Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
Add more filters











Publication year range
1.
J Steroid Biochem Mol Biol ; 187: 166-173, 2019 03.
Article in English | MEDLINE | ID: mdl-30508644

ABSTRACT

Standard therapy for Acute Myeloid Leukemia (AML) is rarely curative, and several suggested improvements have had little success so far. We have reported that in an in vitro model of a potential therapeutic regimen for AML, the activity of cytarabine (AraC) is enhanced by a sequential treatment with a combination of the vitamin D2 analog Doxercalciferol (D2) and the plant-derived antioxidant carnosic acid (CA). Importantly, the enhancement occurred selectively in patient-derived AML blasts, but not in the normal bone marrow cells. We now demonstrate that TXNIP, previously known as Vitamin D up-regulated protein 1 (VDUP1) [PMID 808674] plays a part in signaling cell death (CD) in this regimen. This is shown by the reduced CD when TXNIP protein levels are decreased by the CRISPR/CAS9 or RNAi technology. Further, we show that direct activation of ASK1 kinase by TXNIP is required for the optimal transmission of the CD signal to apoptotic machinery, regulated by JNK and BIM. These studies provide a rationale for a projected clinical trial of this vitamin D-based new therapeutic regimen for AML.


Subject(s)
Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Cytarabine/pharmacology , Ergocalciferols/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Signal Transduction/drug effects , Vitamins/pharmacology , Abietanes/pharmacology , Antioxidants/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 8/metabolism
2.
Curr Top Med Chem ; 6(12): 1267-71, 2006.
Article in English | MEDLINE | ID: mdl-16848740

ABSTRACT

Among the many important physiological functions of the activated vitamin D receptor (VDR) is the signaling of monocytic differentiation, first demonstrated by conversion of malignant myeloid leukemia cells to nonproliferating cells with mature monocyte/macrophage appearance. However, the understanding of how 1, 25-dihydroxyvitamin D3 (1,25D) signals monocytic differentiation is still developing. Recent advances summarized here include the role of the principal "mitogen-activated protein kinase" (MAPK) pathways, their potential downstream target the CCAAT/enhancer binding protein beta (C/EBP beta), cell cycle related proteins, and cyclin-dependent kinase 5 (Cdk5) in 1,25D-induced differentiation. The precise steps by which activated VDR signals differentiation are incompletely understood in any of the cell types known to respond to 1,25D. We have focused our studies on HL60 cells, a widely available cell line derived from a patient with promyeloblastic leukemia, with the goal of achieving as clear a picture as possible with the currently available tools. In this model, outlined in Fig. 1, a plausible sequence of events is presented, with the caveats that these are not the only pathways activated by liganded VDR, and that several other pathways, also operative, remain to be convincingly demonstrated. The details of the scheme will be discussed in the sections below.


Subject(s)
Leukemia/metabolism , Mitogen-Activated Protein Kinases/metabolism , Monocytes/cytology , Monocytes/metabolism , Receptors, Calcitriol/metabolism , Signal Transduction , Transcription Factors/metabolism , HL-60 Cells , Humans , Leukemia/pathology , MAP Kinase Signaling System
3.
Oncogene ; 25(53): 7078-85, 2006 Nov 09.
Article in English | MEDLINE | ID: mdl-16732322

ABSTRACT

Differentiation therapy is being developed as an additional therapeutic option for the treatment of several forms of cancer, including myeloid leukemia. In model systems, the physiologically active form of vitamin D, 1, alpha25-dihydroxyvitamin D3 (1,25D), induces monocytic differentiation of human myeloid cells, but the mechanism is not clear. We report here, the first direct connection between the signal provided by 1,25D and the molecular circuitry known to be involved in monocytic differentiation. Specifically, we show that 1,25D selectively increases the expression of the gene encoding kinase suppressor of Ras-1 (KSR-1) in HL60 cells, while other differentiation-inducing agents such as 12-O-tetradecanoylphorbol-13-acetate, retinoic acid or dimethyl sulfoxide do not significantly increase KSR-1 expression. Further, the upregulation of KSR-1 gene by 1,25D is competed by ZK159222, an antagonist of vitamin D receptor (VDR) action, and can occur in the presence of protein synthesis inhibitor cycloheximide, showing that the effect is direct. Most importantly, we have identified a vitamin D responsive element (VDRE) in the promoter region of the human KSR-1 gene, to which VDR binds in a 1,25D-dependent manner, in vitro and in vivo. This binding is paralleled by increased association of RNA polymerase II with the transcription start site of KSR-1 gene, and the VDRE is functional in reporter assays. Our findings offer a potential mechanism for a signaling pathway that contributes to 1,25D-induced monocytic differentiation of human myeloid leukemia cells.


