Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 481: 116753, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37951547

ABSTRACT

Exposure to nickel, an environmental respiratory toxicant, is associated with lung diseases including asthma, pulmonary fibrosis, bronchitis and cancers. Our previous studies have shown that a majority of the nickel-induced transcriptional changes are persistent and do not reverse even after the termination of exposure. This suggested transcriptional memory, wherein the cell 'remembers' past nickel exposure. Transcriptional memory, due to which the cells respond more robustly to a previously encountered stimulus has been identified in a number of organisms. Therefore, transcriptional memory has been described as an adaptive mechanism. However, transcriptional memory caused by environmental toxicant exposures has not been well investigated. Moreover, how the transcriptional memory caused by an environmental toxicant might influence the outcome of exposure to a second toxicant has not been explored. In this study, we investigated whether nickel-induced transcriptional memory influences the outcome of the cell's response to a second respiratory toxicant, nicotine. Nicotine, an addictive compound in tobacco, is associated with the development of chronic lung diseases including chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis. Our results show that nicotine exposure upregulated a subset of genes only in the cells previously exposed to nickel. Furthermore, our analyses indicate robust activation of interferon (IFN) signaling in these cells. IFN signaling is a driver of inflammation, which is associated with many chronic lung diseases. Therefore, our results suggest that nicotine exposure of lung cells that retain the transcriptional memory of previous nickel exposure could result in increased susceptibility to developing chronic inflammatory lung diseases.


Subject(s)
Nickel , Pulmonary Fibrosis , Humans , Nickel/toxicity , Nicotine/toxicity , Pulmonary Fibrosis/pathology , Lung/pathology , Epithelial Cells , Interferons
2.
Mol Carcinog ; 61(1): 99-110, 2022 01.
Article in English | MEDLINE | ID: mdl-34727382

ABSTRACT

E-cadherin plays a central role in the stability of epithelial tissues by facilitating cell-cell adhesion. Loss of E-cadherin expression is a hallmark of epithelial-mesenchymal transition (EMT), a major event in the pathogenesis of several lung diseases. Our earlier studies showed that nickel, a ubiquitous environmental toxicant, induced EMT by persistently downregulating E-cadherin expression in human lung epithelial cells and that the EMT remained irreversible postexposure. However, the molecular basis of persistent E-cadherin downregulation by nickel exposure is not understood. Here, our studies show that the binding of transcription factor Sp1 to the promoter of E-cadherin encoding gene, CDH1, is essential for its expression. Nickel exposure caused a loss of Sp1 binding at the CDH1 promoter, resulting in its downregulation and EMT induction. Loss of Sp1 binding at the CDH1 promoter was associated with an increase in the binding of ZEB1 adjacent to the Sp1 binding site. ZEB1, an EMT master regulator persistently upregulated by nickel exposure, is a negative regulator of CDH1. CRISPR-Cas9-mediated knockout of ZEB1 restored Sp1 binding at the CDH1 promoter. Furthermore, ZEB1 knockout rescued E-cadherin expression and re-established the epithelial phenotype. Since EMT is associated with a number of nickel-exposure-associated chronic inflammatory lung diseases including asthma, fibrosis and cancer and metastasis, our findings provide new insights into the mechanisms associated with nickel pathogenesis.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Lung/cytology , Nickel/toxicity , Sp1 Transcription Factor/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Cell Line , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Gene Knockout Techniques , Humans , Lung/drug effects , Lung/metabolism , MCF-7 Cells , Promoter Regions, Genetic
3.
Semin Cancer Biol ; 76: 99-109, 2021 11.
Article in English | MEDLINE | ID: mdl-34058338

