Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Article in English | MEDLINE | ID: mdl-38932684

ABSTRACT

Resistance training (RT) remains the most effective treatment for age-related declines in muscle mass. However, many older adults experience attenuated muscle hypertrophy in response to RT when compared to younger adults. This may be attributed to underlying molecular processes that are dysregulated by aging and exacerbated by improperly prescribed RT weekly volume, intensity, and/or frequency doses. MicroRNA (miRNA) are key epigenetic regulators that impact signaling pathways and protein expression within cells, are dynamic and responsive to exercise stimuli, and are often dysregulated in diseases. In this study, we used untargeted miRNA-seq to examine miRNA in skeletal muscle and serum-derived exosomes of older adults (n = 18, 11M/7F, 66±1y) who underwent 3x/wk RT for 30 weeks [e.g., high intensity 3x/wk (HHH, n = 9) or alternating high-low-high intensity (HLH, n = 9)], after a standardized four-week wash-in. Within each tissue, miRNAs were clustered into modules based on pairwise correlation using Weighted Gene Correlation Network Analysis (WGCNA). Modules were tested for association with the magnitude of RT-induced thigh lean mass (TLM) change (as measured by DXA). While no modules were unique to training dose, we identified miRNA modules in skeletal muscle associated with TLM gains irrespective of exercise dose. Using miRNA-target interactions, we analyzed key miRNAs in significant modules for their potential regulatory involvement in biological pathways. Findings point toward potential miRNAs that may be informative biomarkers and could also be evaluated as potential therapeutic targets as an adjuvant to RT in order to maximize skeletal muscle mass accrual in older adults.

2.
Cell Death Discov ; 10(1): 13, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191532

ABSTRACT

ΔNp63α, a member of the p53 family of transcription factors, plays a critical role in maintaining the proliferative potential of stem cells in the stratified epithelium. Although ΔNp63α is considered an oncogene and is frequently overexpressed in squamous cell carcinoma, loss of ΔNp63α expression is associated with increased tumor cell invasion and metastasis. We recently identified a ΔNp63α/miR-320a/PKCγ signaling axis that regulates cancer cell invasion by inhibiting phosphorylation of the small GTPase Rac1, a master switch of cell motility that positively regulates cell invasion in multiple human cancers. In this study, we identified a novel mechanism by which ΔNp63α negatively regulates Rac1 activity, by inhibiting the expression of the Rac-specific Guanine Exchange Factor PREX1. ΔNp63α knockdown in multiple squamous cell carcinoma cell lines leads to increased Rac1 activation, which is abrogated by treatment with the Rac1 inhibitor NSC23766. Furthermore, ΔNp63α negatively regulates PREX1 transcript and protein levels. Using a Rac-GEF activation assay, we also showed that ΔNp63α reduces the levels of active PREX1. The inhibition of the PREX1-Rac1 signaling axis by ΔNp63α leads to impaired cell invasion, thus establishing the functional relevance of this link. Our results elucidated a novel molecular mechanism by which ΔNp63α negatively affects cancer cell invasion and identifies the ΔNp63α/Rac1 axis as a potential target for metastasis.