Subject(s)
5' Flanking Region/genetics , Calcitriol/metabolism , Leukemia/enzymology , Leukemia/genetics , Protein Kinases/genetics , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Cell Differentiation/drug effects , Gene Expression Regulation, Neoplastic , HL-60 Cells , Humans , Leukemia/pathology , Promoter Regions, Genetic/genetics , Protein Biosynthesis , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/metabolism , Signal Transduction , Up-Regulation , Vitamin D Response Element
4.
Leukemia ; 19(10): 1812-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16107889

ABSTRACT

Human myeloid leukemia cell lines are induced to terminal differentiation into monocyte lineage by 1,25-dihydroxyvitamin D3 (1,25D3) or its analogs (deltanoids). However, translation of these findings to the clinic is limited by calcemic effects of deltanoids. Strategies to overcome this problem include combination of deltanoids with other compounds to induce differentiation at lower, noncalcemic, deltanoid concentrations. We previously showed that either carnosic acid, an antioxidant, or SB202190, a p38 MAPK inhibitor, increase the potency of 1,25D3 in the HL60 cell line. Here, we report that simultaneous addition of both these agents further increases differentiation potency of deltanoids in this cell line and in freshly obtained leukemic cells ex vivo. Activity of MAPK pathways showed that increased differentiation was associated with enhanced activity of JNK pathway in all responding cell subtypes. Our studies suggest that patients with CML or AML subtypes M2 and M4, but not M1, M3 or M4eo, are particularly suitable for this combination therapy. We conclude that the established cell line HL60 presents a good model for some, but not all, subtypes of myeloid leukemia, and that the JNK pathway plays an important role in monocytic differentiation of human leukemic cells ex vivo, as well as in vitro.


Subject(s)
Antioxidants/therapeutic use , Cell Differentiation/drug effects , Enzyme Inhibitors/therapeutic use , Leukemia, Myeloid/drug therapy , Vitamin D/analogs & derivatives , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Acute Disease , Adult , Aged , Aged, 80 and over , Calcium/metabolism , Cell Lineage , Drug Therapy, Combination , Female , Humans , Leukemia, Myeloid/classification , Leukemia, Myeloid/metabolism , Male , Middle Aged , Monocytes/metabolism , Vitamin D/therapeutic use , p38 Mitogen-Activated Protein Kinases/drug effects
5.
J Natl Cancer Inst ; 93(16): 1224-33, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11504768

ABSTRACT

BACKGROUND: Carnosic acid is a plant-derived polyphenol food preservative with chemoprotective effects against carcinogens when tested in animals. Recently, we showed that carnosic acid potentiates the effects of 1alpha,25-dihydroxyvitamin D3 (1alpha,25[OH]2D3) and of all-trans-retinoic acid (ATRA) on differentiation of human leukemia cells. We now examine the mechanisms associated with carnosic acid-induced enhancement of cell differentiation (in subline HL60-G) initiated by 1alpha,25(OH)2D3, ATRA, or 12-O-tetradecanoylphorbol-13-acetate (TPA). METHODS: We evaluated monocytic differentiation markers (CD11b, CD14, and monocytic serine esterase), cell cycle parameters, and cell proliferation rates after treatment of cells with different agents with or without carnosic acid. We also assessed the abundance of the vitamin D receptor (VDR), retinoid X receptor (RXR)-alpha, retinoic acid receptor (RAR)-alpha, and cell cycle-associated proteins by immunoblot analysis (p27, early growth response gene [EGR]-1, and p35Nck5a), the expression of corresponding genes by reverse transcription-polymerase chain reaction (RT-PCR), and the activity of VDR by electrophoretic mobility shift analysis. The two-sided nonparametric Kruskal-Wallis one-way analysis-of-variance test with Dunn's adjustment was used for statistical analyses. RESULTS: Monocytic differentiation induced by low (1 nM) concentrations of 1alpha,25(OH)2D3, ATRA, or TPA was enhanced by carnosic acid (10 microM), as shown by the increased expression of monocytic serine esterase (P<.001, P<.001, and P =.043, respectively) and of CD11b (P =.008, P =.046, and P =.041, respectively). Increased expression of CD14 was seen only for 1alpha,25(OH)2D3 and ATRA (P =.009 and P =.048, respectively) and also for several cell cycle-associated proteins. Carnosic acid in combination with 1alpha,25(OH)2D3 and ATRA resulted in decreased cell proliferation and blocked the cell cycle transition from G1 to S phase (P<.05). Carnosic acid alone increased the expression of VDR and RXR-alpha, but the expression was greatly enhanced in the presence of 1alpha,25(OH)2D3 and ATRA. In combination with TPA, carnosic acid potentiated the expression of VDR and RAR-alpha. CONCLUSION: Carnosic acid enhances a program of gene expression consistent with 1alpha,25(OH)2D3-, ATRA-, or TPA-induced monocytic differentiation of HL60-G cells.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Diterpenes/pharmacology , Granulocytes/drug effects , Monocytes/drug effects , Plant Extracts/pharmacology , Abietanes , Analysis of Variance , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Blotting, Western , Calcitriol/pharmacology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Survival/drug effects , DNA-Binding Proteins/analysis , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Receptors, Calcitriol/analysis , Receptors, Retinoic Acid/administration & dosage , Receptors, Retinoic Acid/analysis , Retinoid X Receptors , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/analysis , Tretinoin/pharmacology
6.
Exp Cell Res ; 268(2): 294-300, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11478855

