentially lethal augmentation of ROS by increasing Nrf2-dependent transcription of antioxidant genes. Nrf2 in this context was shown to promote survival in breast cancer, whereas knockdown of Nrf2 lead to Kv11.1-induced cell death. In conclusion, we found that the Kv11.1 channel activity promotes oxidative stress in breast cancer cells and that suppression of the Nrf2-mediated anti-oxidant survival mechanism strongly sensitized breast cancer cells to a lethal effect of pharmacological activation of Kv11.1.


Subject(s)
Antioxidants , Breast Neoplasms , Antioxidants/pharmacology , Breast Neoplasms/genetics , Endoplasmic Reticulum Stress , Female , Humans , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species
4.
Front Pharmacol ; 11: 577, 2020.
Article in English | MEDLINE | ID: mdl-32457608

ABSTRACT

Gynecologic cancers are among the most lethal cancers found in women, and, advanced stage cancers are still a treatment challenge. Ion channels are known to contribute to cellular homeostasis in all cells and mounting evidence indicates that ion channels could be considered potential therapeutic targets against cancer. Nevertheless, the pharmacologic effect of targeting ion channels in cancer is still understudied. We found that the expression of Kir6.2/SUR2 potassium channel is a potential favorable prognostic factor in gynecologic cancers. Also, pharmacological stimulation of the Kir6.2/SUR2 channel activity with the selective activator molecule minoxidil arrests tumor growth in a xenograft model of ovarian cancer. Investigation on the mechanism linking the Kir6.2/SUR2 to tumor growth revealed that minoxidil alters the metabolic and oxidative state of cancer cells by producing mitochondrial disruption and extensive DNA damage. Consequently, application of minoxidil results in activation of a caspase-3 independent cell death pathway. Our data show that repurposing of FDA approved K+ channel activators may represent a novel, safe adjuvant therapeutic approach to traditional chemotherapy for the treatment of gynecologic cancers.

5.
Biol Blood Marrow Transplant ; 26(7): 1273-1279, 2020 07.
Article in English | MEDLINE | ID: mdl-32194286

ABSTRACT

High-dose melphalan (MEL) and autologous stem cell transplantation (ASCT) is the standard of care in the treatment of multiple myeloma (MM). Resistance to MEL has been linked to increased DNA repair. Here we sought to identify whether inhibition of poly(ADP-ribose) polymerase (PARP) synergizes with MEL and can overcome resistance. We tested the synergistic cytotoxicity of 3 inhibitors of PARP (PARPi)-veliparib (VEL), olaparib (OLA), and niraparib (NIRA)-combined with MEL in RPMI8226 and U266 MM cell lines, as well as in their MEL resistance counterparts, RPMI8226-LR5 (LR5) and U266-LR6 (LR6). The addition of VEL, OLA, and NIRA to MEL reduced the half maximal inhibitory concentration (IC50) in RPMI8226 cells from 27.8 µM to 23.1 µM, 22.5 µM, and 18.0 µM, respectively. Similarly, the IC50 of MEL in U266 cells was decreased from 6.2 µM to 3.2 µM, 3.3 µM, and 3.0 µM, respectively. In LR5 and LR6 cells, PARPi did not reverse MEL resistance. We confirmed this in a NOD/SCID/gamma null xenograft mouse model with either MEL-sensitive (RPMI8226) or MEL-resistant (LR5) MM. Treatment with a MEL-VEL combination prolonged survival compared with MEL alone in RPMI8226 mice (107 days versus 67.5 days; P = .0009), but not in LR5 mice (41 versus 39 days; P = .09). We next tested whether 2 double-stranded DNA repair mechanisms, homologous recombination (HR) and nonhomologous end-joining (NHEJ), cause MEL resistance in LR5 and LR6 cells. In an HR assay, LR6 cells had a 4.5-fold greater HR capability than parent U226 cells (P = .05); however, LR5 cells had an equivalent HR ability as parent RPMI8226 cells. We hypothesized that NHEJ may be a mediator of MEL resistance in LR5 cells. Given that DNA-PK is integral to NHEJ and may be a therapeutic target, we treated LR5 cells with the DNA-PK inhibitor NU7026 in combination with MEL. Although NU7026 alone at 2.5 µM had no cytotoxicity, in combination it completely reversed resistance to MEL (MEL IC50, 46.4 µM versus 14.4 µM). We examined the clinical implications of our findings in a dataset of 414 patients treated with tandem ASCT. High PARP1 expressers had lower survival compared with patients with low expression (median 42.7 months versus median not reached; P = .003). We hypothesized that combined expression of the HR gene BRCA1, the NHEJ gene PRKDC (DNA-PK), and PARP1 may predict survival and found that overexpression of 0 (n = 101), 1 or 2 (n = 287), or all 3 (n = 26) genes had a negative impact on median survival (undefined versus 57.8 months versus 14.8 months; P < .0001). Here we demonstrate that PARPi synergized with MEL, but that resistance (which may be due to HR and NHEJ pathways) is not completely reversed by PARPi. In addition, we observed that a 3-gene analysis may be tested to identify patients resistant or sensitive to high-dose MEL.