3.
BMC Cancer ; 21(1): 155, 2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33579235

ABSTRACT

BACKGROUND: p63, a member of the p53 gene family, is an important regulator for epithelial tissue growth and development. ∆Np63α is the main isoform of p63 and highly expressed in Non-melanoma skin cancer (NMSC). Extracellular signal-regulated kinase 3 (ERK3) is an atypical mitogen-activated protein kinase (MAPK) whose biochemical features and cellular regulation are distinct from those of conventional MAPKs such as ERK1/2. While ERK3 has been shown to be upregulated in lung cancers and head and neck cancers, in which it promotes cancer cell migration and invasion, little is known about the implication of ERK3 in NMSCs. METHODS: Fluorescent immunohistochemistry was performed to evaluate the expression levels of ΔNp63α and ERK3 in normal and NMSC specimens. Dunnett's test was performed to compare mean fluorescence intensity (MFI, indicator of expression levels) of p63 or ERK3 between normal cutaneous samples and NMSC samples. A mixed effects (ANOVA) test was used to determine the correlation between ΔNp63α and ERK3 expression levels (MFI). The regulation of ERK3 by ΔNp63α was studied by qRT-PCR, Western blot and luciferase assay. The effect of ERK3 regulation by ΔNp63α on cell migration was measured by performing trans-well migration assay. RESULTS: The expression level of ∆Np63α is upregulated in NMSCs compared to normal tissue. ERK3 level is significantly upregulated in AK and SCC in comparison to normal tissue and there is a strong positive correlation between ∆Np63α and ERK3 expression in normal skin and skin specimens of patients with AK, SCC or BCC. Further, we found that ∆Np63α positively regulates ERK3 transcript and protein levels in A431 and HaCaT skin cells, underlying the upregulation of ERK3 expression and its positive correlation with ∆Np63α in NMSCs. Moreover, similar to the effect of ∆Np63α depletion, silencing ERK3 greatly enhanced A431 cell migration. Restoration of ERK3 expression under the condition of silencing ∆Np63α counteracted the increase in cell migration induced by the depletion of ∆Np63α. Mechanistically, ERK3 inhibits the phosphorylation of Rac1 G-protein and the formation of filopodia of A431 skin SCC cells. CONCLUSIONS: ERK3 is positively regulated by ∆Np63α and mediates the role of ∆Np63α in suppressing cell migration in NMSC.


Subject(s)
Cell Movement , Gene Expression Regulation, Neoplastic , Mitogen-Activated Protein Kinase 6/metabolism , Skin Neoplasms/pathology , Transcription Factors/metabolism , Transcriptional Activation , Tumor Suppressor Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Line , Cell Line, Tumor , Humans , Mitogen-Activated Protein Kinase 6/genetics , Phosphorylation , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , rac1 GTP-Binding Protein/genetics
4.
Clin Transl Gastroenterol ; 11(1): e00125, 2020 01.
Article in English | MEDLINE | ID: mdl-31934893

ABSTRACT

OBJECTIVES: Barrett's esophagus (BE) is the precursor lesion and a major risk factor for esophageal adenocarcinoma (EAC). Although patients with BE undergo routine endoscopic surveillance, current screening methodologies have proven ineffective at identifying individuals at risk of EAC. Since microRNAs (miRNAs) have potential diagnostic and prognostic value as disease biomarkers, we sought to identify an miRNA signature of BE and EAC. METHODS: High-throughput sequencing of miRNAs was performed on serum and tissue biopsies from 31 patients identified either as normal, gastroesophageal reflux disease (GERD), BE, BE with low-grade dysplasia (LGD), or EAC. Logistic regression modeling of miRNA profiles with Lasso regularization was used to identify discriminating miRNA. Quantitative reverse transcription polymerase chain reaction was used to validate changes in miRNA expression using 46 formalin-fixed, paraffin-embedded specimens obtained from normal, GERD, BE, BE with LGD or HGD, and EAC subjects. RESULTS: A 3-class predictive model was able to classify tissue samples into normal, GERD/BE, or LGD/EAC classes with an accuracy of 80%. Sixteen miRNAs were identified that predicted 1 of the 3 classes. Our analysis confirmed previous reports indicating that miR-29c-3p and miR-193b-5p expressions are altered in BE and EAC and identified miR-4485-5p as a novel biomarker of esophageal dysplasia. Quantitative reverse transcription polymerase chain reaction validated 11 of 16 discriminating miRNAs. DISCUSSION: Our data provide an miRNA signature of normal, precancerous, and cancerous tissue that may stratify patients at risk of progressing to EAC. We found that serum miRNAs have a limited ability to distinguish between disease states, thus limiting their potential utility in early disease detection.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , Esophageal Neoplasms/genetics , Esophagus/metabolism , Gastroesophageal Reflux/genetics , MicroRNAs/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Case-Control Studies , Discriminant Analysis , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophagus/pathology , Gastroesophageal Reflux/metabolism , Gastroesophageal Reflux/pathology , Humans , Logistic Models , MicroRNAs/blood , MicroRNAs/metabolism , Middle Aged , Neoplasm Grading , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome
5.
J Biol Chem ; 294(45): 17007-17016, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31601649