ABSTRACT

Determination of the involvement of MAP kinase cascades in signaling cell growth or differentiation is aided by the use of the inhibitors PD 098059 [2-(2'-amino-3'-methoxyphenyl)oxananphthalen-4-one] and U0126 [1,4-diamino-2,3-dicyano-1,4-bis(2-aminophenylthio)butadiene], believed to be MEK-specific kinase inhibitors. We report here that the activity of kinase suppressor of ras (KSR-1), a kinase upstream of raf-1, is inhibited by both these compounds at concentrations similar to those that inhibit MEK-1. Further, in HL60 cells induced to differentiate with 1,25-dihydroxyvitamin D(3) raf-1 and p90RSK, but not ERK1/2, are coregulated, and their expression as well as monocytic differentiation is inhibited in parallel by PD 098059. Thus, in this system raf-1 is phosphorylated by KSR-1, and PD 098059 as well as U0126 inhibits this phosphorylation. This suggests great caution in the interpretation of experiments that utilize these pharmacological inhibitors of kinase activity as evidence for a role for the MEK--ERK module in ras or raf-1 signaling.


Subject(s)
Calcitriol/pharmacology , MAP Kinase Kinase Kinase 1 , Monocytes/cytology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/metabolism , Butadienes/pharmacology , Cell Differentiation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , HL-60 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Nitriles/pharmacology , Phosphorylation/drug effects , Protein Kinases/drug effects , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction
7.
J Cell Biochem ; 82(1): 68-77, 2001.
Article in English | MEDLINE | ID: mdl-11400164

ABSTRACT

Monocytic differentiation of HL60 cells induced by 1,25-dihydroxyvitamin D(3) (1,25 D(3)) has been recently shown (Exp Cell Res 258, 425, 2000) to be enhanced by an exposure to SB203580 or to SB202190, specific inhibitors of p38MAP kinase, with concomitant up-regulation of the c-jun N terminal kinase (JNK) pathway. In the present study we inquired if this enhancement and the JNK up-regulation are limited to 1,25 D(3)-induced differentiation, or if they occur more generally in HL60 cell differentiation. We found that dimethylsulfoxide (DMSO)-induced differentiation, and to a lesser extent tetradecanoylphorbol acetate (TPA)-induced macrophage differentiation were also potentiated by the p38MAPK inhibitors, but that granulocytic differentiation in response to all-trans retinoic acid (RA) was not. The enhancement of differentiation by p38MAPK inhibitors was accompanied by an activation of the JNK MAPK pathway, as shown by the phosphorylation levels of these kinases and by AP-1 binding, but only in 1,25 D(3)-treated cells. This shows that an up-regulation of the JNK stress pathway during 1,25 D(3)-induced monocytic differentiation occurs selectively in this lineage of differentiation and is not necessary for the expression of the differentiated phenotype.


