Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
BMB Rep ; 55(12): 633-638, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36284441

ABSTRACT

Liver regeneration is a well-known systemic homeostatic phenomenon. The N6-methyladenosine (m6A) modification pathway has been associated with liver regeneration and hepatocellular carcinoma. m6A methyltransferases, such as methyltransferase 3 (METTL3) and methyltransferase 14 (METTL14), are involved in the hepatocyte-specific-regenerative pathway. To illustrate the role of METTL14, secreted from non-parenchymal liver cells, in the initiation phase of liver regeneration, we performed 70% partial hepatectomy (PH) in Mettl14 heterozygous (HET) and wild-type (WT) mice. Next, we analyzed the ratio of liver weight to body weight and the expression of mitogenic stimulators derived from non-parenchymal liver cells. Furthermore, we evaluated the expression of cell cycle-related genes and the hepatocyte proliferation rate via MKI67-immunostaining. During regeneration after PH, the weight ratio was lower in Mettl14 HET mice compared to WT mice. The expressions of hepatocyte growth factor (HGF) and tumor necrosis factor (TNF)-α, mitogens derived from non-parenchymal liver cells that stimulate the cell cycle, as well as the expressions of cyclin B1 and D1, which regulate the cell cycle, and the number of MKI67-positive cells, which indicate proliferative hepatocyte in the late G1-M phase, were significantly reduced in Mettl14 HET mice 72 h after PH. Our findings demonstrate that global Mettl14 mutation may interrupt the homeostasis of liver regeneration after an acute injury like PH by restraining certain mitogens, such as HGF and TNF-α, derived from sinusoidal endothelial cells, stellate cells, and Kupffer cells. These results provide new insights into the role of METTL14 in the clinical treatment strategies of liver disease. [BMB Reports 2022; 55(12): 633-638].


Subject(s)
Liver Regeneration , Methyltransferases , Mitogens , Animals , Mice , Endothelial Cells , Hepatectomy , Liver/metabolism , Liver Regeneration/physiology , Mitogens/metabolism , Tumor Necrosis Factor-alpha/metabolism , Methyltransferases/genetics
3.
Aging Cell ; 20(3): e13332, 2021 03.
Article in English | MEDLINE | ID: mdl-33709472

ABSTRACT

We previously demonstrated that ibrutinib modulates LPS-induced neuroinflammation in vitro and in vivo, but its effects on the pathology of Alzheimer's disease (AD) and cognitive function have not been investigated. Here, we investigated the effects of ibrutinib in two mouse models of AD. In 5xFAD mice, ibrutinib injection significantly reduced Aß plaque levels by promoting the non-amyloidogenic pathway of APP cleavage, decreased Aß-induced neuroinflammatory responses, and significantly downregulated phosphorylation of tau by reducing levels of phosphorylated cyclin-dependent kinase-5 (p-CDK5). Importantly, tau-mediated neuroinflammation and tau phosphorylation were also alleviated by ibrutinib injection in PS19 mice. In 5xFAD mice, ibrutinib improved long-term memory and dendritic spine number, whereas in PS19 mice, ibrutinib did not alter short- and long-term memory but promoted dendritic spinogenesis. Interestingly, the induction of dendritic spinogenesis by ibrutinib was dependent on the phosphorylation of phosphoinositide 3-kinase (PI3K). Overall, our results suggest that ibrutinib modulates AD-associated pathology and cognitive function and may be a potential therapy for AD.


Subject(s)
Adenine/analogs & derivatives , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Brain/pathology , Cognition , Inflammation/pathology , Piperidines/pharmacology , tau Proteins/metabolism , Adenine/pharmacology , Animals , Brain/drug effects , Brain/physiopathology , Cognition/drug effects , Cyclin-Dependent Kinase 5/metabolism , Cytokines/metabolism , Dendritic Spines/drug effects , Dendritic Spines/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Gliosis/complications , Inflammation Mediators/metabolism , Memory, Long-Term/drug effects , Mice, Transgenic , Neurogenesis/drug effects , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Phosphorylation/drug effects , Plaque, Amyloid/pathology
4.
Oncotarget ; 10(17): 1606-1624, 2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30899431