Subject(s)
Hematopoietic Stem Cell Transplantation , Melphalan , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Melphalan/pharmacology , Mice , Mice, Inbred NOD , Mice, SCID , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Transplantation, Autologous
6.
J Immunol ; 202(8): 2493-2501, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30842275

ABSTRACT

The limited number of hematopoietic stem cells (HSC) within a single unit of human cord blood currently limits its use as an alternate graft source. However, we have developed a strategy using 5-aza-2'-deoxycytidine (5azaD) and trichostatin A (TSA), which expands transplantable HSC 7- to 10-fold. In our current studies, we have assessed the allostimulatory capacity of the 5azaD/TSA-expanded grafts. The coexpression of immunophenotypic dendritic cell (DC) markers, such as HLA-DR/CD86 and HLA-DR/CD11c as determined by flow cytometry, and the allostimulatory capacity of 5azaD/TSA-expanded cells as determined by MLC were both significantly lower than control. It has been previously demonstrated that STAT3 is indispensable for the differentiation of DC from HSC. Real-time quantitative PCR analysis revealed that 5azaD/TSA-expanded cells expressed more STAT3 transcript than control while also expressing increased transcripts for STAT3 inhibitors including SHP1, p21, and GATA1. Western blot analysis indicates that chromatin-modifying agent-expanded grafts displayed a reduced ratio of p-STAT3 to total STAT3 than control cultures, which is likely indicative of STAT3 inactivity in 5azD/TSA-expanded grafts. Culturing 5azaD/TSA-expanded cord blood cells in extended cultures reveals that they are still capable of generating DC. Notably, STAT3 inactivity was transient because the transcript levels of STAT3 and its inhibitors, including SHP1, were comparable between 5azaD/TSA and control cultures following extended culture. Taken together, our studies indicate that the reduced allostimulatory capacity of 5azaD/TSA-expanded cells is likely because of reversible inhibition of STAT3-dependent DC differentiation. These results suggest that a graft composed of 5azaD/TSA-expanded cells possesses relatively less allostimulatory response but is still capable of generating DC in permissive conditions.


Subject(s)
Antigens, Differentiation/immunology , Chromatin/immunology , Decitabine/pharmacology , Fetal Blood/immunology , Hematopoietic Stem Cells/immunology , Hydroxamic Acids/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Hematopoietic Stem Cells/cytology , Humans
7.
Biol Blood Marrow Transplant ; 25(5): 855-860, 2019 05.
Article in English | MEDLINE | ID: mdl-30615982