Subject(s)
HL-60 Cells/cytology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Monocytes/metabolism , Transcription Factor AP-1/metabolism , Antigens, Differentiation, Myelomonocytic/drug effects , Antigens, Surface/drug effects , Calcitriol/pharmacology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Dimethyl Sulfoxide/pharmacology , Granulocytes/cytology , Granulocytes/metabolism , HL-60 Cells/metabolism , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases , Macrophages/cytology , Macrophages/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Pyridines/metabolism , Pyridines/pharmacology , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factor AP-1/agonists , Up-Regulation , p38 Mitogen-Activated Protein Kinases
8.
Blood ; 97(12): 3763-7, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11389014

ABSTRACT

Although cyclin-dependent kinase 5 (Cdk5) is widely expressed in human tissues, its activator p35Nck5a is generally considered to be neuron specific. In addition to neuronal cells, active Cdk5 complexes have been reported in developing tissues, such as the embryonic muscle and ocular lens, and in human leukemia HL60 cells induced to differentiate by an exposure to 1,25-dihydroxyvitamin D(3); however, its activator in these cells has not been demonstrated. The results of this study indicate that p35Nck5a is associated with Cdk5 in monocytic differentiation of hematopoietic cells. Specifically, p35Nck5a is expressed in normal human monocytes and in leukemic cells induced to differentiate toward the monocytic lineage, but not in lymphocytes or cells induced to granulocytic differentiation by retinoic acid. It is present in a complex with Cdk5 that has protein kinase activity, and when ectopically expressed together with Cdk5 in undifferentiated HL60 cells, it induces the expression of CD14 and "nonspecific" esterase, markers of monocytic phenotype. These observations not only indicate a functional relationship between Cdk5 and p35Nck5a, but also support a role for this complex in monocytic differentiation. (Blood. 2001;97:3763-3767)


Subject(s)
Cell Cycle Proteins , Monocytes/cytology , Nerve Tissue Proteins/biosynthesis , Saccharomyces cerevisiae Proteins , Cell Differentiation/drug effects , Granulocytes/cytology , Granulocytes/metabolism , HL-60 Cells , Humans , Immunophenotyping , Lymphocytes/cytology , Lymphocytes/metabolism , Lymphoma/pathology , Monocytes/metabolism , Precipitin Tests , Protein Binding , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Transfection , Tumor Cells, Cultured
9.
Steroids ; 66(3-5): 463-71, 2001.
Article in English | MEDLINE | ID: mdl-11179755

ABSTRACT

The secosteroid hormone 1alpha,25-dihydroxyvitamin D(3) [1alpha,25(OH)(2)D(3)] is metabolized in its target tissues through modifications of both the side chain and the A-ring. The C-24 oxidation pathway, the main side chain modification pathway is initiated by hydroxylation at C-24 of the side chain and leads to the formation of the end product, calcitroic acid. The C-23 and C-26 oxidation pathways, the minor side chain modification pathways are initiated by hydroxylations at C-23 and C-26 of the side chain and lead to the formation of the end product, calcitriol lactone. The C-3 epimerization pathway, the newly discovered A-ring modification pathway is initiated by epimerization of the hydroxyl group at C-3 of the A-ring to form 1alpha,25(OH)(2)-3-epi-D(3). A rational design for the synthesis of potent analogs of 1alpha,25(OH)(2)D(3) is developed based on the knowledge of the various metabolic pathways of 1alpha,25(OH)(2)D(3). Structural modifications around the C-20 position, such as C-20 epimerization or introduction of the 16-double bond affect the configuration of the side chain. This results in the arrest of the C-24 hydroxylation initiated cascade of side chain modifications at the C-24 oxo stage, thus producing the stable C-24 oxo metabolites which are as active as their parent analogs. To prevent C-23 and C-24 hydroxylations, cis or trans double bonds, or a triple bond are incorporated in between C-23 and C-24. To prevent C-26 hydroxylation, the hydrogens on these carbons are replaced with fluorines. Furthermore, testing the metabolic fate of the various analogs with modifications of the A-ring, it was found that the rate of C-3 epimerization of 5,6-trans or 19-nor analogs is decreased to a significant extent. Assembly of all these protective structural modifications in single molecules has then produced the most active vitamin D(3) analogs 1alpha,25(OH)(2)-16,23-E-diene-26,27-hexafluoro-19-nor-D(3) (Ro 25-9022), 1alpha,25(OH)(2)-16,23-Z-diene-26,27-hexafluoro-19-nor-D(3) (Ro 26-2198), and 1alpha,25(OH)(2)-16-ene-23-yne-26,27-hexafluoro-19-nor-D(3) (Ro 25-6760), as indicated by their antiproliferative activities.