ABSTRACT

Nickel compounds are environmental toxicants, prevalent in the atmosphere due to their widespread use in several industrial processes, extensive consumption of nickel containing products, as well as burning of fossil fuels. Exposure to nickel is associated with a multitude of chronic inflammatory lung diseases including asthma, chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis. In addition, nickel exposure is implicated in the development of nasal and lung cancers. Interestingly, a common pathogenic mechanism underlying the development of diseases associated with nickel exposure is epithelial-mesenchymal transition (EMT). EMT is a process by which the epithelial cells lose their junctions and polarity and acquire mesenchymal traits, including increased ability to migrate and invade. EMT is a normal and essential physiological process involved in differentiation, development and wound healing. However, EMT also contributes to a number of pathological conditions, including fibrosis, cancer and metastasis. Growing evidence suggest that EMT induction could be an important outcome of nickel exposure. In this review, we discuss the role of EMT in nickel-induced lung diseases and the mechanisms associated with EMT induction by nickel exposure.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Lung Diseases/chemically induced , Lung Diseases/pathology , Nickel/adverse effects , Animals , Humans
4.
Epigenetics Chromatin ; 12(1): 75, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31856895

ABSTRACT

BACKGROUND: Nickel is an occupational and environmental toxicant associated with a number of diseases in humans including pulmonary fibrosis, bronchitis and lung and nasal cancers. Our earlier studies showed that the nickel-exposure-induced genome-wide transcriptional changes, which persist even after the termination of exposure may underlie nickel pathogenesis. However, the mechanisms that drive nickel-induced persistent changes to the transcriptome remain elusive. RESULTS: To elucidate the mechanisms that underlie nickel-induced long-term transcriptional changes, in this study, we examined the transcriptome and the epigenome of human lung epithelial cells during nickel exposure and after the termination of exposure. We identified two categories of persistently differentially expressed genes: (i) the genes that were differentially expressed during nickel exposure; and (ii) the genes that were differentially expressed only after the termination of exposure. Interestingly, > 85% of the nickel-induced gene expression changes occurred only after the termination of exposure. We also found extensive genome-wide alterations to the activating histone modification, H3K4me3, after the termination of nickel exposure, which coincided with the post-exposure gene expression changes. In addition, we found significant post-exposure alterations to the repressive histone modification, H3K27me3. CONCLUSION: Our results suggest that while modest first wave of transcriptional changes occurred during nickel exposure, extensive transcriptional changes occurred during a second wave of transcription for which removal of nickel ions was essential. By uncovering a new category of transcriptional and epigenetic changes, which occur only after the termination of exposure, this study provides a novel understanding of the long-term deleterious consequences of nickel exposure on human health.


Subject(s)
Epigenesis, Genetic/drug effects , Nickel/toxicity , Cell Line , DNA Methylation/drug effects , Down-Regulation/drug effects , Histones/metabolism , Humans , Up-Regulation/drug effects
5.
Mutat Res Rev Mutat Res ; 780: 61-68, 2019.
Article in English | MEDLINE | ID: mdl-31395350

ABSTRACT

CCCTC-binding factor (CTCF) is a highly conserved, ubiquitously expressed zinc finger protein. CTCF is a multifunctional protein, associated with a number of vital cellular processes such as transcriptional activation, repression, insulation, imprinting and genome organization. Emerging evidence indicates that CTCF is also involved in DNA damage response. In this review, we focus on the newly identified role of CTCF in facilitating DNA double-strand break repair. Due to the large number of cellular processes in which CTCF is involved, factors that functionally affect CTCF could have serious implications on genomic stability. It is becoming increasingly clear that exposure to environmental toxicants could have adverse effects on CTCF functions. Here we discuss the various ways that environmental toxicants could impact CTCF functions and the potential consequences on DNA damage response.


Subject(s)
CCCTC-Binding Factor/genetics , DNA Damage/genetics , DNA Repair/genetics , Animals , DNA Breaks, Double-Stranded , Genome/genetics , Genomic Instability/genetics , Humans
6.
Toxicol Appl Pharmacol ; 373: 1-9, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30998937

ABSTRACT

Cadmium (Cd) is a known human lung carcinogen. In addition, Cd exposure is associated with several lung diseases including emphysema, chronic obstructive pulmonary disease (COPD), asthma and fibrosis. Although earlier studies have identified several processes dysregulated by Cd exposure, the underlying mechanisms remain unclear. Here, we examined the transcriptome of lung epithelial cells exposed to Cd to understand the molecular basis of Cd-induced diseases. Computational analysis of the transcriptome predicted a significant number of Cd-upregulated genes to be targets of miR-30 family miRNAs. Experimental validation showed downregulation of all the miR-30 family members in Cd exposed cells. We found SNAIL, an EMT master regulator, to be the most upregulated among the miR-30 targets. Furthermore, we found decrease in the levels of epithelial marker E- cadherin (CDH1) and increase in the levels of mesenchymal markers, ZEB1 and vimentin. This suggested induction of EMT in Cd exposed cells. Luciferase reporter assays showed that miR-30 repressed SNAIL by directly targeting its 3' UTR. Over expression of miR-30e and transfection of miR-30e mimics reduced Cd-induced SNAIL upregulation. Our results suggest that miR-30 negatively regulates SNAIL in lung epithelial cells and that Cd-induced downregulation of miR-30 relieves this repression, resulting in SNAIL upregulation and EMT induction. EMT plays a major role in many diseases associated with Cd exposure including fibrosis, COPD, and cancer and metastasis. Therefore, our identification of miR-30 downregulation in Cd exposed cells and the consequent activation of SNAIL provides important mechanistic insights into lung diseases associated with Cd exposure.


Subject(s)
Cadmium Chloride/toxicity , Epithelial Cells/drug effects , Lung/drug effects , MicroRNAs/metabolism , Snail Family Transcription Factors/metabolism , 3' Untranslated Regions , Antigens, CD/genetics , Antigens, CD/metabolism , Binding Sites , Cadherins/genetics , Cadherins/metabolism , Cell Line , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung/metabolism , Lung/pathology , MicroRNAs/genetics , Signal Transduction , Snail Family Transcription Factors/genetics , Transcriptome , Up-Regulation , Vimentin/genetics , Vimentin/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
7.
Mol Carcinog ; 57(6): 794-806, 2018 06.
Article in English | MEDLINE | ID: mdl-29528143

ABSTRACT

Nickel (Ni) is an environmental and occupational carcinogen, and exposure to Ni is associated with lung and nasal cancers in humans. Furthermore, Ni exposure is implicated in several lung diseases including chronic inflammatory airway diseases, asthma, and fibrosis. However, the mutagenic potential of Ni is low and does not correlate with its potent toxicity and carcinogenicity. Therefore, mechanisms underlying Ni exposure-associated diseases remain poorly understood. Since the health risks of environmental exposures often continue post exposure, understanding the exposure effects that persist after the termination of exposure could provide mechanistic insights into diseases. By examining the persistent effects of Ni exposure, we report that Ni induces epithelial-mesenchymal transition (EMT) and that the mesenchymal phenotype remains irreversible even after the termination of exposure. Ni-induced EMT was dependent on the irreversible upregulation of ZEB1, an EMT master regulator, via resolution of its promoter bivalency. ZEB1, upon activation, downregulated its repressors as well as the cell-cell adhesion molecule, E-cadherin, resulting in the cells undergoing EMT and switching to persistent mesenchymal status. ZEB1 depletion in cells exposed to Ni attenuated Ni-induced EMT. Moreover, Ni exposure did not induce EMT in ZEB1-depleted cells. Activation of EMT, during which the epithelial cells lose cell-cell adhesion and become migratory and invasive, plays a major role in asthma, fibrosis, and cancer and metastasis, lung diseases associated with Ni exposure. Therefore, our finding of irreversible epigenetic activation of ZEB1 by Ni exposure and the acquisition of persistent mesenchymal phenotype would have important implications in understanding Ni-induced diseases.


Subject(s)
Epigenesis, Genetic/drug effects , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Nickel/pharmacology , Zinc Finger E-box-Binding Homeobox 1/genetics , Cell Hypoxia , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Humans , Phenotype , RNA Interference , Zinc Finger E-box-Binding Homeobox 1/metabolism
8.
Toxicol Res (Camb) ; 6(3): 312-323, 2017 May 01.
Article in English | MEDLINE | ID: mdl-29057067

ABSTRACT

Mercury (Hg) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) are major environmental contaminants that commonly co-occur in the environment. Both Hg and TCDD are associated with a number of human diseases including cancers. While the individual toxicological effects of Hg and TCDD have been extensively investigated, studies on co-exposure are limited to a few genes and pathways. Therefore, a significant knowledge gap exists in the understanding of the deleterious effects of co-exposure to Hg and TCDD. Due to the prevalence of Hg and TCDD co-contamination in the environment and the major human health hazards they pose, it is important to obtain a fuller understanding of genome-wide effects of Hg and TCDD co-exposure. In this study, by performing a comprehensive transcriptomic analysis of human bronchial epithelial cells (BEAS-2B) exposed to Hg and TCDD individually and in combination, we have uncovered a subset of genes with altered expression only in the co-exposed cells. We also identified the additive as well as antagonistic effects of Hg and TCDD on gene expression. Moreover, we found that co-exposure impacted several biological and disease processes not affected by Hg or TCDD individually. Our studies show that the consequences of Hg and TCDD co-exposure on the transcriptional program and biological processes could be substantially different from single exposures, thus providing new insights into the co-exposure-specific pathogenic processes.

9.
J Cell Physiol ; 231(7): 1611-20, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26588041

ABSTRACT

Oxygen levels range from 2% to 9% in vivo. Atmospheric O2 levels (21%) are known to induce cell proliferation defects and cellular senescence in primary cell cultures. However, the mechanistic basis of the deleterious effects of higher O2 levels is not fully understood. On the other hand, immortalized cells including cancer cell lines, which evade cellular senescence are normally cultured at 21% O2 and the effects of higher O2 on these cells are understudied. Here, we addressed this problem by culturing immortalized human bronchial epithelial (BEAS-2B) cells at ambient atmospheric, 21% O2 and lower, 10% O2. Our results show increased inflammatory response at 21% O2 but not at 10% O2. We found higher RelA binding at the NF-κB1/RelA target gene promoters as well as upregulation of several pro-inflammatory cytokines in cells cultured at 21% O2. RelA knockdown prevented the upregulation of the pro-inflammatory cytokines at 21% O2, suggesting NF-κB1/RelA as a major mediator of inflammatory response in cells cultured at 21% O2. Interestingly, unlike the 21% O2 cultured cells, exposure of 10% O2 cultured cells to H2O2 did not elicit inflammatory response, suggesting increased ability to tolerate oxidative stress in cells cultured at lower O2 levels.


Subject(s)
Inflammation/metabolism , Lung/metabolism , Oxygen/metabolism , Transcription Factor RelA/genetics , Cell Proliferation , Cellular Senescence , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Knockdown Techniques , Humans , Inflammation/pathology , Lung/pathology , Promoter Regions, Genetic , Transcription Factor RelA/metabolism
10.
Proc Natl Acad Sci U S A ; 111(40): 14631-6, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25246589

ABSTRACT

Investigations into the genomic landscape of histone modifications in heterochromatic regions have revealed histone H3 lysine 9 dimethylation (H3K9me2) to be important for differentiation and maintaining cell identity. H3K9me2 is associated with gene silencing and is organized into large repressive domains that exist in close proximity to active genes, indicating the importance of maintenance of proper domain structure. Here we show that nickel, a nonmutagenic environmental carcinogen, disrupted H3K9me2 domains, resulting in the spreading of H3K9me2 into active regions, which was associated with gene silencing. We found weak CCCTC-binding factor (CTCF)-binding sites and reduced CTCF binding at the Ni-disrupted H3K9me2 domain boundaries, suggesting a loss of CTCF-mediated insulation function as a potential reason for domain disruption and spreading. We furthermore show that euchromatin islands, local regions of active chromatin within large H3K9me2 domains, can protect genes from H3K9me2-spreading-associated gene silencing. These results have major implications in understanding H3K9me2 dynamics and the consequences of chromatin domain disruption during pathogenesis.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic/drug effects , Epithelial Cells/drug effects , Nickel/pharmacology , Acetylation , Amino Acid Sequence , Binding Sites/genetics , Blotting, Western , Bronchi/cytology , CCCTC-Binding Factor , Cell Line , Chromatin/genetics , Epigenesis, Genetic/genetics , Epithelial Cells/metabolism , Gene Expression Profiling , Genome, Human/genetics , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism
11.
PLoS One ; 7(5): e36365, 2012.
Article in English | MEDLINE | ID: mdl-22570707

ABSTRACT

Polycomb group (PcG) proteins are key chromatin regulators implicated in multiple processes including embryonic development, tissue homeostasis, genomic imprinting, X-chromosome inactivation, and germ cell differentiation. The PcG proteins recognize target genomic loci through cis DNA sequences known as Polycomb Response Elements (PREs), which are well characterized in Drosophila. However, mammalian PREs have been elusive until two groups reported putative mammalian PREs recently. Consistent with the existence of mammalian PREs, here we report the identification and characterization of a potential PRE from human T cells. The putative human PRE has enriched binding of PcG proteins, and such binding is dependent on a key PcG component SUZ12. We demonstrate that the putative human PRE carries both genetic and molecular features of Drosophila PRE in transgenic flies, implying that not only the trans PcG proteins but also certain features of the cis PREs are conserved between mammals and Drosophila.


Subject(s)
Drosophila/genetics , Polycomb-Group Proteins/metabolism , Response Elements , Animals , Animals, Genetically Modified , Binding Sites , Cell Cycle Proteins/metabolism , Cell Line , Drosophila/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Polycomb-Group Proteins/chemistry , Transcription, Genetic
12.
BMC Cell Biol ; 11: 35, 2010 May 24.
Article in English | MEDLINE | ID: mdl-20497571

ABSTRACT

BACKGROUND: Connective Tissue Growth Factor (CTGF/CCN2), a known matrix-associated protein, is required for the lactogenic differentiation of mouse mammary epithelial cells. An HC11 mammary epithelial cell line expressing CTGF/CCN2 was constructed to dissect the cellular responses to CTGF/CCN2 that contribute to this differentiation program. RESULTS: Tetracycline-regulated expression of CTGF/CCN2 in HC11 cells enhanced multiple markers of lactogenic differentiation including beta-casein transcription and mammosphere formation. In a separate measure of mammary differentiation the addition of CTGF/CCN2 to cultures of MCF10A cells increased the development of acini in vitro. In HC11 cells the elevated levels of CTGF/CCN2 diminished the requirement for extracellular matrix proteins in the activation of beta-casein transcription, indicating that CTGF/CCN2 contributed to lactogenic differentiation through the regulation of matrix dependent cell adhesion. CTGF/CCN2 expression in HC11 cells increased expression of extracellular matrix proteins and integrins, enhanced the formation of focal adhesion complexes, and increased survival signaling. In addition, HC11 cells adhered to immobilized CTGF/CCN2 and this was inhibited by function-blocking antibodies to the integrins alpha6 and beta1, and to a lesser degree by antibody to beta3 integrin. CONCLUSIONS: CTGF/CCN2 expression in HC11 cells led to an increase in multiple markers of lactogenic differentiation. The mechanisms by which CTGF/CCN2 contributed to lactogenic differentiation include direct binding of CTGF/CCN2 to integrin complexes and CTGF/CCN2-induced matrix protein expression resulting in elevated integrin functionality.


Subject(s)
Antigens, Differentiation/biosynthesis , Caseins/biosynthesis , Connective Tissue Growth Factor/metabolism , Epithelial Cells/metabolism , Integrin alpha6beta1/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antigens, Differentiation/genetics , Caseins/genetics , Cell Differentiation , Cell Line, Tumor , Cloning, Molecular , Connective Tissue Growth Factor/genetics , Epithelial Cells/drug effects , Epithelial Cells/pathology , Female , Focal Adhesions/drug effects , Integrin alpha6beta1/immunology , Mammary Glands, Animal/pathology , Mice , Signal Transduction/drug effects , Transgenes/genetics
13.
J Cell Physiol ; 214(1): 38-46, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17541935

ABSTRACT

The response of mammary epithelial cells to basement membrane and stroma induced signals contributes to the degree of differentiation in this tissue. The studies reported here indicate that connective tissue growth factor (CTGF) is highly elevated during lactogenic differentiation of the HC11 mouse mammary epithelial cell line. In addition, CTGF is expressed in the mouse mammary gland during pregnancy and lactation and it is expressed in primary mammary epithelial cell cultures established from pregnant mice. In HC11 cells CTGF is transcriptionally regulated by dexamethasone, but not by estrogen or progesterone, and CTGF expression is not dependent on TGFbeta. CTGF contributes to and is required for lactogenic differentiation of HC11 cells, as demonstrated by increased differentiation following expression of plasmid-encoded CTGF and decreased differentiation following depletion of endogenous CTGF with siRNA. Moreover, HC11 mouse mammary epithelial cells infected with an adenoviral vector encoding CTGF exhibit increased lactogenic differentiation. Plasmid vector-induced elevation of CTGF levels also increased the level of beta1 integrin in HC11 cells. Because the production of stromal factors is an important component of differentiation in mammary epithelial cells, the regulation of CTGF by glucocorticoids may play a critical role in this aspect of the control of differentiation. The studies reported here provide important information on the role of CTGF in mammary epithelial cell differentiation.


Subject(s)
Cell Differentiation/physiology , Dexamethasone/pharmacology , Epithelial Cells/metabolism , Glucocorticoids/pharmacology , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Adenoviridae/genetics , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Line , Connective Tissue Growth Factor , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Luciferases/analysis , Luciferases/metabolism , Mammary Glands, Animal/cytology , Mice , Mice, Inbred BALB C , Pregnancy , Prolactin/pharmacology
14.
BMC Cell Biol ; 7: 34, 2006 Sep 19.
Article in English | MEDLINE | ID: mdl-16984645

ABSTRACT

BACKGROUND: HC11 mouse mammary epithelial cells differentiate in response to lactogenic hormone resulting in expression of milk proteins including beta-casein. Previous studies have shown that epidermal growth factor (EGF) blocks differentiation not only through activation of the Ras/Mek/Erk pathway but also implicated phosphatidylinositol-3-kinase (PI-3-kinase) signaling. The current study analyzes the mechanism of the PI-3-kinase pathway in an EGF-induced block of HC11 lactogenic differentiation. RESULTS: HC11 and HC11-luci cells, which contain luciferase gene under the control of a beta-casein promotor, were treated with specific chemical inhibitors of signal transduction pathways or transiently infected/transfected with vectors encoding dominant negative-Akt (DN-Akt) or conditionally active-Akt (CA-Akt). The expression of CA-Akt inhibited lactogenic differentiation of HC11 cells, and the infection with DN-Akt adenovirus enhanced beta-casein transcription and rescued beta-casein promotor-regulated luciferase activity in the presence of EGF. Treatment of cells with Rapamycin, an inhibitor of mTOR, blocked the effects of EGF on beta-casein promotor driven luciferase activity as effectively as PI-3-kinase inhibitors. While expression of CA-Akt caused a constitutive activation of p70S6 kinase (p70S6K) in HC11 cells, the inhibition of either PI-3-kinase or mTOR abolished the activation of p70S6K by EGF. The activation of p70S6K by insulin or EGF resulted in the phosphorylation of ribosomal protein S6 (RPS6), elongation initiation factor 4E (elF4E) and 4E binding protein1 (4E-BP1). But lower levels of PI-3-K and mTOR inhibitors were required to block insulin-induced phosphorylation of RPS6 than EGF-induced phosphorylation, and insulin-induced phosphorylation of elF4E and 4E-BP1 was not completely mTOR dependent suggesting some diversity of signaling for EGF and insulin. In HC11 cells undergoing lactogenic differentiation the phosphorylation of p70S6K completely diminished by 12 hours, and this was partly attributable to dexamethasone, a component of lactogenic hormone mix. However, p70S6K phosphorylation persisted in the presence of lactogenic hormone and EGF, but the activation could be blocked by a PI-3-kinase inhibitor. CONCLUSION: PI-3-kinase signaling contributes to the EGF block of lactogenic differentiation via Akt and p70S6K. The EGF-induced activation of PI-3-kinase-Akt-mTOR regulates phosphorylation of molecules including ribosomal protein S6, eIF4E and 4E-BP1 that influence translational control in HC11 cells undergoing lactogenic differentiation.


Subject(s)
Epidermal Growth Factor/pharmacology , Protein Kinases/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/physiology , Lactose/metabolism , Mice , Milk Proteins/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinases/physiology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...