ABSTRACT

An estimated 5.4 million cases of nonmelanoma skin cancer are reported in the United States at an associated cost of $4.8 billion. ΔNp63α, a proto-oncogene in the p53 family of transcription factors, is overexpressed in squamous cell carcinoma (SCC) and associated with poor prognosis and survival. ΔNp63α elicits its tumorigenic effects in part by promoting cellular proliferation and cell survival. Despite its importance in SCC, the upstream regulation of ΔNp63α is poorly understood. In this study, we identify TIP60 as a novel upstream regulator of ΔNp63α. Using a combination of overexpression, silencing, stable expression, and pharmacological approaches in multiple cell lines, we showed that TIP60 up-regulates ΔNp63α expression. Utilizing cycloheximide treatment, we showed that TIP60 catalytic activity is required for stabilization of ΔNp63α protein levels. We further showed that TIP60 coexpression inhibits ΔNp63α ubiquitination and proteasomal degradation. Stabilization of ΔNp63α protein was further associated with TIP60-mediated acetylation. Finally, we demonstrated that TIP60-mediated regulation of ΔNp63α increases cellular proliferation by promoting G2/M progression through MTS assays and flow cytometry. Taken together, our findings provide evidence that TIP60 may contribute to SCC progression by increasing ΔNp63α protein levels, thereby promoting cellular proliferation.


Subject(s)
Lysine Acetyltransferase 5/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Up-Regulation , Acetylation , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Proto-Oncogene Mas , RNA, Messenger/genetics
6.
Cell Death Dis ; 10(9): 680, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31515469

ABSTRACT

ΔNp63α, a member of the p53 family of transcription factors, is overexpressed in a number of cancers and plays a role in proliferation, differentiation, migration, and invasion. ΔNp63α has been shown to regulate several microRNAs that are involved in development and cancer. We identified miRNA miR-320a as a positively regulated target of ΔNp63α. Previous studies have shown that miR-320a is downregulated in colorectal cancer and targets the small GTPase Rac1, leading to a reduction in noncanonical WNT signaling and EMT, thereby inhibiting tumor metastasis and invasion. We showed that miR-320a is a direct target of ΔNp63α. Knockdown of ΔNp63α in HaCaT and A431 cells downregulates miR-320a levels and leads to a corresponding elevation in PKCγ transcript and protein levels. Rac1 phosphorylation at Ser71 was increased in the absence of ΔNp63α, whereas overexpression of ΔNp63α reversed S71 phosphorylation of Rac1. Moreover, increased PKCγ levels, Rac1 phosphorylation and cell invasion observed upon knockdown of ΔNp63α was reversed by either overexpressing miR-320a mimic or Rac1 silencing. Finally, silencing PKCγ or treatment with the PKC inhibitor Gö6976 reversed increased Rac1 phosphorylation and cell invasion observed upon silencing ΔNp63α. Taken together, our data suggest that ΔNp63α positively regulates miR-320a, thereby inhibiting PKCγ expression, Rac1 phosphorylation, and cancer invasion.