Subject(s)
Calcitriol/metabolism , Calcitriol/pharmacology , Animals , Calcitriol/analogs & derivatives , Calcitriol/chemistry , Humans , Molecular Structure , Oxidation-Reduction , Structure-Activity Relationship , Vitamin D/analogs & derivatives , Vitamin D/chemistry , Vitamin D/metabolism , Vitamin D/pharmacology
10.
J Cell Biochem ; 80(4): 471-82, 2001.
Article in English | MEDLINE | ID: mdl-11169731

ABSTRACT

Activation of ERK1 and ERK2 protein kinases has been implicated in diverse cellular processes, including the control of cell proliferation and cell differentiation (Marshall [1995] Cell 80:179). In human myeloblastoid leukemia HL60 cells rapid (ca. 15 min) but transient activation of ERK1/2 has been reported following induction of macrophage/monocyte differentiation by phorbol esters, or by very high (10(-6) M) concentrations of 1,25-dihydroxyvitamin D(3) (1,25D3), while retinoic acid-induced granulocytic differentiation was accompanied by sustained activation of ERK1/2. We report here that monocytic differentiation of HL60 cells induced by moderate (10(-9) to 10(-7) M) concentrations of 1,25D3 could be divided into at least two stages. In the first phase, which lasts 24-48 h, the cells continued in the normal cell cycle while expressing markers of monocytic phenotype, such as CD14. In the next phase the onset of G1 cell cycle block became apparent and expression of CD11b was prominent, indicating a more mature myeloid phenotype. The first phase was characterized by high levels of ERKs activated by phosphorylation, and these decreased as the cells entered the second phase, while the levels of p27/Kip1 increased at that time. Serum-starved or PD98059-treated HL60 cells had reduced growth rate and slower differentiation, but the G1 block also coincided with decreased levels of activated ERK1/2. The data suggest that the MEK/ERK pathway maintains cell proliferation during 1,25D3-induced monocytic differentiation of HL60 cells, but that ERK1/2 activity becomes suppressed during the later stages of differentiation, and the consequent G1 block leads to "terminal" differentiation.


Subject(s)
Calcitriol/metabolism , Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Biomarkers , Blotting, Western , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Nucleus/metabolism , Culture Media, Serum-Free/pharmacology , Cytoplasm/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , G1 Phase/drug effects , HL-60 Cells , Humans , Immunoblotting , Kinetics , Lipopolysaccharide Receptors/metabolism , Macrophage-1 Antigen/metabolism , Models, Biological , Monocytes/cytology , Phenotype , Phosphorylation , Time Factors , Tretinoin/pharmacology
11.
Clin Cancer Res ; 6(7): 2951-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914745

ABSTRACT

Differentiation agents use existing cellular systems to induce neoplastic cells to regain a normal phenotype and/or to cause growth arrest and therefore may offer novel chemotherapeutic approaches to treating solid tumors. In this study, we demonstrate in Caco-2 colon cancer cells that the differentiation agent phenylbutyrate (PB) causes a decrease in viable cells, an increase in cell differentiation, and a G1-S-phase block. The mechanism of this last effect is related to a PB-induced increase in p27Kip1, leading to a decrease in the activity of cyclin-dependent kinase 2 (CDK2), a positive regulator of the G1-S-phase cell cycle transition. Consistent with the decreased CDK2 kinase activity, we also observed a decrease in the phosphorylation state of the retinoblastoma protein after PB treatment. This was associated with increased binding and consequent inactivation of E2F, a transactivator of genes that regulate the G1 to S phase cell cycle transition. These data suggest that the differentiation agent PB inhibits tumor growth by limiting the availability of active E2F, with a subsequent G1-S-phase block. Additional studies should show whether PB is a clinically effective therapeutic agent against colorectal cancer.


Subject(s)
CDC2-CDC28 Kinases , Carrier Proteins , Cell Cycle Proteins , Cell Cycle/physiology , Cell Differentiation/physiology , DNA-Binding Proteins , Phenylbutyrates/pharmacology , Proto-Oncogene Proteins , Transcription Factors/metabolism , Tumor Suppressor Proteins , Alkaline Phosphatase/metabolism , Caco-2 Cells , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , E2F Transcription Factors , G1 Phase , Humans , Kinetics , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma-Binding Protein 1 , S Phase , Time Factors , Transcription Factor DP1
12.
Exp Cell Res ; 258(2): 425-37, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10896794

ABSTRACT

Differentiation therapy for neoplastic diseases has potential for supplementing existing treatment modalities but its implementation has been slow. One of the reasons is the lack of full understanding of the complexities of cellular pathways through which signals for differentiation lead to cell maturation. This was addressed in this study using HL60 cells, a well-established model of differentiation of neoplastic cells. SB 203580 and SB 202190, specific inhibitors of a signaling protein p38 MAP kinase, were found to markedly accelerate monocytic differentiation of HL60 cells induced by low concentrations of 1,25-dihydroxyvitamin D(3) (1,25D(3)). Surprisingly, inhibition of p38 activity resulted in sustained enhancement of p38 phosphorylation and of its in vitro activity in the absence of the inhibitor, indicating up-regulation of the upstream components of the p38 pathway. In addition, SB 203580 or SB 202190 treatment of HL60 cells resulted in a prolonged activation of the JNK and, to a lesser extent, the ERK pathways. The data are consistent with the hypothesis that in HL60 cells an interruption of a negative feedback loop from a p38 target activates a common regulator of multiple MAPK pathways. The possibility also exists that JNK and/or ERK pathways amplify a differentiation signal provided by 1,25D(3).


Subject(s)
Calcitriol/pharmacology , Cell Differentiation , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Up-Regulation , Calcitriol/metabolism , Cell Count , Cell Differentiation/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , HL-60 Cells , Humans , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/metabolism , Monocytes/drug effects , Monocytes/metabolism , Phosphorylation , Pyridines/pharmacology , S Phase , Time Factors , p38 Mitogen-Activated Protein Kinases
13.
Anticancer Res ; 20(2A): 661-7, 2000.
Article in English | MEDLINE | ID: mdl-10810337

ABSTRACT

The role of the transcription factor Sp1 in cell growth and survival was investigated by induced expression of its DNA-binding C-terminal fragment. Transfection of a constitutively expressed Sp1-170C plasmid construct into HeLa cells failed to produce viable clones, suggesting that this peptide interferes with cell growth. However, transfection with the muristerone A-inducible vector system produced four clones with low levels of expression of Sp1-170C. Muristerone A transiently induced higher levels of Sp1-170C, and this was accompanied by a reduced growth rate and prolongation of the S phase of the cell cycle. This is the first report that a dominant negative Sp1 can affect the cell cycle.


Subject(s)
Cell Cycle , Ecdysterone/analogs & derivatives , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Base Sequence , Cell Division/drug effects , Ecdysterone/pharmacology , HeLa Cells , Humans , Kinetics , Recombinant Proteins/metabolism , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction , S Phase/drug effects , Transfection
14.
J Leukoc Biol ; 67(4): 559-66, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10770290

ABSTRACT

Hematopoietic cell differentiation takes place in phenotypically recognizable stages characterized by morphology as well as by the expression of enzymes and surface markers. It is recognized that differentiation results from an interaction of environmental cues, such as cytokines and hormones, with internal cellular programs, but the precise mechanisms are not entirely clear. HL60 cells, a human acute myeloid leukemia (AML) cell line with promyelocytic features, provide a model for such studies because they behave like stem cells, which can differentiate into two different lineages, granulocytic or monocytic/macrophage, depending on the inducer. Protein levels and kinase activity of cyclin-dependent kinase 5 (Cdk5) were reported [F. Chen and G. P. Studzinski (1999) Exp. Cell Res. 249, 422, 1999] to increase in HL60 cells induced to monocytic differentiation by 1alpha,25-dihydroxyvitamin D3 (1,25D3), but the specificity of the association of Cdk5 with the monocytic phenotype has not been established. We show here that up-regulation of Cdk5 does not occur in granulocytic differentiation, whereas inhibition of Cdk5 activity by olomoucine, or its expression by a plasmid construct expressing antisense Cdk5, switches the 1,25D3-induced monocytic phenotype (a combination of positive nonspecific esterase reaction, expression of the CD14 marker, and morphology) to general myeloid phenotype (positive nitro-blue tetrazolium reaction, CD11b marker and morphology). The transcriptional up-regulation of Cdk5 by 1,25D3 was not inhibited by olomoucine. These findings show that in human myeloid cells up-regulation of Cdk5 is specifically associated with the monocytic phenotype.


Subject(s)
Cell Lineage , Cyclin-Dependent Kinases/biosynthesis , Monocytes/pathology , Biomarkers , Calcitriol/pharmacology , Cell Differentiation/drug effects , Cyclin-Dependent Kinase 5 , HL-60 Cells , Humans , Monocytes/enzymology , Up-Regulation/drug effects
15.
J Cell Physiol ; 183(2): 238-46, 2000 May.
Article in English | MEDLINE | ID: mdl-10737899

ABSTRACT

p53/56(lyn) is a member of the src family that is predominantly expressed in hematopoietic cells and is thought to play a role in cellular proliferation. In this study, we demonstrate the participation of p53/56(lyn) in 1,25-dihydroxyvitamin D(3) (1, 25D(3))-induced growth arrest in HL60 cells. We show that the mRNA and protein levels of p53/56(lyn) are markedly elevated after 1, 25D(3) treatment, which is accompanied by an increase of p53/56(lyn) kinase activity. We also demonstrate that treatment with p53/56(lyn) antisense oligodeoxynucleotides reverses the 1,25D(3)-induced G1/S block, and results in an accumulation of cells with S-phase DNA content. BrdU pulse-chase experiments reveal that this accumulation results from an increased proportion of cells actively synthesizing DNA, which are inhibited from exiting the S-phase compartment. These results indicate that upregulation of p53/56(lyn) contributes significantly to the G1/S growth arrest induced by 1,25D(3) in HL60 cells and thus its activation may be a desirable outcome of chemotherapeutic regimens.


Subject(s)
Calcitriol/pharmacology , G1 Phase/drug effects , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , S Phase/drug effects , src-Family Kinases/genetics , Base Sequence , Cell Differentiation/drug effects , Cell Division/drug effects , DNA/metabolism , DNA Primers/genetics , Gene Expression/drug effects , HL-60 Cells , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , src-Family Kinases/metabolism
16.
Proc Soc Exp Biol Med ; 222(2): 150-6, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10564539

ABSTRACT

Butyrate, a short-chain fatty acid, has been reported to inhibit proliferation and stimulate differentiation in multiple cancer cell lines. Whereas the effects of butyrate on cellular differentiation are well documented, the relationship between butyrate-induced differentiation and its effect on cell cycle traverse is less well understood. The purpose of this study was to investigate the effects of butyrate on the regulatory proteins of the G2/M traverse in the Caco-2 colon cancer cell model. We demonstrated that the inhibition of proliferation and increased cellular differentiation after treatment of Caco-2 cells with butyrate were associated with a significant G2/M cell cycle block. Although protein levels of the major G2/M regulatory protein, p34cdc2, were unchanged, a decrease in p34cdc2 activity was noted. Despite this decrease in activity, the inhibitory tyrosine phosphorylation of p34cdc2 was decreased, suggesting that other factors are responsible for the decreased kinase activity. The reduced activity of p34cdc2 provides a possible mechanism for the accumulation of Caco-2 cells in the G2/M cell cycle compartment following exposure to butyrate. This cell system provides a new model for studies of G2/M cell cycle perturbations.


Subject(s)
Butyrates/pharmacology , CDC2 Protein Kinase/metabolism , Cell Cycle/drug effects , Alkaline Phosphatase/metabolism , Caco-2 Cells , Cell Cycle/physiology , Cell Differentiation/drug effects , Cell Division/drug effects , DNA, Neoplasm/metabolism , Flow Cytometry , G2 Phase , Humans , Kinetics , Mitosis , Models, Biological
17.
J Cell Biochem ; 75(2): 226-34, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10502295

ABSTRACT

Cellular differentiation of neoplastic cells after exposure to 1, 25-dihydroxyvitamin D(3) (1,25 D(3)) is accompanied by altered cell cycle regulation. In previous studies, blocks in both G(1)/S and G(2)/M checkpoints have been observed in 1,25D(3)-treated HL60 cells, but the mechanism of the 1,25D(3)-induced G(2)/M block has not been previously reported. In this study, we show by cell cycle analysis, using bromodeoxyuridine pulse-chase labeling, that the G(2)/M block in 1,25D(3)-treated HL60 cells is incomplete. We also demonstrate that although the 1,25D(3)-treated cells exhibit elevated levels of cyclin B1, Cdc25C, and Cdk7, which are positive regulators of the G(2)/M traverse, these cells have decreased protein levels of p34(cdc2) and decreased p34(cdc2) kinase activity. This provides potential mechanisms for the observed accumulation of cells in the G(2) cell cycle compartment and occasional polyploidization following treatment of HL60 cells with 1,25D(3). The data also suggest that the ability of some cells to traverse this block may be the result of cellular compensatory mechanisms responding to decreased p34(cdc2) activity by increasing the levels of other regulators of the G(2) traverse, such as cyclin B1, Cdc25C, and Cdk7.


Subject(s)
CDC2 Protein Kinase/metabolism , Calcitriol/pharmacology , Cell Division/drug effects , Cyclin-Dependent Kinases , G2 Phase/drug effects , Mitosis/drug effects , Bromodeoxyuridine/metabolism , CDC2 Protein Kinase/drug effects , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cyclin B/metabolism , Cyclin B1 , Flow Cytometry , HL-60 Cells , Histones/metabolism , Humans , Immunoblotting , Models, Biological , Precipitin Tests , Protein Serine-Threonine Kinases/metabolism , Time Factors , Up-Regulation , cdc25 Phosphatases/metabolism , Cyclin-Dependent Kinase-Activating Kinase
18.
Exp Cell Res ; 249(2): 422-8, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10366442

ABSTRACT

The function of most cyclin-dependent kinases (Cdks) is to facilitate progression through the checkpoints of the cell cycle, but Cdk5 is known to be involved in differentiation of CNS, muscle, and lens cells, though not in the cell cycle traverse. Here we show an additional role for Cdk5, an enhancement of monocytic differentiation with abrogation of the G1 checkpoint. Human leukemia HL60 cells exposed to 1alpha,25-dihydroxyvitamin D3 (1,25D3) displayed monocytic phenotype and increased Cdk5 kinase activity. An analog of 1,25D3 which does not induce differentiation failed to upregulate Cdk5, and 1,25D3-resistant cells had reduced Cdk5 activity. Active or inactive Cdk5 was associated with cyclin D1, but only active Cdk5 exhibited threonine phosphorylation. Inhibition of Cdk5 expression by an antisense construct reduced the intensity of 1, 25D3-induced expression of CD14, a marker of monocytes, and increased the 1,25D3-induced G1 block. These findings demonstrate a novel aspect of Cdk5 activity-facilitation of the G1- to S-phase transition in cells which are approaching replicative quiescence and a concomitant enhancement of monocytic differentiation.


Subject(s)
Calcitriol/pharmacology , Cell Cycle , Cyclin-Dependent Kinases/metabolism , Monocytes/enzymology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cyclin-Dependent Kinase 5 , Cyclin-Dependent Kinases/genetics , DNA, Antisense/genetics , DNA, Antisense/pharmacology , G1 Phase/drug effects , HL-60 Cells , Humans , Monocytes/cytology , Monocytes/drug effects , Phagocytosis/drug effects , Phenotype , Transfection
19.
Int Rev Cytol ; 189: 1-58, 1999.
Article in English | MEDLINE | ID: mdl-10333577

ABSTRACT

This review examines recent developments relating to the interface between cell proliferation and differentiation. It is suggested that the mechanism responsible for this transition is more akin to a "dimmer" than to a "switch," that it is more useful to refer to early and late stages of differentiation rather than to "terminal" differentiation, and examples of the reversibility of differentiation are provided. An outline of the established paradigm of cell cycle regulation is followed by summaries of recent studies that suggest that this paradigm is overly simplified and should be interpreted in the context of different cell types. The role of inhibitors of cyclin-dependent kinases in differentiation is discussed, but the data are still inconclusive. An increasing interest in the changes in G2/M transition during differentiation is illustrated by examples of polyploidization during differentiation, such as megakaryocyte maturation. Although the retinoblastoma protein is currently maintaining its prominent role in control of proliferation and differentiation, it is anticipated that equally important regulators will be discovered and provide an explanation at the molecular level for the gradual transition from proliferation to differentiation.


Subject(s)
Cell Cycle/physiology , Cell Differentiation/physiology , Animals , Cell Division/physiology , Cell Lineage , Colon/cytology , Hematopoietic Stem Cells/cytology , Humans , Models, Biological , Transcription Factors/physiology
20.
Exp Cell Res ; 248(1): 58-73, 1999 Apr 10.
Article in English | MEDLINE | ID: mdl-10094813

ABSTRACT

Differentiation of mammalian cells implies cessation of DNA replication and cell proliferation; the potential controls of this coupling are examined here. It is clear that the known or proposed mechanisms of down-regulation of replicative cellular activities vary in different lineages of cell differentiation, and occur in all phases of the cell cycle. In G1 these regulators include p21/Cip1 or p27/Kip1, pRb, and p53; the novel, recently reported mechanisms of their action are summarized. In S phase the availability of nucleotide precursors, the origin recognition complex (ORC), and other replication proteins may be important in differentiation, and in G2 phase the cdc2/cyclin B complex and replication licensing factors determine normal G2 traverse versus an arrest or polyploidisation. Other replication-related mechanisms include transcription factors, e.g., Sp1, telomerase, and nuclear matrix changes. Thus, differentiation alters the activity not only of the various checkpoint proteins, but also of the components of the replicative machinery itself.


Subject(s)
DNA Replication , Animals , G1 Phase , G2 Phase , Humans , Mitosis , S Phase
SELECTION OF CITATIONS
SEARCH DETAIL