ABSTRACT

RARRES1, a retinoic acid regulated carboxypeptidase inhibitor associated with fatty acid metabolism, stem cell differentiation and tumorigenesis is among the most commonly methylated loci in multiple cancers but has no known mechanism of action. Here we show that RARRES1 interaction with cytoplasmic carboxypeptidase 2 (CCP2) inhibits tubulin deglutamylation, which in turn regulates the mitochondrial voltage dependent anion channel (VDAC1), mitochondrial membrane potential, AMPK activation, energy balance and metabolically reprograms cells and zebrafish to a more energetic and anabolic phenotype. Depletion of RARRES1 also increases expression of stem cell markers, promotes anoikis, anchorage independent growth and insensitivity to multiple apoptotic stimuli. As depletion of CCP2 or inhibition of VDAC1 reverses the effects of RARRES1 depletion on energy balance and cell survival we conclude that RARRES1 modulation of CCP2-modulated tubulin-mitochondrial VDAC1 interactions is a fundamental regulator of cancer and stem cell metabolism and survival.

5.
Cell Death Dis ; 9(9): 864, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154465

ABSTRACT

The role of Ahnak in obesity has been reported previously. Loss of Ahnak leads to decreased Bmp4/Smad1 signaling, resulting in the downregulation of adipocyte differentiation. However, the biological significance of Ahnak remains largely unknown. In this study, we demonstrate that Ahnak-mediated impaired adipogenesis results in decreased Bmpr1α transcriptional expression. To confirm this, Ahnak siRNA was used to knock-down Ahnak in C3H10T1/2 and primary stromal vascular fraction cells. Ahnak siRNA transfected cells showed suppression of Bmpr1α expression and decreased BMP4/ Bmpr1α signaling. The differential adipogenesis was further confirmed by knock-down of Bmpr1α in C3H10T1/2 cells, which resulted in reduced adipogenesis. Moreover, stable Ahnak knock-out C3H10T1/2 cells stably transfected with Ahnak CRISPR/Cas9 plasmid suppressed expression of Bmpr1α and prevented differentiation into adipocytes. Furthermore, we developed immortalized pre-adipocytes from wild-type or Ahnak Knock-out mice's stromal vascular fraction (SVF) to confirm the function of Ahnak in pre-adipocyte transition. Immortalized Ahnak knock-out SVF cells showed lower level of Bmpr1α expression, evidence by their impaired BMP4/Bmpr1α signaling. Upon adipogenic induction, immortalized Ahnak knock-out SVF cells exhibited a marked decrease in adipocyte differentiation compared with immortalized wild-type pre-adipocytes. Furthermore, over-expression of Bmpr1α restored the adipogenic activity of Ahnak knock-out C3H10T1/2 cells and immortalized Ahnak knock-out SVF cells. Our data reveal the missing link in Ahnak-mediated adipose tissue remodeling and suggest that precise regulation of Ahnak in adipose tissue might have a therapeutic advantage for metabolic disease treatment.


Subject(s)
Adipocytes/physiology , Bone Morphogenetic Protein Receptors, Type I/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Transcription, Genetic/genetics , Adipogenesis/genetics , Adipose Tissue/physiology , Animals , Cell Differentiation/genetics , Cell Line , Down-Regulation/genetics , Gene Expression Regulation/genetics , Male , Mice , Mice, Knockout , Obesity/genetics
6.
Nat Genet ; 48(12): 1517-1526, 2016 12.
Article in English | MEDLINE | ID: mdl-27776116

ABSTRACT

The functional rules for microRNA (miRNA) targeting remain controversial despite their biological importance because only a small fraction of distinct interactions, called site types, have been examined among an astronomical number of site types that can occur between miRNAs and their target mRNAs. To systematically discover functional site types and to evaluate the contradicting rules reported previously, we used large-scale transcriptome data and statistically examined whether each of approximately 2 billion site types is enriched in differentially downregulated mRNAs responding to overexpressed miRNAs. Accordingly, we identified seven non-canonical functional site types, most of which are novel, in addition to four canonical site types, while also removing numerous false positives reported by previous studies. Extensive experimental validation and significantly elevated 3' UTR sequence conservation indicate that these non-canonical site types may have biologically relevant roles. Our expanded catalog of functional site types suggests that the gene regulatory network controlled by miRNAs may be far more complex than currently understood.


Subject(s)
3' Untranslated Regions/genetics , Computational Biology/methods , Gene Expression Regulation , Gene Regulatory Networks , MicroRNAs/genetics , RNA, Messenger/metabolism , Binding Sites , Gene Expression Profiling , Humans , RNA, Messenger/genetics
7.
J Alzheimers Dis ; 51(2): 591-604, 2016.
Article in English | MEDLINE | ID: mdl-26890742

ABSTRACT

BACKGROUND: The accumulation of amyloid-ß (Aß) leads to the loss of dendritic spines and synapses, which is hypothesized to cause cognitive impairments in Alzheimer's disease (AD) patients. In our previous study, we demonstrated that a novel mercaptoacetamide-based class II histone deacetylase inhibitor (HDACI), known as W2, decreased Aß levels and improved learning and memory in mice. However, the underlying mechanism of this effect is unknown. OBJECTIVE: Because dendritic spine formation is associated with cognitive performance, here we investigated whether HDACI W2 regulates dendritic spine density and its molecular mechanism of action. METHODS: To examine the effect of HDACI W2 on dendritic spine density, we conducted morphological analysis of dendritic spines using GFP transfection and Golgi staining. In addition, to determine the molecular mechanism of W2 effects on spines, we measured the levels of mRNAs and proteins involved in the Ras signaling pathway using quantitative real-time PCR, immunocytochemistry, and western analysis. RESULTS: We found that HDACI W2 altered dendritic spine density and morphology in vitro and in vivo. Additionally, W2 increased the mRNA or protein levels of Ras GRF1 and phospho-ERK. Moreover, knockdown of RasGRF1 and inhibition of ERK activity prevented the W2-mediated spinogenesis in primary hippocampal neurons. CONCLUSION: Our Class II-selective HDACI W2 promotes the formation and growth of dendritic spines in a RasGRF1 and ERK dependent manner in primary hippocampal neurons.


Subject(s)
Acetamides/pharmacology , Dendritic Spines/drug effects , Histone Deacetylase Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , Thioglycolates/pharmacology , ras-GRF1/metabolism , Animals , Blotting, Western , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Dendritic Spines/enzymology , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/enzymology , Immunohistochemistry , MAP Kinase Signaling System/physiology , Mice, Inbred C57BL , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Transfection , ras-GRF1/genetics
8.
Biochim Biophys Acta ; 1862(2): 284-95, 2016 02.
Article in English | MEDLINE | ID: mdl-26675527

ABSTRACT

Our recent study demonstrated that an amyloid-ß binding molecule, BTA-EG4, increases dendritic spine number via Ras-mediated signaling. To potentially optimize the potency of the BTA compounds, we synthesized and evaluated an amyloid-ß binding analog of BTA-EG4 with increased solubility in aqueous solution, BTA-EG6. We initially examined the effects of BTA-EG6 on dendritic spine formation and found that BTA-EG6-treated primary hippocampal neurons had significantly increased dendritic spine number compared to control treatment. In addition, BTA-EG6 significantly increased the surface level of AMPA receptors. Upon investigation into the molecular mechanism by which BTA-EG6 promotes dendritic spine formation, we found that BTA-EG6 may exert its effects on spinogenesis via RasGRF1-ERK signaling, with potential involvement of other spinogenesis-related proteins such as Cdc42 and CDK5. Taken together, our data suggest that BTA-EG6 boosts spine and synapse number, which may have a beneficial effect of enhancing neuronal and synaptic function in the normal healthy brain.


Subject(s)
Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Dendritic Spines/drug effects , Signal Transduction/drug effects , ras Proteins/metabolism , ras-GRF1/metabolism , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Cells, Cultured , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Ethylene Glycol/chemistry , Ethylene Glycol/pharmacology , MAP Kinase Signaling System/drug effects , Rats, Sprague-Dawley , Receptors, AMPA/metabolism
9.
Neuron ; 84(1): 63-77, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25242217

ABSTRACT

Alzheimer's disease (AD) is an age-related neurological disorder characterized by synaptic loss and dementia. The low-density lipoprotein receptor-related protein 6 (LRP6) is an essential coreceptor for Wnt signaling, and its genetic variants have been linked to AD risk. Here we report that neuronal LRP6-mediated Wnt signaling is critical for synaptic function and cognition. Conditional deletion of Lrp6 gene in mouse forebrain neurons leads to age-dependent deficits in synaptic integrity and memory. Neuronal LRP6 deficiency in an amyloid mouse model also leads to exacerbated amyloid pathology due to increased APP processing to amyloid-ß. In humans, LRP6 and Wnt signaling are significantly downregulated in AD brains, likely by a mechanism that depends on amyloid-ß. Our results define a critical pathway in which decreased LRP6-mediated Wnt signaling, synaptic dysfunction, and elevated Aß synergistically accelerate AD progression and suggest that restoring LRP6-mediated Wnt signaling can be explored as a viable strategy for AD therapy.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/deficiency , Synapses/metabolism , Wnt Signaling Pathway/physiology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Cell Line, Tumor , Female , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Synapses/pathology
10.
Exp Neurol ; 252: 105-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24316432

ABSTRACT

We recently reported that the tetra(ethylene glycol) derivative of benzothiazole aniline, BTA-EG4, acts as an amyloid-binding small molecule that promotes dendritic spine density and cognitive function in wild-type mice. This raised the possibility that BTA-EG4 may benefit the functional decline seen in Alzheimer's disease (AD). In the present study, we directly tested whether BTA-EG4 improves dendritic spine density and cognitive function in a well-established mouse model of AD carrying mutations in APP, PS1 and tau (APPswe;PS1M146V;tauP301L, 3xTg AD mice). We found that daily injections of BTA-EG4 for 2 weeks improved dendritic spine density and cognitive function of 3xTg AD mice in an age-dependent manner. Specifically, BTA-EG4 promoted both dendritic spine density and morphology alterations in cortical layers II/III and in the hippocampus at 6-10 months of age compared to vehicle-injected mice. However, at 13-16 months of age, only cortical spine density was improved without changes in spine morphology. The changes in dendritic spine density correlated with Ras activity, such that 6-10 month old BTA-EG4 injected 3xTg AD mice had increased Ras activity in the cortex and hippocampus, while 13-16 month old mice only trended toward an increase in Ras activity in the cortex. Finally, BTA-EG4 injected 3xTg AD mice at 6-10 months of age showed improved learning and memory; however, only minimal improvement was observed at 13-16 months of age. This behavioral improvement corresponds to a decrease in soluble Aß 40 levels. Taken together, these findings suggest that BTA-EG4 may be beneficial in ameliorating the synaptic loss seen in early AD.


Subject(s)
Alzheimer Disease/complications , Aniline Compounds/therapeutic use , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Dendritic Spines/drug effects , Hippocampus/pathology , Age Factors , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Aniline Compounds/pharmacology , Animals , Disease Models, Animal , Hippocampus/ultrastructure , Humans , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Mutation/genetics , Presenilin-1/genetics , tau Proteins/genetics
11.
Exp Neurol ; 239: 192-201, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23063601

ABSTRACT

Histone deacetylase inhibitors (HDACIs) alter gene expression epigenetically by interfering with the normal functions of HDAC. Given their ability to decrease Aß levels, HDACIs are a potential treatment for Alzheimer's disease (AD). However, it is unclear how HDACIs alter Aß levels. We developed two novel HDAC inhibitors with improved pharmacological properties, such as a longer half-life and greater penetration of the blood-brain barrier: mercaptoacetamide-based class II HDACI (coded as W2) and hydroxamide-based class I and IIHDACI (coded as I2) and investigated how they affect Aß levels and cognition. HDACI W2 decreased Aß40 and Aß42 in vitro. HDACI I2 also decreased Aß40, but not Aß42. We systematically examined the molecular mechanisms by which HDACIs W2 and I2 can decrease Aß levels. HDACI W2 decreased gene expression of γ-secretase components and increased the Aß degradation enzyme Mmp2. Similarly, HDACI I2 decreased expression of ß- and γ-secretase components and increased mRNA levels of Aß degradation enzymes. HDACI W2 also significantly decreased Aß levels and rescued learning and memory deficits in aged hAPP 3xTg AD mice. Furthermore, we found that the novel HDACI W2 decreased tau phosphorylation at Thr181, an effect previously unknown for HDACIs. Collectively, these data suggest that class II HDACls may serve as a novel therapeutic strategy for AD.


Subject(s)
Acetamides/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Learning/drug effects , Memory/drug effects , Thioglycolates/pharmacology , Amyloid beta-Protein Precursor/metabolism , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Humans , Maze Learning/drug effects , Mice , Mice, Transgenic , Microinjections , Neurons/drug effects , Phosphorylation , Real-Time Polymerase Chain Reaction , Transfection , tau Proteins/metabolism
12.
Clin Exp Metastasis ; 29(2): 143-53, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22116632

ABSTRACT

Almost half of breast Ductal Carcinoma in situ are likely to remain non threatening in situ lesions with no invasion to the surrounding stroma and no metastases. The majority of focal disruptions in myoepithelial (ME) cell layers indicative of invasion onset were found to be overlying epithelial cell clusters with no or substantially reduced estrogen receptor α (ERα) expression. Here we report the down-regulation of tyrosine kinase-2 (TYK2) and up-regulation of strumpellin expression, among other proteins in ERα(-) cells located at disrupted ME layers compared to adjacent ERα(+) cells overlying an intact myoepithelial layer. ERα(+) and ERα(-) cells were microdissected from the same in vivo human breast cancer tissues, proteins were extracted and separated utilizing Differential in-Gel Electrophoresis followed by trypsin digestion, MALDI-TOF analysis, and protein identification. Proteins expressed by ERα(-) cell clusters were found to express higher levels of strumpellin that binds to valosin-containing protein (VCP) to slow-down wound closure and promote growth; and lower levels of TYK2, a jak protein necessary for lineage specific differentiation. TYK2 levels were further analyzed by immunohistochemistry in a cohort composed of 70 patients with broad clinical characteristics. TYK2 levels were minimal in TxN1M0 breast cancers which is the stage where the initial regional lymph node metastasis is observed. Our data highlight the role of TYK2 downregulation in breast cancer cell de-differentitation and initiation of regional metastasis. In addition, the aggressiveness of the ERα(-) cell clusters compared to ERα(+) ones present in the same duct of the same patient was confirmed.


Subject(s)
Breast Neoplasms/enzymology , Lymphatic Metastasis , TYK2 Kinase/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Middle Aged , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , TYK2 Kinase/genetics
13.
FASEB J ; 24(1): 58-69, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19720620

ABSTRACT

The goal of this study was to determine the effect of X11alpha on ApoE receptor 2 (ApoEr2) trafficking and the functional significance of this interaction on cell movement in MCF 10A epithelial cells. We found that X11alpha increased surface levels of ApoEr2 by 64% compared to vector control, as determined by surface protein biotinylation. To examine the functional significance of this effect, we tested whether ApoEr2 played a novel role in cell movement in a wound-healing assay. We found that overexpression of ApoEr2 in MCF 10A cells increased cell migration velocity by 87% (P<0.01, n=4) compared to GFP control. Cotransfection of X11alpha had an additive effect on average velocity compared to ApoEr2 alone (13%; P<0.05, n=4). In addition, we tested whether ApoEr2 ligands altered the effect of ApoEr2 on cell movement. We found that treatment with concentrated medium containing the extracellular matrix protein Reelin, but not control medium, further increased the velocity of ApoEr2- but not APP-transfected cells (20%; P<0.001, n=4). Similarly, Reelin treatment increased cell velocity in the presence of ApoEr2 and X11alpha (10%; P<0.05, n=4). In the present study, we are the first to demonstrate that ApoEr2 regulates cell movement, and both X11alpha and Reelin enhance this effect.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/physiology , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Lipoprotein/metabolism , Serine Endopeptidases/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites/genetics , Biological Transport, Active , COS Cells , Cadherins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Exons , Extracellular Matrix Proteins/genetics , Humans , LDL-Receptor Related Proteins , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons/metabolism , PDZ Domains , Phosphorylation , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , RNA, Small Interfering/genetics , Rats , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reelin Protein , Serine Endopeptidases/genetics , Transfection , Two-Hybrid System Techniques , Wound Healing/physiology
14.
PLoS One ; 4(10): e7445, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19829710

ABSTRACT

The normal function of Syk in epithelium of the developing or adult breast is not known, however, Syk suppresses tumor growth, invasion, and metastasis in breast cancer cells. Here, we demonstrate that in the mouse mammary gland, loss of one Syk allele profoundly increases proliferation and ductal branching and invasion of epithelial cells through the mammary fat pad during puberty. Mammary carcinomas develop by one year. Syk also suppresses proliferation and invasion in vitro. siRNA or shRNA knockdown of Syk in MCF10A breast epithelial cells dramatically increased proliferation, anchorage independent growth, cellular motility, and invasion, with formation of functional, extracellular matrix-degrading invadopodia. Morphological and gene microarray analysis following Syk knockdown revealed a loss of luminal and differentiated epithelial features with epithelial to mesenchymal transition and a gain in invadopodial cell surface markers CD44, CD49F, and MMP14. These results support the role of Syk in limiting proliferation and invasion of epithelial cells during normal morphogenesis, and emphasize the critical role of Syk as a tumor suppressor for breast cancer. The question of breast cancer risk following systemic anti-Syk therapy is raised since only partial loss of Syk was sufficient to induce mammary carcinomas.


Subject(s)
Genes, Tumor Suppressor , Intracellular Signaling Peptides and Proteins/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Protein-Tyrosine Kinases/metabolism , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/biosynthesis , Integrin alpha6/biosynthesis , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Transgenic , Neoplasm Invasiveness , Syk Kinase
15.
Carcinogenesis ; 28(10): 2063-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17277233

ABSTRACT

The EP2 prostanoid receptor is one of the four subtypes of receptors for prostaglandin E2 (PGE2). We previously reported that deletion of EP2 led to resistance to chemically induced mouse skin carcinogenesis, whereas overexpression of EP2 resulted in enhanced tumor development. The purpose of this study was to investigate the underlying molecular mechanisms. We found that EP2 knockout mice had reduced cyclooxygenase-2 (COX-2) expression after 12-O-tetradecanoylphorbol-13-acetate (TPA) treatment compared with wild-type (WT) mice. Further, primary keratinocytes from EP2 transgenic mice had increased COX-2 expression after either TPA or PGE2 treatment and COX-2 expression was blocked by 10 microM SQ 22,536, an adenylate cyclase inhibitor. EP2 knockout mice had significantly decreased, whereas EP2 transgenic mice had significantly increased PGE2 production in response to a single treatment of TPA. Cyclic AMP response element-binding protein (CREB) phosphorylation was elevated to a greater extent in keratinocytes from EP2 transgenic mice compared with those of WT mice following PGE2 treatment. A protein kinase A (PKA) inhibitor reduced PGE2-mediated CREB phosphorylation in keratinocytes from EP2 transgenic mice. Furthermore, we found that there was no CREB phosphorylation in EP2 knockout mice following PGE2 treatment. PGE2-induced DNA synthesis (cell proliferation) was significantly decreased in keratinocytes from EP2 knockout mice following pretreatment with 10 microM SQ 22,536. Taken together, EP2 activation of the PKA/CREB-signaling pathway is responsible for keratinocyte proliferation and our findings reveal a positive feedback loop between COX-2 and PGE2 that is mediated by the EP2 receptor.


Subject(s)
Cyclooxygenase 2/genetics , Dinoprostone/pharmacology , Receptors, Prostaglandin E/physiology , Skin Physiological Phenomena , Skin/enzymology , Animals , Cell Culture Techniques , Dinoprostone/metabolism , Epidermis/drug effects , Epidermis/enzymology , Epidermis/physiology , Gene Expression Regulation, Enzymologic , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratinocytes/physiology , Mice , Mice, Knockout , Mice, Transgenic , RNA/genetics , RNA/isolation & purification , Receptors, Prostaglandin E/deficiency , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Skin/drug effects , Thymidine/metabolism
16.
Article in English | MEDLINE | ID: mdl-16446079

ABSTRACT

Cyclooxygenase-2 (COX-2) plays an important role in tumorigenesis of several tissues, including skin. We report here that troglitazone, a thiazolidinedione class of antidiabetic drug, induced COX-2 expression at both the protein and mRNA levels and increased production of prostaglandin E2 (PGE2) in cultured keratinocytes. Troglitazone-induced COX-2 expression in keratinocytes was likely peroxisome proliferator-activated receptor gamma (PPARgamma)-independent. Troglitazone treatment of these cells also resulted in a sustained increase in phosphorylation of ERK. We show that induction of COX-2 by troglitazone was almost completely inhibited by specific inhibitors of ERK activation. These data suggest that troglitazone is capable of inducing COX-2 expression through an ERK-dependent mechanism in mouse skin keratinocytes.


Subject(s)
Chromans/pharmacology , Cyclooxygenase 2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Keratinocytes/metabolism , Thiazolidinediones/pharmacology , Animals , Blotting, Northern , Blotting, Western , Cell Line , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Gene Expression/drug effects , Keratinocytes/drug effects , Mice , PPAR gamma/agonists , PPAR gamma/genetics , Peroxisome Proliferator-Activated Receptors/agonists , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Troglitazone , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Cancer Res ; 66(3): 1873-8, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452250

ABSTRACT

Thiazolidinediones are a novel class of antidiabetic drugs that improve insulin sensitivity in type 2 diabetic patients. Recently, these compounds have also been shown to suppress tumor development in several animal models. The molecular basis for their antitumor action, however, is largely unknown. We report here that oral administration of thiazolidinediones (rosiglitazone and troglitazone) remarkably inhibited insulin-like growth factor-I (IGF-I)-promoted skin tumor development by 73% in BK5.IGF-1 transgenic mice, although they were previously found to be ineffective in inhibiting UV- or chemically induced mouse skin tumorigenesis. The anti-IGF-I effect of troglitazone in mouse skin keratinocytes was due to, at least partially, inhibition of IGF-I-induced phosphorylation of p70S6 kinase (p70S6K) at Thr(389), a site specifically phosphorylated by mammalian target of rapamycin (mTOR). Troglitazone did not directly inhibit mTOR kinase activity as shown by mTOR in vitro kinase assay but rapidly activated AMP-activated protein kinase (AMPK) through a yet undefined peroxisome proliferator-activated receptor gamma-independent mechanism. Expression of a dominant-negative AMPK reversed the inhibitory effect of troglitazone on IGF-I-induced phosphorylation of p70S6K, suggesting that troglitazone inhibited IGF-I and p70S6K signaling through activation of AMPK. Collectively, these data suggest that thiazolidinediones specifically inhibit IGF-I tumor-promoting activity in mouse skin through activation of AMPK and subsequent inhibition of p70S6K.


Subject(s)
Antineoplastic Agents/pharmacology , Chromans/pharmacology , Insulin-Like Growth Factor I/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Skin Neoplasms/prevention & control , Thiazolidinediones/pharmacology , AMP-Activated Protein Kinases , Animals , Cell Growth Processes/drug effects , Enzyme Activation/drug effects , Humans , Insulin-Like Growth Factor I/pharmacology , Keratinocytes/drug effects , Mice , Mice, Transgenic , Multienzyme Complexes/metabolism , Phosphorylation/drug effects , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Rosiglitazone , Signal Transduction/drug effects , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , TOR Serine-Threonine Kinases , Transfection , Troglitazone
18.
Cancer Res ; 65(20): 9304-11, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16230392

ABSTRACT

The EP2 receptor for prostaglandin E2 (PGE2) is a membrane receptor that mediates at least part of the action of PGE2. It has been shown that EP2 plays a critical role in tumorigenesis in mouse mammary gland and colon. However, the possibility that the EP2 receptor is involved in the development of skin tumors was unknown. The purpose of this study was to investigate the role of the EP2 receptor in mouse skin carcinogenesis. Unlike EP3 knockout mice, the EP2 knockout mice produced significantly fewer tumors and reduced tumor incidence compared with wild type (WT) mice in a 7,12-dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA) two-stage carcinogenesis protocol. EP2 knockout mice had significantly reduced cellular proliferation of mouse skin keratinocytes in vivo and in vitro compared with that in WT mice. In addition, the epidermis of EP2 knockout mice 48 hours after topical TPA treatment was significantly thinner compared with that of WT mice. The inflammatory response to TPA was reduced in EP2 knockout mice, based on a reduced number of macrophages in the dermis and a reduced level of interleukin-1alpha mRNA expression, compared with WT mice. EP2 knockout mice also had significantly reduced epidermal cyclic AMP levels after PGE2 treatment compared with WT mice. Tumors from WT mice produced more blood vessels and fewer apoptotic cells than those of EP2 knockout mice as determined by immunohistochemical staining. Our data suggest that the EP2 receptor plays a significant role in the protumorigenic action of PGE2 in skin tumor development.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Dinoprostone/metabolism , Receptors, Prostaglandin E/deficiency , Skin Neoplasms/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Animals , Apoptosis/genetics , Apoptosis/physiology , Carcinogens , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Transformation, Neoplastic/drug effects , Cyclic AMP/metabolism , Dinoprostone/pharmacology , Drug Eruptions/genetics , Drug Eruptions/metabolism , Drug Eruptions/pathology , Female , Hyperplasia , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Knockout , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Receptors, Prostaglandin E/biosynthesis , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate
SELECTION OF CITATIONS
SEARCH DETAIL
...