ABSTRACT

Patients with high-risk myeloproliferative neoplasms (MPNs), and in particular myelofibrosis (MF), can be cured only with allogeneic hematopoietic stem cell transplantation (HSCT). Because MPNs and JAK2V617F-mutated cells show genomic instability, stalled replication forks, and baseline DNA double-strand breaks, DNA repair inhibition with poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors represents a potential novel therapy. Because the alkylating agent busulfan is integral in conditioning regimens for HSCT and leads to stalled replication forks through DNA strand cross-linking, we hypothesized that PARP inhibition with veliparib in combination with busulfan may lead to synergistic cytotoxicity in MPN cells. We first treated 2 MPN cell lines harboring the JAK2V617F mutation (SET2 and HEL) with veliparib at increasing concentrations and measured cell proliferation. SET2 and HEL cells were relatively sensitive to veliparib (IC50 of 11.3 µM and 74.2 µM, respectively). We next treated cells with increasing doses of busulfan in combination with 4 µM veliparib and found that the busulfan IC50 decreased from 27 µM to 4 µM in SET2 cells and from 45.1 µM to 28.1 µM in HEL cells. The mean combination index was .55 for SET2 cells and .40 for HEL cells. Combination treatment of SET2 cells caused G2M arrest in 53% of cells, compared with 30% with veliparib alone and 35% with busulfan alone. G2M arrest was associated with activation of the ATR-Chk1 pathway, as shown by an immunofluorescence assay for phosphorylated Chk1 (p-Chk1). We then tested in vivo the effect of combined low doses of busulfan and veliparib in a JAK2V617F MPN-AML xenotransplant model. Vehicle- and veliparib-treated mice had similar median survival of 39 and 40 days, respectively. Combination treatment increased median survival from 47 days (busulfan alone) to 50 days (P = .02). Finally, we tested the combined effect of busulfan and veliparib on CD34+ cells obtained from the bone marrow or peripheral blood of 5 patients with JAK2V617F-mutated and 2 patients with CALR-mutated MF. MF cells treated with the combination of veliparib and busulfan showed reduced colony formation compared with busulfan alone (87% versus 68%; P = .001). In contrast, treatment of normal CD34+ cells with veliparib did not affect colony growth. Here we show that in vivo confirmation that treatment with the PARP-1 inhibitor veliparib and busulfan results in synergistic cytotoxicity in MPN cells. Our data provide the rationale for testing novel pretransplantation conditioning regimens with combinations of PARP-1 inhibition and reduced doses of alkylators, such as busulfan and melphalan, for high-risk MPNs or MPN-derived acute myelogenous leukemia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/pharmacology , Busulfan/pharmacology , Myeloproliferative Disorders/drug therapy , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/therapeutic use , Busulfan/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Heterografts , Humans , Mice , Myeloproliferative Disorders/pathology , Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
8.
Sci Rep ; 8(1): 6519, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29695725

ABSTRACT

MicroRNAs (miRNAs) are emerging as critical regulators of normal and malignant hematopoiesis. In previous studies of acute myeloid leukemia miR-9 overexpression was commonly observed. Here, we show that ectopic expression of miR-9 in vitro and in vivo significantly blocks differentiation of erythroid progenitor cells with an increase in reactive oxygen species (ROS) production. Consistent with this observation, ROS scavenging enzymes, including superoxide dismutase (Sod2), Catalase (Cat), and glutathine peroxidase (Gpx1), are down-regulated by miR-9. In addition, miR-9 suppresses expression of the erythroid transcriptional regulator FoxO3, and its down-stream targets Btg1 and Cited 2 in erythroid progenitor cells, while expression of a constitutively active form of FoxO3 (FoxO3-3A) reverses miR-9-induced suppression of erythroid differentiation, and inhibits miR-9-induced ROS production. Thus, our findings indicate that aberrant expression of miR-9 blocks erythropoiesis by deregulating FoxO3-mediated pathways, which may contribute to the ineffective erythropoiesis observed in patients with hematological malignancies.


Subject(s)
Erythropoiesis/genetics , Forkhead Box Protein O3/genetics , MicroRNAs/genetics , Up-Regulation/genetics , Animals , Catalase/genetics , Cell Differentiation/genetics , Cell Line , Down-Regulation/genetics , Erythroid Cells/metabolism , Erythroid Precursor Cells/metabolism , Gene Expression Regulation/genetics , Glutathione Peroxidase/genetics , HEK293 Cells , Humans , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Glutathione Peroxidase GPX1
9.
Front Immunol ; 9: 3186, 2018.
Article in English | MEDLINE | ID: mdl-30723481

ABSTRACT

Early release of TNFα after hematopoietic stem cell transplantation (HSCT) correlates with development of acute graft-vs.-host disease (GVHD). Here we tested the effect of TNFα and alloreactive T cells on early hematopoietic HSC genotype and function. Addition of TNFα (10 ng/ml) in liquid cultures with CD34+ cells for 6-72 h resulted in the downregulation of genes associated with stem cell activity, such as DNMT3A, DNMT3B, TET1, TET2, SOX2, NANOG, and OCT4, whereas no significant effect was observed on DNMT1 and GATA2 expression. These findings were reversed by using an anti-TNFα antibody. Similar gene downregulation was observed when CD34+ cells were co-cultured with alloreactive T cells CD34+ cells for 48-72 h, and this effect was partially prevented by rapamycin and an anti-TNFα antibody. CD34+ cells pre-incubated with TNFα for 48 h and transplanted in irradiated NOD-SCID ɤnull (NSG) mice showed a reduced myeloid engraftment compared to control mice. By using a xenograft model recently developed in our lab, we co-transplanted CD34+ cells and allogeneic T lymphocytes at 1:0.1 ratio in one group that also received etanercept (TNFα inhibitor) at 100 µg intra-peritoneum (i.p.) on days -1,+1,+3,+5 post-HSCT, and in the control group. At 6 weeks post-transplant, mice that received etanercept had a significantly higher number of marrow huCD45+CD34+CD38- early stem cells (p = 0.03) and a reduced number of huCD45+CD3+ splenic T cells (p = 0.04) compared to controls. The repopulating activity of marrow cells from mice treated with etanercept vs. controls was tested in secondary transplants. Although the overall engraftment was similar in the two groups, CD34+ cells isolated from recipients of marrow from the etanercept group showed a significantly greater expression of stem cell-associated genes and a higher number of CD45+CD34+CD38- cells than in controls (p = 0.03). Our findings suggest that early TNFα increase post-transplant can affect long-term stem cell engraftment, and that blockade of TNFα early after transplant may limit a cytokine-mediated suppressive effect on repopulating stem cell function.


Subject(s)
Cell Differentiation , Cell Self Renewal , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Animals, Genetically Modified , Cell Communication/genetics , Cell Communication/immunology , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Cytokines/metabolism , Gene Expression Profiling , Graft Survival/immunology , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/drug effects , Humans , Immunosuppressive Agents/pharmacology , Mice , Models, Animal , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/antagonists & inhibitors
10.
Biol Blood Marrow Transplant ; 23(12): 2048-2056, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28818684

ABSTRACT

A xenograft model of stem cell rejection was developed by co-transplantating human CD34+ and allogeneic CD3+ T cells into NOD-scid ɣ-chainnull mice. T cells caused graft failure when transplanted at any CD34/CD3 ratio between 1:50 and 1:.1. Kinetics experiments showed that 2 weeks after transplantation CD34+ cells engrafted the marrow and T cells expanded in the spleen. Then, at 4 weeks only memory T cells populated both sites and rejected CD34+ cells. Blockade of T cell costimulation was tested by injecting the mice with abatacept (CTLA4-IgG1) from day -1 to +27 (group A), from day -1 to +13 (group B), or from day +14 to +28 (group C). On day +56 groups B and C had rejected the graft, whereas in group A graft failure was completely prevented, although with lower stem cell engraftment than in controls (P = .03). Retransplantation of group A mice with same CD34+ cells obtained a complete reconstitution of human myeloid and B cell lineages and excluded latent alloreactivity. In this first xenograft model of stem cell rejection we showed that transplantation of HLA mismatched CD34+ cells may be facilitated by treatment with abatacept and late stem cell boost.


Subject(s)
Abatacept/pharmacology , Antigens, CD34 , CD3 Complex , Graft Rejection/prevention & control , T-Lymphocytes/transplantation , Abatacept/administration & dosage , Abatacept/therapeutic use , Animals , Graft Rejection/etiology , Heterografts , Humans , Mice, Inbred NOD , Reoperation , T-Lymphocytes/immunology , Time Factors
11.
Proc Natl Acad Sci U S A ; 110(14): 5594-9, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23509296

ABSTRACT

MicroRNA-9 (miR-9) is emerging as a critical regulator of organ development and neurogenesis. It is also deregulated in several types of solid tumors; however, its role in hematopoiesis and leukemogenesis is not yet known. Here we show that miR-9 is detected in hematopoietic stem cells and hematopoietic progenitor cells, and that its expression increases during hematopoietic differentiation. Ectopic expression of miR-9 strongly accelerates terminal myelopoiesis and promotes apoptosis in vitro and in vivo. Conversely, in hematopoietic progenitor cells, the inhibition of miR-9 with a miRNA sponge blocks myelopoiesis. Ecotropic viral integration site 1 (EVI1), required for normal embryogenesis, is considered an oncogene because its inappropriate up-regulation induces malignant transformation in solid and hematopoietic cancers. Here we show that EVI1 binds to the promoter of miR-9-3, leading to DNA hypermethylation of the promoter and repression of miR-9. Moreover, miR-9 expression reverses a myeloid differentiation block that is induced by EVI1. Our findings indicate that EVI1, when inappropriately expressed, delays or blocks myeloid differentiation at least in part by DNA hypermethylation and down-regulation of miR-9. It was reported that Forkhead box class O genes (FoxOs) inhibit myeloid differentiation and prevent differentiation of leukemia-initiating cells. Here we identify both FoxO1 and FoxO3 as direct targets of miR-9 in hematopoietic cells and find that up-regulation of FoxO3 inhibits miR-9-induced myelopoiesis. These results reveal a unique role of miR-9 in myelopoiesis and in the pathogenesis of EVI1-induced myeloid neoplasms and provide insights into the epigenetic regulation of miR9 in tumorigenesis.


Subject(s)
DNA Methylation , DNA-Binding Proteins/metabolism , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/metabolism , MicroRNAs/metabolism , Myelopoiesis/physiology , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Colony-Forming Units Assay , DNA Primers/genetics , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , HEK293 Cells , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , Myelopoiesis/genetics , NIH 3T3 Cells , Proto-Oncogenes , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
12.
J Biol Chem ; 287(27): 22683-90, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22589536

ABSTRACT

The canonical Wnt/ß-catenin signaling is activated during development, tumorigenesis, and in adult homeostasis, yet its role in maintenance of hematopoietic stem/progenitor cells is not firmly established. Here, we demonstrate that conditional expression of an active form of ß-catenin in vivo induces a marked increase in the frequency of apoptosis in hematopoietic stem/progenitor cells (HSCs/HPCs). Activation of Wnt/ß-catenin signaling in HPCs in vitro elevates the activity of caspases 3 and 9 and leads to a loss of mitochondrial membrane potential (ΔΨ(m)), indicating that it induces the intrinsic mitochondrial apoptotic pathway. In vivo, expression of activated ß-catenin in HPCs is associated with down-regulation of Bcl2 and expression of Casp3. Bone marrow transplantation assays reveal that enhanced cell survival by a Bcl2 transgene re-establishes the reconstitution capacity of HSCs/HPCs that express activated ß-catenin. In addition, a Bcl2 transgene prevents exhaustion of these HSCs/HPCs in vivo. Our data suggest that activation of the Wnt/ß-catenin pathway contributes to the defective function of HPCs in part by deregulating their survival.


Subject(s)
Apoptosis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mitochondria/physiology , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Survival/physiology , Cells, Cultured , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , beta Catenin/genetics
13.
PLoS One ; 6(6): e20793, 2011.
Article in English | MEDLINE | ID: mdl-21695170

ABSTRACT

EVI1 has pleiotropic functions during murine embryogenesis and its targeted disruption leads to prenatal death by severely affecting the development of virtually all embryonic organs. However, its functions in adult tissues are still unclear. When inappropriately expressed, EVI1 becomes one of the most aggressive oncogenes associated with human hematopoietic and solid cancers. The mechanisms by which EVI1 transforms normal cells are unknown, but we showed recently that EVI1 indirectly upregulates self-renewal and cell-cycling genes by inappropriate methylation of CpG dinucleotides in the regulatory regions of microRNA-124-3 (miR-124-3), leading to the repression of this small gene that controls normal differentiation and cell cycling of somatic cells. We used the regulatory regions of miR-124-3 as a read-out system to investigate how EVI1 induces de novo methylation of DNA. Here we show that EVI1 physically interacts with DNA methyltransferases 3a and 3b (Dnmt3a/b), which are the only de novo DNA methyltransferases identified to date in mouse and man, and that it forms an enzymatically active protein complex that induces de novo DNA methylation in vitro. This protein complex targets and binds to a precise region of miR-124-3 that is necessary for repression of a reporter gene by EVI1. Based on our findings, we propose that in cooperation with Dnmt3a/b EVI1 regulates the methylation of DNA as a sequence-specific mediator of de novo DNA methylation and that inappropriate EVI1 expression contributes to carcinogenesis through improper DNA methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Catalytic Domain , DNA Methyltransferase 3A , DNA-Binding Proteins/chemistry , Genome/genetics , Humans , Immunoprecipitation , MDS1 and EVI1 Complex Locus Protein , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , NIH 3T3 Cells , Oncogene Proteins/chemistry , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogenes , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/chemistry , Zinc Fingers , DNA Methyltransferase 3B
14.
Blood ; 116(26): 6023-6, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-20861460

ABSTRACT

Recently, Dawson et al identified a previously unrecognized nuclear role of JAK2 in the phosphorylation of histone H3 in hematopoietic cell lines. We searched nuclear JAK2 in total bone marrow (BM) cells and in 4 sorted BM cell populations (CD34(+), CD15(+), CD41(+), and CD71(+)) of 10 myeloproliferative neoplasia (MPN) patients with JAK2V617F mutation and 5 patients with wild-type JAK2 MPN. Confocal immunofluorescent images and Western blot analyses of nuclear and cytoplasmic fractions found nuclear JAK2 in CD34(+) cells of 10 of 10 JAK2-mutated patients but not in patients with wild-type JAK2. JAK2 was predominantly in the cytoplasmic fraction of differentiated granulocytic, megakaryocytic, or erythroid cells obtained from all patients. JAK2V617F up-regulates LMO2 in K562 and in JAK2V617F-positive CD34(+) cells. The selective JAK2 inhibitor AG490 normalizes the LMO2 levels in V617F-positive K562 and restores the cyto-plasmic localization of JAK2.


Subject(s)
Antigens, CD34/metabolism , Cell Nucleus/metabolism , Erythroid Cells/metabolism , Granulocytes/metabolism , Janus Kinase 2/genetics , Megakaryocytes/metabolism , Myeloproliferative Disorders/metabolism , Philadelphia Chromosome , Apoptosis , Blotting, Western , Bone Marrow/metabolism , Cells, Cultured , Cytoplasm/metabolism , Granulocytes/cytology , Humans , Janus Kinase 2/metabolism , K562 Cells , Megakaryocytes/cytology , Microscopy, Fluorescence , Mutation/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phosphorylation , Prognosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
15.
Proc Natl Acad Sci U S A ; 107(21): 9783-8, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20448201

ABSTRACT

By expressing EVI1 in murine bone marrow (BM), we previously described a myelodysplastic syndrome (MDS) model characterized by pancytopenia, dysmegakaryopoiesis, dyserythropoiesis, and BM failure. The mice invariably died 11-14 months after BM transplantation (BMT). Here, we show that a double point mutant EVI1-(1+6Mut), unable to bind Gata1, abrogates the onset of MDS in the mouse and re-establishes normal megakaryopoiesis, erythropoiesis, BM function, and peripheral blood profiles. These normal features were maintained in the reconstituted mice until the study was ended at 21 months after BMT. We also report that EVI1 deregulates several genes that control cell division and cell self-renewal. In striking contrast, these genes are normalized in the presence of the EVI1 mutant. Moreover, EVI1, but not the EVI1 mutant, seemingly deregulates these cellular processes by altering miRNA expression. In particular, the silencing of miRNA-124 by DNA methylation is associated with EVI1 expression, but not that of the EVI1 mutant, and appears to play a key role in the up-regulation of cell division in murine BM cells and in the hematopoietic cell line 32Dcl3. The results presented here demonstrate that EVI1 induces MDS in the mouse through two major pathways, both of which require the interaction of EVI1 with other factors: one, results from EVI1-Gata1 interaction, which deregulates erythropoiesis and leads to fatal anemia, whereas the other occurs by interaction of EVI1 with unidentified factors causing perturbation of the cell cycle and self-renewal, as a consequence of silencing miRNA-124 by EVI1 and, ultimately, ensues in BM failure.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/cytology , MicroRNAs/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , RNA Interference , Transcription Factors/metabolism , Animals , Bone Marrow Transplantation , Cell Differentiation , Cell Proliferation , Cells, Cultured , CpG Islands , DNA Replication , DNA-Binding Proteins/genetics , Gene Expression Regulation , MDS1 and EVI1 Complex Locus Protein , Methylation , Mice , Molecular Sequence Data , Mutation , Myelodysplastic Syndromes/pathology , Promoter Regions, Genetic , Proto-Oncogenes/genetics , Transcription Factors/genetics
16.
Cancer Res ; 69(1): 262-71, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19118011

ABSTRACT

Janus-activated kinase 2 (JAK2) mutations are common in myeloproliferative disorders; however, although they are detected in virtually all polycythemia vera patients, they are found in approximately 50% of essential thrombocythemia (ET) patients, suggesting that converging pathways/abnormalities underlie the onset of ET. Recently, the chromosomal translocation 3;21, leading to the fusion gene AML1/MDS1/EVI1 (AME), was observed in an ET patient. After we forced the expression of AME in the bone marrow (BM) of C57BL/6J mice, all the reconstituted mice died of a disease with symptoms similar to ET with a latency of 8 to 16 months. Peripheral blood smears consistently showed an elevated number of dysplastic platelets with anisocytosis, degranulation, and giant size. Although the AME-positive mice did not harbor Jak2 mutations, the BM of most of them had significantly higher levels of activated Stat3 than the controls. With combined biochemical and biological assays we found that AME binds to the Stat3 promoter leading to its up-regulation. Signal transducers and activators of transcription 3 (STAT3) analysis of a small group of ET patients shows that in about half of the patients, there is STAT3 hyperactivation independently of JAK2 mutations, suggesting that the hyperactivation of STAT3 by JAK2 mutations or promoter activation may be a critical step in development of ET.


Subject(s)
Janus Kinase 2/genetics , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/metabolism , Thrombocythemia, Essential/genetics , Aged , Animals , Blood Platelets/pathology , Bone Marrow Cells/pathology , Core Binding Factor Alpha 2 Subunit/biosynthesis , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Disease Models, Animal , Enzyme Activation , Humans , Janus Kinase 2/metabolism , K562 Cells , Megakaryocytes/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT3 Transcription Factor/genetics , Thrombocythemia, Essential/blood , Thrombocythemia, Essential/enzymology , Thrombocythemia, Essential/metabolism , Up-Regulation
17.
Cancer Res ; 67(12): 5658-66, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575132

ABSTRACT

Recurring chromosomal translocations observed in human leukemia often result in the expression of fusion proteins that are DNA-binding transcription factors. These altered proteins acquire new dimerization properties that result in the assembly of inappropriate multimeric transcription complexes that deregulate hematopoietic programs and induce leukemogenesis. Recently, we reported that the fusion protein AML1/MDS1/EVI1 (AME), a product of a t(3;21)(q26;q22) associated with chronic myelogenous leukemia and acute myelogenous leukemia, displays a complex pattern of self-interaction. Here, we show that the 8th zinc finger motif of MDS1/EVI1 is an oligomerization domain involved not only in interaction of AME with itself but also in interactions with the parental proteins, RUNX1 and MDS1/EVI1, from which AME is generated. Because the 8th zinc finger motif is also present in the oncoprotein EVI1, we have evaluated the effects of the interaction between RUNX1 and EVI1 in vitro and in vivo. We found that in vitro, this interaction alters the ability of RUNX1 to bind to DNA and to regulate a reporter gene, whereas in vivo, the expression of the isolated 8th zinc finger motif of EVI1 is sufficient to block the granulocyte colony-stimulating factor-induced differentiation of 32Dcl3 cells, leading to cell death. As EVI1 is not detected in normal bone marrow cells, these data suggest that its inappropriate expression could contribute to hematopoietic transformation in part by a new mechanism that involves EVI1 association with key hematopoietic regulators, leading to their functional impairment.


Subject(s)
Cell Transformation, Neoplastic , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Leukemia/genetics , Proto-Oncogenes/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Animals , Blotting, Western , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cloning, Molecular , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , NIH 3T3 Cells , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Transcription Factors/metabolism , Transfection , Zinc Fingers/physiology
18.
Gene ; 368: 1-11, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16314052

ABSTRACT

The ecotropic viral integration site 1 (EVI1) gene was identified almost 20 years ago as the integration site of an ecotropic retrovirus leading to murine myeloid leukemia. Since its identification, EVI1 has slowly been recognized as one of the most aggressive oncogenes associated with human leukemia. Despite the effort of many investigators, still very little is known about this gene. The mechanism by which EVI1 operates in the transformation of hematopoietic cells is not known, but it is clear that EVI1 upregulates cell proliferation, impairs cell differentiation, and induces cell transformation. In this review, we summarize the biochemical properties of EVI1 and the effects of EVI1 in biological models.


Subject(s)
DNA-Binding Proteins , Hematologic Diseases/metabolism , Proto-Oncogenes , Transcription Factors , Amino Acid Sequence , Animals , DNA-Binding Proteins/history , DNA-Binding Proteins/physiology , Hematologic Diseases/pathology , History, 20th Century , History, 21st Century , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , Molecular Sequence Data , Oncogene Proteins, Fusion , Phylogeny , Proto-Oncogenes/physiology , Sequence Homology, Amino Acid , Transcription Factors/history , Transcription Factors/physiology
19.
Cancer Res ; 65(17): 7603-11, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140925

ABSTRACT

AML1/MDS1/EVI1 (AME) is a chimeric transcription factor produced by the (3;21)(q26;q22) translocation. This chromosomal translocation is associated with de novo and therapy-related acute myeloid leukemia and with the blast crisis of chronic myelogenous leukemia. AME is obtained by in-frame fusion of the AML1 and MDS1/EVI1 (ME) genes. The mechanisms by which AME induces a neoplastic transformation in bone marrow cells are unknown. AME interacts with the corepressors CtBP and HDAC1, and it was shown that AME is a repressor in contrast to the parent transcription factors AML1 and ME, which are transcription activators. Studies with murine bone marrow progenitors indicated that the introduction of a point mutation that destroys the CtBP-binding consensus impairs but does not abolish the disruption of cell differentiation and replication associated with AME expression, suggesting that additional events are required. Several chimeric proteins, such as AML1/ETO, BCR/ABL, and PML/RARa, are characterized by the presence of a self-interaction domain critical for transformation. We report that AME is also able to oligomerize and displays a complex pattern of self-interaction that involves at least three oligomerization regions, one of which is the distal zinc finger domain. Although the deletion of this short domain does not preclude the self-interaction of AME, it significantly reduces the differentiation defects caused in vitro by AME in primary murine bone marrow progenitors. The addition of a point mutation that inhibits CtBP binding completely abrogates the effects of AME on differentiation, suggesting that AME induces hematopoietic differentiation defects through at least two separate but cooperating pathways.


Subject(s)
Hematopoietic Stem Cells/cytology , Oncogene Proteins, Fusion/metabolism , Repressor Proteins/metabolism , Zinc Fingers/physiology , Alcohol Oxidoreductases , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Humans , Mice , NIH 3T3 Cells , Oncogene Proteins, Fusion/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Repressor Proteins/genetics , Transfection , Up-Regulation , Zinc Fingers/genetics
20.
Arch Biochem Biophys ; 441(2): 168-73, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16122695

ABSTRACT

Transcription repression in eukaryotes is mediated by a wide variety of transcription factors that usually recruit corepressors and form corepressor complexes at the specific promoter sites. One of these corepressors is the C-terminal-binding protein (CtBP) which was first identified as a protein that binds to the C-terminal region of the adenovirus E1A protein. CtBP has a strong role in both development and oncogenesis. Till date, the mechanism of transcription repression by CtBP is unknown. Here, we report that CtBP physically interacts in vivo with HAT enzymes from different families. The vast majority of the HAT enzymes have a potential consensus site for CtBP binding within the bromodomain but we show that additional site(s) exists for CBP. The interaction between CtBP and CBP is functionally important and leads to impairment of histone H3 acetylation by CBP at specific lysine residues (Lys9, Lys14, and Lys18) in a dose-dependent and NADH-dependent manner. Based on these results, we propose that CtBP1 mediates repression by blocking histone acetylation by HAT complexes.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Kidney/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Alcohol Oxidoreductases , Amino Acid Sequence , Binding Sites , CREB-Binding Protein , Cell Line , Humans , Molecular Sequence Data , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...