Subject(s)
MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Protein Kinase C-delta/metabolism , Tumor Suppressor Protein p53/metabolism , rac1 GTP-Binding Protein/metabolism , Blotting, Western , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , MicroRNAs/genetics , Neoplasm Invasiveness/pathology , Phosphorylation/genetics , Phosphorylation/physiology , Protein Kinase C-delta/genetics , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics , rac1 GTP-Binding Protein/genetics
7.
ACS Appl Mater Interfaces ; 11(15): 13927-13934, 2019 Apr 17.
Article in English | MEDLINE | ID: mdl-30884221

ABSTRACT

Transmission electron microscopy (TEM) is being pushed to new capabilities which enable studies on systems that were previously out of reach. Among recent innovations, TEM through liquid cells (LC-TEM) enables in operando observation of biological phenomena. This work applies LC-TEM to the study of biological components as they interact on an abiotic surface. Specifically, analytes or target molecules like neuropeptide Y (NPY) are observed in operando on functional graphene field-effect transistor (GFET) biosensors. Biological recognition elements (BREs) identified using biopanning with affinity to NPY are used to functionalize graphene to obtain selectivity. On working devices capable of achieving picomolar responsivity to neuropeptide Y, LC-TEM reveals translational motion, stochastic positional fluctuations due to constrained Brownian motion, and rotational dynamics of captured analyte. Coupling these observations with the electrical responses of the GFET biosensors in response to analyte capture and/or release will potentially enable new insights leading to more advanced and capable biosensor designs.


Subject(s)
Biosensing Techniques/methods , Graphite/chemistry , Neuropeptide Y/analysis , Transistors, Electronic , Area Under Curve , Biosensing Techniques/instrumentation , Cryoelectron Microscopy , Humans , Neuropeptide Y/chemistry , Peptide Library , Protein Binding , ROC Curve , Sweat/metabolism
8.
Sci Rep ; 8(1): 10069, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29968742

ABSTRACT

Advances in high-throughput sequencing have enabled profiling of microRNAs (miRNAs), however, a consensus pipeline for sequencing of small RNAs has not been established. We built and optimized an analysis pipeline using Partek Flow, circumventing the need for analyzing data via scripting languages. Our analysis assessed the effect of alignment reference, normalization method, and statistical model choice on biological data. The pipeline was evaluated using sequencing data from HaCaT cells transfected with either a non-silencing control or siRNA against ΔNp63α, a p53 family member protein which is highly expressed in non-melanoma skin cancer and shown to regulate a number of miRNAs. We posit that 1) alignment and quantification to the miRBase reference provides the most robust quantitation of miRNAs, 2) normalizing sample reads via Trimmed Mean of M-values is the most robust method for accurate downstream analyses, and 3) use of the lognormal with shrinkage statistical model effectively identifies differentially expressed miRNAs. Using our pipeline, we identified previously unrecognized regulation of miRs-149-5p, 18a-5p, 19b-1-5p, 20a-5p, 590-5p, 744-5p and 93-5p by ΔNp63α. Regulation of these miRNAs was validated by RT-qPCR, substantiating our small RNA-Seq pipeline. Further analysis of these miRNAs may provide insight into ΔNp63α's role in cancer progression. By defining the optimal alignment reference, normalization method, and statistical model for analysis of miRNA sequencing data, we have established an analysis pipeline that may be carried out in Partek Flow or at the command line. In this manner, our pipeline circumvents some of the major hurdles encountered during small RNA-Seq analysis.


Subject(s)
MicroRNAs/analysis , Sequence Analysis, RNA/methods , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Algorithms , Cell Line , Gene Expression Profiling/methods , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing/methods , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction/methods , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
9.
Oncotarget ; 9(8): 8179-8196, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29487725

ABSTRACT

Esophageal adenocarcinoma (EAC) is a highly aggressive malignancy that develops from Barrett's esophagus (BE), an intestinal metaplasia of the distal esophagus. microRNAs (miRNAs), short non-coding regulatory RNAs, are frequently dysregulated in BE and are thought to play key roles in the onset of BE and its progression to EAC. miRNAs thus have potential diagnostic and prognostic value and are increasingly being used as cancer biomarkers. This review summarizes the current literature related to miRNAs that are dysregulated in BE within the context of Hedgehog, Notch, MAPK, NF kappa-B, Wnt and epithelial-mesenchymal transition (EMT) signaling which are thought to drive BE onset and progression. This comprehensive analysis of miRNAs and their associated signaling in the regulation of BE provides an overview of vital discoveries in this field and highlights gaps in our understanding of BE pathophysiology that warrant further investigation.

10.
Oncotarget ; 8(2): 2114-2129, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27924063

ABSTRACT

The epithelial-mesenchymal transition (EMT) is a cellular reprogramming mechanism that is an underlying cause of cancer metastasis. Recent investigations have uncovered an intricate network of regulation involving the TGFß, Wnt, and Notch signaling pathways and small regulatory RNA species called microRNAs (miRNAs). The activity of a transcription factor vital to the maintenance of epithelial stemness, ΔNp63α, has been shown to modulate the activity of these EMT pathways to either repress or promote EMT. Furthermore, ΔNp63α is a known regulator of miRNA, including those directly involved in EMT. This review discusses the evidence of ΔNp63α as a master regulator of EMT components and miRNA, highlighting the need for a deeper understanding of its role in EMT. This expanded knowledge may provide a basis for new developments in the diagnosis and treatment of metastatic cancer.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Transcription Factors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Transformation, Neoplastic/genetics , Cellular Reprogramming/genetics , Gene Expression Regulation , Humans , Signal Transduction/genetics
11.
Am J Hum Biol ; 28(5): 736-42, 2016 09 10.
Article in English | MEDLINE | ID: mdl-27135761

ABSTRACT

OBJECTIVES: To examine the association between 25-hydroxyvitamin D [25(OH)D] concentrations and cardiometabolic risk factors in obese American children. METHODS: A cross-sectional study was conducted on 209 obese children (55% females, 25.8% black) aged between 6 and 19 years old. Study measurements included plasma 25(OH)D concentrations, blood pressure, lipids and oxidized LDL levels, insulin resistance (IR) indices from glucose, insulin and 5 hour oral glucose tolerance test. RESULTS: Fifty-one percent of the children had vitamin D deficiency. Older age [OR (95% CI) = 1.16 (1.00, 1.35)], black race/ethnicity [15.39 (5.79, 40.92)], winter/spring season [3.46 (1.69, 7.02)] and higher body mass index (BMI) [1.05 (0.99, 1.11)] were associated with increased odds of having vitamin D deficiency. None of cardiometabolic risk factors examined were significantly associated with vitamin D deficiency in age, race/ethnicity, season, and BMI adjusted models. In age, race/ethnicity, season and BMI adjusted models, total cholesterol (ß = -0.001, P = 0.013), non-HDL-C (ß = -0.001, P = 0.014), and oxidized LDL (ß = -0.087, P = 0.045) were inversely associated with log-transformed 25(OH)D. An approximate 10 mg/dl increase in total cholesterol or in non-HDL-C was associated with an approximate 1.3% decrease in the geometric mean of 25(OH)D concentration. Further a 10% increase in ox-LDL levels was associated with an approximate 0.8% decrease in the geometric mean of 25(OH)D. CONCLUSION: Vitamin D deficiency is prevalent in obese American children. There was evidence that some cardiometabolic risk factors including lipid levels and oxidized LDL levels were significantly inversely associated with 25(OH)D concentration in our sample. Am. J. Hum. Biol. 28:736-742, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cardiovascular Diseases/epidemiology , Obesity/metabolism , Vitamin D Deficiency/epidemiology , Vitamin D/analogs & derivatives , Adolescent , Cardiovascular Diseases/etiology , Child , Cross-Sectional Studies , Female , Humans , Male , Ohio/epidemiology , Risk Factors , Vitamin D/blood , Vitamin D Deficiency/etiology
12.
Toxicol Res (Camb) ; 5(6): 1733-1743, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-30090472

ABSTRACT

Regulation of gene expression by non-coding RNAs, such as microRNAs (miRNAs), is increasingly being examined in a variety of disciplines. Here we evaluated changes in miRNA expression following metallic nanoparticle (NP) exposure in a mouse neuronal co-culture model. Exposure to manganese (Mn) NPs resulted in oxidative stress, inflammation, and toxicity. Next-generation sequencing (NGS) following an 8 h exposure to Mn NPs (low and high doses) revealed several miRNA candidates that modulate NP induced responses. The lead candidate identified was miR-155, which showed a dose dependent decrease in expression upon Mn exposure. Introduction of a miR-155 mimic into the co-culture to restore miR-155 expression completely abrogated the Mn NP-induced gene and protein expression of inflammatory markers TNF-α and IL-6. Taken together, this study is the first report where global NP-induced miRNA expression changes were used to identify and then modulate negative impacts of metallic NP exposure in a neuronal model. These findings demonstrate that unique miRNA expression profiles provide novel targets for manipulating gene and protein expression, and therefore provide the potential of modifying cellular responses to NP exposure.

13.
PLoS One ; 9(9): e107052, 2014.
Article in English | MEDLINE | ID: mdl-25191969

ABSTRACT

ΔNp63α, a proto-oncogene, is up-regulated in non-melanoma skin cancers and directly regulates the expression of both Vitamin D receptor (VDR) and phosphatase and tensin homologue deleted on chromosome ten (PTEN). Since ΔNp63α has been shown to inhibit cell invasion via regulation of VDR, we wanted to determine whether dietary Vitamin D3 protected against UVB induced tumor formation in SKH-1 mice, a model for squamous cell carcinoma development. We examined whether there was a correlation between dietary Vitamin D3 and ΔNp63α, VDR or PTEN expression in vivo in SKH-1 mice chronically exposed to UVB radiation and fed chow containing increasing concentrations of dietary Vitamin D3. Although we observed differential effects of the Vitamin D3 diet on ΔNp63α and VDR expression in chronically irradiated normal mouse skin as well as UVB induced tumors, Vitamin D3 had little effect on PTEN expression in vivo. While low-grade papillomas in mice exposed to UV and fed normal chow displayed increased levels of ΔNp63α, expression of both ΔNp63α and VDR was reduced in invasive tumors. Interestingly, in mice fed high Vitamin D3 chow, elevated levels of ΔNp63α were observed in both local and invasive tumors but not in normal skin suggesting that oral supplementation with Vitamin D3 may increase the proliferative potential of skin tumors by increasing ΔNp63α levels.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cholecalciferol/pharmacology , Phosphoproteins/genetics , Skin Neoplasms/genetics , Trans-Activators/genetics , Ultraviolet Rays , Animals , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Diet , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Male , Mice , Mice, Hairless , PTEN Phosphohydrolase/genetics , Receptors, Calcitriol/genetics , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin/radiation effects , Skin Neoplasms/pathology
14.
Cell Rep ; 6(5): 818-26, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24582960

ABSTRACT

Inactivation of the Pten tumor suppressor negatively regulates the PI3K-mTOR pathway. In a model of cutaneous squamous cell carcinoma (SCC), we demonstrate that deletion of Pten strongly elevates Fgf10 protein levels without increasing Fgf10 transcription in vitro and in vivo. The translational activation of Fgf10 by Pten deletion is reversed by genetic disruption of the mTORC1 complex, which also prevents skin tumorigenesis in Pten mutants. We further show that ectopic expression of Fgf10 causes skin papillomas, whereas Pten deletion-induced skin tumors are inhibited by epidermal deletion of Fgfr2. Collectively, our data identify autocrine activation of FGF signaling as an essential mechanism in promoting Pten-deficient skin tumors.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Transformation, Neoplastic/metabolism , Fibroblast Growth Factor 10/metabolism , PTEN Phosphohydrolase/deficiency , Skin Neoplasms/metabolism , Animals , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Fibroblast Growth Factor 10/genetics , Humans , Mice , Mice, Transgenic , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Transfection
15.
PLoS One ; 8(9): e74741, 2013.
Article in English | MEDLINE | ID: mdl-24040331

ABSTRACT

Serdemetan (JNJ-26854165), an antagonist to Mdm2, was anticipated to promote the activation of p53. While regulation of p53 by Mdm2 is important, Mdm2 also regulates numerous proteins involved in diverse cellular functions. We investigated if Serdemetan would alter the Mdm2-HIF1α axis and affect cell survival in human glioblastoma cells independently of p53. Treatment of cells with Serdemetan under hypoxia resulted in a decrease in HIF1α levels. HIF1α downstream targets, VEGF and the glycolytic enzymes (enolase, phosphoglycerate kinase1/2, and glucose transporter 1), were all decreased in response to Serdemetan. The involvement of Mdm2 in regulating gene expression of glycolytic enzymes raises the possibility of side effects associated with therapeutically targeting Mdm2.


Subject(s)
Brain Neoplasms/enzymology , Gene Expression Regulation, Neoplastic , Glioblastoma/enzymology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tryptamines/pharmacology , Cell Line, Tumor/drug effects , Cell Survival , Glycolysis , Humans , Hypoxia , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism
16.
Arch Dermatol Res ; 305(8): 733-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23589096

ABSTRACT

ΔNp63α maintains the proliferative potential of keratinocytes by inhibiting the transcription and nuclear localization of the tumor suppressor PTEN as shown earlier by our laboratory. The goal of this study was to define the mechanisms by which ΔNp63α mediates the nuclear exclusion of PTEN. We demonstrate here that ΔNp63α reduces the ubiquitination of PTEN, a key signaling event in the nuclear translocation of PTEN. The decrease in ubiquitinated PTEN correlated with the ability of ΔNp63α to bind to neuronal precursor developmentally down regulated 4 (NEDD4) promoter and transcriptionally repress the E3 ubiquitin ligase NEDD4-1. Knockdown of NEDD4-1 in cultured keratinocytes was sufficient to attenuate the increase in nuclear PTEN observed upon silencing of ΔNp63α. In vivo examination of normal skin demonstrated that ΔNp63α and NEDD4-1 were both expressed in the basal layer of the epidermis and this correlated with nuclear exclusion of PTEN. Altogether, these studies suggest that ΔNp63α-mediated suppression of nuclear PTEN in basal layer keratinocytes occurs through repression of NEDD4-1.


Subject(s)
Active Transport, Cell Nucleus , Endosomal Sorting Complexes Required for Transport/metabolism , Keratinocytes/metabolism , PTEN Phosphohydrolase/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line , Cell Proliferation , Down-Regulation , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/genetics , Epidermis/metabolism , Humans , Nedd4 Ubiquitin Protein Ligases , Promoter Regions, Genetic , Signal Transduction , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination
17.
Cell Cycle ; 12(9): 1406-15, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23574721

ABSTRACT

Loss of the tumor suppressor PTEN is observed in many human cancers that display increased chromosome instability and aneuploidy. The subcellular fractions of PTEN are associated with different functions that regulate cell growth, invasion and chromosome stability. In this study, we show a novel role for PTEN in regulating mitotic centrosomes. PTEN localization at mitotic centrosomes peaks between prophase and metaphase, paralleling the centrosomal localization of PLK-1 and γ-tubulin and coinciding with the time frame of centrosome maturation. In primary keratinocytes, knockdown of PTEN increased whole-cell levels of γ-tubulin and PLK-1 in an Akt-dependent manner and had little effect on recruitment of either protein to mitotic centrosomes. Conversely, knockdown of PTEN reduced centrosomal levels of pericentrin in an Akt-independent manner. Inhibition of Akt activation with MK2206 reduced the whole-cell and centrosome levels of PLK-1 and γ-tubulin and also prevented the recruitment of PTEN to mitotic centrosomes. This reduction in centrosome-associated proteins upon inhibition of Akt activity may contribute to the increase in defects in centrosome number and separation observed in metaphase cells. Concomitant PTEN knockdown and Akt inhibition reduced the frequency of metaphase cells with centrosome defects when compared with MK2206 treatment alone, indicating that both PTEN and pAkt are required to properly regulate centrosome composition during mitosis. The findings presented in this study demonstrate a novel role for PTEN and Akt in controlling centrosome composition and integrity during mitosis and provide insight into how PTEN functions as a multifaceted tumor suppressor.


Subject(s)
Centrosome/metabolism , Mitosis , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Antigens/metabolism , Cell Cycle Proteins/metabolism , Enzyme Activation , Humans , Infant, Newborn , Keratinocytes/cytology , Keratinocytes/enzymology , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins/metabolism , Tubulin/metabolism , Polo-Like Kinase 1
18.
J Biol Chem ; 287(46): 39115-24, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-22995914

ABSTRACT

Recent evidence suggests a major role of tumor-stromal interactions in pancreatic cancer pathobiology. The chemokine CXCL12 (stromal cell-derived factor 1 (SDF-1)), abundantly produced by stromal cells, promotes progression, metastasis, and chemoresistance of pancreatic cancer cells. On the other hand, pancreatic tumor cell-derived sonic hedgehog (SHH) acts predominantly on stromal cells to induce desmoplasia and, thus, has a paracrine effect on tumorigenesis and therapeutic outcome. In this study, we examined the association between these two proteins of pathological significance in pancreatic cancer. Our data demonstrate that CXCL12 leads to a dose- and time-dependent up-regulation of SHH in pancreatic cancer cells. CXCL12-induced SHH up-regulation is specifically mediated through the receptor CXCR4 and is dependent on the activation of downstream Akt and ERK signaling pathways. Both Akt and ERK cooperatively promote nuclear accumulation of NF-κB by inducing the phosphorylation and destabilization of its inhibitory protein, IκB-α. Using dominant negative IκB-α, a SHH promoter (deletion mutant) reporter, and chromatin immunoprecipitation assays, we demonstrate that CXCL12 exposure enhances direct binding of NF-κB to the SHH promoter and that suppression of NF-κB activation abrogates CXCL12-induced SHH expression. Finally, our data demonstrate a strong correlative expression of CXCR4 and SHH in human pancreatic cancer tissues, whereas their expression is not observed in the normal pancreas. Altogether, our data reveal a novel mechanism underlying aberrant SHH expression in pancreatic cancer and identify a molecular link facilitating bidirectional tumor-stromal interactions.


Subject(s)
Chemokine CXCL12/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Chemokines/metabolism , Cytoplasm/metabolism , Genes, Dominant , Humans , Models, Biological , Promoter Regions, Genetic , Signal Transduction
19.
Mol Cancer Res ; 10(6): 778-86, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22622028

ABSTRACT

Epidermal melanocytes are skin cells specialized in melanin production. Activation of the melanocortin 1 receptor (MC1R) on melanocytes by α-melanocyte-stimulating hormone (α-MSH) induces synthesis of the brown/black pigment eumelanin that confers photoprotection from solar UV radiation (UVR). Contrary to keratinocytes, melanocytes are slow proliferating cells that persist in the skin for decades, in an environment with high levels of UVR-induced reactive oxygen species (ROS). We previously reported that in addition to its role in pigmentation, α-MSH also reduces oxidative stress and enhances the repair of DNA photoproducts in melanocytes, independent of melanin synthesis. Given the significance of ROS in carcinogenesis, here we investigated the mechanisms by which α-MSH exerts antioxidant effects in melanocytes. We show that activation of the MC1R by α-MSH contributes to phosphorylation of p53 on serine 15, a known requirement for stabilization and activation of p53, a major sensor of DNA damage. This effect is mediated by the cAMP/PKA pathway and by the activation of phosphoinositide 3-kinase (PI3K) ATR and DNA protein kinase (DNA-PK). α-MSH increases the levels of 8-oxoguanine DNA glycosylase (OGG1) and apurinic apyrimidinic endonuclease 1 (APE-1/Ref-1), enzymes essential for base excision repair. Nutlin-3, an HDM2 inhibitor, mimicked the effects of α-MSH resulting in reduced phosphorylation of H2AX (γ-H2AX), a marker of DNA damage. