Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892135

ABSTRACT

Podophyllotoxin (PPT) is an active pharmaceutical ingredient (API) with established antitumor potential. However, due to its systemic toxicity, its use is restricted to topical treatment of anogenital warts. Less toxic PPT derivatives (e.g., etoposide and teniposide) are used intravenously as anticancer agents. PPT has been exploited as a scaffold of new potential therapeutic agents; however, fewer studies have been conducted on the parent molecule than on its derivatives. We have undertaken a study of ultrastructural changes induced by PPT on HaCaT keratinocytes. We have also tracked the intracellular localization of PPT using its fluorescent derivative (PPT-FL). Moreover, we performed molecular docking of both PPT and PPT-FL to compare their affinity to various binding sites of tubulin. Using the Presto blue viability assay, we established working concentrations of PPT in HaCaT cells. Subsequently, we have used selected concentrations to determine PPT effects at the ultrastructural level. Dynamics of PPT distribution by confocal microscopy was performed using PPT-FL. Molecular docking calculations were conducted using Glide. PPT induces a time-dependent cytotoxic effect on HaCaT cells. Within 24 h, we observed the elongation of cytoplasmic processes, formation of cytoplasmic vacuoles, progressive ER stress, and shortening of the mitochondrial long axis. After 48 h, we noticed disintegration of the cell membrane, progressive vacuolization, apoptotic/necrotic vesicles, and a change in the cell nucleus's appearance. PPT-FL was detected within HaCaT cells after ~10 min of incubation and remained within cells in the following measurements. Molecular docking confirmed the formation of a stable complex between tubulin and both PPT and PPT-FL. However, it was formed at different binding sites. PPT is highly toxic to normal human keratinocytes, even at low concentrations. It promptly enters the cells, probably via endocytosis. At lower concentrations, PPT causes disruptions in both ER and mitochondria, while at higher concentrations, it leads to massive vacuolization with subsequent cell death. The novel derivative of PPT, PPT-FL, forms a stable complex with tubulin, and therefore, it is a useful tracker of intracellular PPT binding and trafficking.


Subject(s)
HaCaT Cells , Keratinocytes , Molecular Docking Simulation , Podophyllotoxin , Tubulin , Humans , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Podophyllotoxin/chemistry , Tubulin/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Cell Survival/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Fluorescent Dyes/chemistry , Binding Sites , Endoplasmic Reticulum Stress/drug effects
2.
J Am Heart Assoc ; 9(9): e015611, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32319345

ABSTRACT

Background Approximately 1 in 6 adolescents report regular binge alcohol consumption, and we hypothesize it affects heart growth during this period. Methods and Results Adolescent, genetically diverse, male Wistar rats were gavaged with water or ethanol once per day for 6 days. In vivo structure and function were assessed before and after exposure. Binge alcohol exposure in adolescence significantly impaired normal cardiac growth but did not affect whole-body growth during adolescence, therefore this pathology was specific to the heart. Binge rats also exhibited signs of accelerated pathological growth (concentric cellular hypertrophy and thickening of the myocardial wall), suggesting a global reorientation from physiologic to pathologic growth. Binge rats compensated for their smaller filling volumes by increasing systolic function and sympathetic stimulation. Consequently, binge alcohol exposure increased PKA (protein kinase A) phosphorylation of troponin I, inducing myofilament calcium desensitization. Binge alcohol also impaired in vivo relaxation and increased titin-based cellular stiffness due to titin phosphorylation by PKCα (protein kinase C α). Mechanistically, alcohol inhibited extracellular signal-related kinase activity, a nodal signaling kinase activating physiology hypertrophy. Thus, binge alcohol exposure depressed genes involved in growth. These cardiac structural alterations from binge alcohol exposure persisted through adolescence even after cessation of ethanol exposure. Conclusions Alcohol negatively impacts function in the adult heart, but the adolescent heart is substantially more sensitive to its effects. This difference is likely because adolescent binge alcohol impedes the normal rapid physiological growth and reorients it towards pathological hypertrophy. Many adolescents regularly binge alcohol, and here we report a novel pathological consequence as well as mechanisms involved.


Subject(s)
Binge Drinking/complications , Cardiomegaly/etiology , Heart/growth & development , Myocardium/pathology , Adaptation, Physiological , Age Factors , Animals , Calcium Signaling , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Connectin/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Myocardium/enzymology , Phosphorylation , Protein Kinase C-alpha/metabolism , Rats, Wistar , Troponin I/metabolism
3.
Cancer Res ; 79(19): 4965-4977, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31387918

ABSTRACT

Estrogen receptor (ER)-positive breast cancer recurrence is thought to be driven by tumor-initiating cells (TIC). TICs are enriched by endocrine therapy through NOTCH signaling. Side effects have limited clinical trial testing of NOTCH-targeted therapies. Death-associated factor 6 (DAXX) is a newly identified marker whose RNA expression inversely correlates with NOTCH in human ER+ breast tumor samples. In this study, knockdown and overexpression approaches were used to investigate the role of DAXX on stem/pluripotent gene expression, TIC survival in vitro, and TIC frequency in vivo, and the mechanism by which DAXX suppresses TICs in ER+ breast cancer. 17ß-Estradiol (E2)-mediated ER activation stabilized the DAXX protein, which was required for repressing stem/pluripotent genes (NOTCH4, SOX2, OCT4, NANOG, and ALDH1A1), and TICs in vitro and in vivo. Conversely, endocrine therapy promoted rapid protein depletion due to increased proteasome activity. DAXX was enriched at promoters of stem/pluripotent genes, which was lost with endocrine therapy. Ectopic expression of DAXX decreased stem/pluripotent gene transcripts to levels similar to E2 treatment. DAXX-mediated repression of stem/pluripotent genes and suppression of TICs was dependent on DNMT1. DAXX or DNMT1 was necessary to inhibit methylation of CpGs within the SOX2 promoter and moderately within the gene body of NOTCH4, NOTCH activation, and TIC survival. E2-mediated stabilization of DAXX was necessary and sufficient to repress stem/pluripotent genes by recruiting DNMT1 to methylate some promoters and suppress TICs. These findings suggest that a combination of endocrine therapy and DAXX-stabilizing agents may inhibit ER+ tumor recurrence. SIGNIFICANCE: Estradiol-mediated stabilization of DAXX is necessary and sufficient to repress genes associated with stemness, suggesting that the combination of endocrine therapy and DAXX-stabilizing agents may inhibit tumor recurrence in ER+ breast cancer.


Subject(s)
Breast Neoplasms/pathology , Co-Repressor Proteins/metabolism , Drug Resistance, Neoplasm/physiology , Molecular Chaperones/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Female , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/pathology , Receptors, Estrogen/metabolism
4.
Clin Cancer Res ; 22(1): 175-86, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26350262

ABSTRACT

PURPOSE: Breast cancer is the second leading cause of cancer mortality among women worldwide. The major problem with current treatments is tumor resistance, recurrence, and disease progression. ErbB-2-positive breast tumors are aggressive and frequently become resistant to trastuzumab or lapatinib. We showed previously that Notch-1 is required for trastuzumab resistance in ErbB-2-positive breast cancer. EXPERIMENTAL DESIGN: Here, we sought to elucidate mechanisms by which ErbB-2 attenuates Notch signaling and how this is reversed by trastuzumab or lapatinib. RESULTS: The current study elucidates a novel Notch inhibitory mechanism by which PKCα downstream of ErbB-2 (i) restricts the availability of Jagged-1 at the cell surface to transactivate Notch, (ii) restricts the critical interaction between Jagged-1 and Mindbomb-1, an E3 ligase that is required for Jagged-1 ubiquitinylation and subsequent Notch activation, (iii) reverses trastuzumab resistance in vivo, and (iv) predicts better outcome in women with ErbB-2-positive breast cancer. CONCLUSIONS: The clinical impact of these studies is PKCα is potentially a good prognostic marker for low Notch activity and increased trastuzumab sensitivity in ErbB-2-positive breast cancer. Moreover, women with ErbB-2-positive breast tumors expressing high Notch activation and low PKCα expression could be the best candidates for anti-Notch therapy.


Subject(s)
Breast Neoplasms/metabolism , Calcium-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Protein Kinase C-alpha/metabolism , Receptor, ErbB-2/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Trastuzumab/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Jagged-1 Protein , Neoplasm Recurrence, Local , Prognosis , Protein Binding , Protein Transport , Serrate-Jagged Proteins , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
5.
Front Oncol ; 4: 360, 2014.
Article in English | MEDLINE | ID: mdl-25566499

ABSTRACT

The Notch pathway is a well-established mediator of cell-cell communication that plays a critical role in stem cell survival, self-renewal, cell fate decisions, tumorigenesis, invasion, metastasis, and drug resistance in a variety of cancers. An interesting form of crosstalk exists between the Notch receptor and the Epidermal Growth Factor Receptor Tyrosine Kinase family, which consists of HER-1, -2, -3, and -4. Overexpression of HER and/or Notch occurs in several human cancers including brain, lung, breast, ovary, and skin making them potent oncogenes capable of advancing malignant disease. Continued assessment of interplay between these two critical signaling networks uncovers new insight into mechanisms used by HER-driven cancer cells to exploit Notch as a compensatory pathway. The compensatory Notch pathway maintains HER-induced downstream signals transmitted to pathways such as Mitogen Activated Protein Kinase and Phosphatidylinositol 3-Kinase (PI3K), thereby allowing cancer cells to survive molecular targeted therapies, undergo epithelial to mesenchymal transitioning, and increase cellular invasion. Uncovering the critical crosstalk between the HER and Notch pathways can lead to improved screening for the expression of these oncogenes enabling patients to optimize their personal treatment options and predict potential treatment resistance. This review will focus on the current state of crosstalk between the HER and Notch receptors and the effectiveness of current therapies targeting HER-driven cancers.

6.
Onco Targets Ther ; 6: 943-55, 2013.
Article in English | MEDLINE | ID: mdl-23901284

ABSTRACT

The numerous processes involved in the etiology of breast cancer such as cell survival, metabolism, proliferation, differentiation, and angiogenesis are currently being elucidated. However, underlying mechanisms that drive breast cancer progression and drug resistance are still poorly understood. As we discuss here in detail, the Notch signaling pathway is an important regulatory component of normal breast development, cell fate of normal breast stem cells, and proliferation and survival of breast cancer initiating cells. Notch exerts a wide range of critical effects through a canonical pathway where it is expressed as a type I membrane precursor heterodimer followed by at least two subsequent cleavages induced by ligand engagement to ultimately release an intracellular form to function as a transcriptional activator. Notch and its ligands are overexpressed in breast cancer, and one method of effectively blocking Notch activity is preventing its cleavage at the cell surface with γ-secretase inhibitors. In the context of Notch signaling, the application of clinically relevant anti-Notch drugs in treatment regimens may contribute to novel therapeutic interventions and promote more effective clinical response in women with breast cancer.

7.
Development ; 136(18): 3131-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19700617

ABSTRACT

Trimethylation of histone H3 lysine 27 (H3K27me3) by Polycomb repressive complex 2 (PRC2) is essential for transcriptional silencing of Polycomb target genes, whereas acetylation of H3K27 (H3K27ac) has recently been shown to be associated with many active mammalian genes. The Trithorax protein (TRX), which associates with the histone acetyltransferase CBP, is required for maintenance of transcriptionally active states and antagonizes Polycomb silencing, although the mechanism underlying this antagonism is unknown. Here we show that H3K27 is specifically acetylated by Drosophila CBP and its deacetylation involves RPD3. H3K27ac is present at high levels in early embryos and declines after 4 hours as H3K27me3 increases. Knockdown of E(Z) decreases H3K27me3 and increases H3K27ac in bulk histones and at the promoter of the repressed Polycomb target gene abd-A, suggesting that these indeed constitute alternative modifications at some H3K27 sites. Moderate overexpression of CBP in vivo causes a global increase in H3K27ac and a decrease in H3K27me3, and strongly enhances Polycomb mutant phenotypes. We also show that TRX is required for H3K27 acetylation. TRX overexpression also causes an increase in H3K27ac and a concomitant decrease in H3K27me3 and leads to defects in Polycomb silencing. Chromatin immunoprecipitation coupled with DNA microarray (ChIP-chip) analysis reveals that H3K27ac and H3K27me3 are mutually exclusive and that H3K27ac and H3K4me3 signals coincide at most sites. We propose that TRX-dependent acetylation of H3K27 by CBP prevents H3K27me3 at Polycomb target genes and constitutes a key part of the molecular mechanism by which TRX antagonizes or prevents Polycomb silencing.


Subject(s)
CREB-Binding Protein/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Gene Silencing , Histones/metabolism , Lysine/metabolism , Acetylation , Animals , Animals, Genetically Modified , CREB-Binding Protein/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryonic Development/physiology , Gene Expression Regulation, Developmental , Histones/genetics , Humans , Polycomb Repressive Complex 1 , Promoter Regions, Genetic
8.
Cancer Res ; 65(11): 4554-61, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15930272

ABSTRACT

Indolent non-Hodgkin lymphomas are characterized by a prolonged phase that is typically followed by a clinical progression associated with an accelerated clinical course and short survival time. Previous studies have not identified a consistent cytogenetic or molecular abnormality associated with transformation. The development of a transformed phenotype, evolving from the original low-grade component, most likely depends on multiple genetic events, including the activation of synergistic dominant oncogenes and a loss of tumor suppressor gene functions. Complex karyotypes and relatively bad chromosome morphology are typical of transformed non-Hodgkin lymphomas, rendering complete cytogenetic analysis difficult. Here, we report the use of transformed non-Hodgkin lymphoma cell lines and primary samples to identify the involvement of the silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) gene that maps at chromosome 12q24 in transformed non-Hodgkin lymphomas. We also show that down-regulation of SMRT in the immortalized "Weinberg's model" cell lines induces transformation of the cells. Assessment of cDNA array profiles should further help us to design a working model for SMRT involvement in non-Hodgkin lymphoma transformation as a novel, nonclassical tumor suppressor.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Genes, Tumor Suppressor , Lymphoma, Non-Hodgkin/genetics , Repressor Proteins/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromosomes, Human, Pair 12/genetics , DNA-Binding Proteins/biosynthesis , Down-Regulation , Gene Deletion , Gene Rearrangement , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 2 , Oligonucleotide Array Sequence Analysis , Repressor Proteins/biosynthesis , Transcriptional Activation
9.
J Cell Physiol ; 199(3): 418-33, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15095289

ABSTRACT

Notch signaling is a potential therapeutic target for various solid and hematopoietic malignancies. We have recently shown that downregulation of Notch-1 expression has significant anti-neoplastic activity in pre-clinical models. However, the mechanisms through which Notch modulation may affect cell fate in cancer remain poorly understood. We had previously shown that Notch-1 prevents apoptosis and is necessary for pharmacologically induced differentiation in murine erythroleukemia (MEL) cells. We investigated the mechanisms of these effects using three experimental strategies: (1) MEL cells stably transfected with antisense Notch-1 or constitutively active Notch-1, (2) activation of Notch-1 by a cell-associated ligand, and (d3) activation of Notch-1 by a soluble peptide ligand. We show that: (1) downregulation of Notch-1 sensitizes MEL cells to apoptosis induced by a Ca(2+) influx or anti-neoplastic drugs; (2) Notch-1 downregulation induces phosphorylation of c-Jun N-terminal kinase (JNK) while constitutive activation of Notch-1 or prolonged exposure to a soluble Notch ligand abolishes it; (3) Notch-1 has dose- and time-dependent effects on the levels of apoptotic inhibitor Bcl-x(L) and cell cycle regulators p21(cip1/waf1), p27(kip1), and Rb; and (4) Notch-1 activation by a cell-associated ligand is accompanied by rapid and transient induction of NF-kappaB DNA-binding activity. The relative effects of Notch-1 signaling on these pathways depend on the levels of Notch-1 expression, the mechanism of activation, and the timing of activation. The relevance of these findings to the role of Notch signaling in differentiation and cancer are discussed.


Subject(s)
Apoptosis/physiology , Cell Cycle/physiology , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Transcription Factors , Animals , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/metabolism , Down-Regulation , Electrophoretic Mobility Shift Assay , JNK Mitogen-Activated Protein Kinases , Leukemia, Erythroblastic, Acute/physiopathology , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, Notch1 , Transfection , Tumor Suppressor Proteins/metabolism , bcl-X Protein
10.
J Cell Physiol ; 194(3): 356-62, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12548555

ABSTRACT

Notch receptor signaling has been implicated in cellular transformation. Notch-1 receptor expression is increased during the progression from cervical intraepithelial lesions (CIN) to invasive cervical carcinoma. Moreover, the main cellular localization of Notch-1 protein changes from cytoplasmic to nuclear with the transition from CIN III to microinvasive carcinoma. Since the E6 and E7 proteins encoded by human papilloma virus (HPV) are a causative agent of cervical carcinoma, this study determined whether E6 and E7 protein expression causes the observed upregulation in Notch-1 expression. Mouse and human primary cell lines were transfected with HPV16 E6 and E7 and Notch-1 expression and activity were analyzed. We show that Notch-1 expression and activity are upregulated by E6 and E7 independently. This was due to both transcriptional and post-transcriptional mechanisms. A protein involved in Notch processing, Presenilin-1 (PS-1), was also upregulated by E6 and E7. In the presence of E6 and E7, Notch-1 protein expression is localized in the cytoplasm. Downregulation of Notch-1 expression in a human cervical carcinoma cell line expressing E6/E7 caused striking inhibition of proliferation in vitro and tumorigenicity in vivo. These data suggest that E6- and E7-mediated upregulation of Notch signaling may contribute to disruption of regular cell growth in cervical cancer.


Subject(s)
Avian Proteins , Cell Transformation, Neoplastic/metabolism , Membrane Proteins/genetics , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Oncogene Proteins , Receptors, Cell Surface , Repressor Proteins , Transcription Factors , Viral Proteins , Animals , Cell Division/physiology , Cell Line , Cell Survival/physiology , Cervix Uteri/cytology , Epithelial Cells/cytology , Epithelial Cells/physiology , Female , Forkhead Transcription Factors , Gene Expression Regulation, Neoplastic/physiology , Mice , Papillomavirus E7 Proteins , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor, Notch1 , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/physiopathology , Uterine Cervical Neoplasms/virology
11.
Nat Med ; 8(9): 979-86, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12185362

ABSTRACT

Truncated Notch receptors have transforming activity in vitro and in vivo. However, the role of wild-type Notch signaling in neoplastic transformation remains unclear. Ras signaling is deregulated in a large fraction of human malignancies and is a major target for the development of novel cancer treatments. We show that oncogenic Ras activates Notch signaling and that wild-type Notch-1 is necessary to maintain the neoplastic phenotype in Ras-transformed human cells in vitro and in vivo. Oncogenic Ras increases levels and activity of the intracellular form of wild-type Notch-1, and upregulates Notch ligand Delta-1 and also presenilin-1, a protein involved in Notch processing, through a p38-mediated pathway. These observations place Notch signaling among key downstream effectors of oncogenic Ras and suggest that it might be a novel therapeutic target.


Subject(s)
Cell Transformation, Neoplastic/genetics , Membrane Proteins/metabolism , Receptors, Cell Surface , Signal Transduction/genetics , Transcription Factors , ras Proteins/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cells, Cultured , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mice , Mice, SCID , Mitogen-Activated Protein Kinases/metabolism , Phenotype , Presenilin-1 , Receptor, Notch1 , Up-Regulation , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , p38 Mitogen-Activated Protein Kinases , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...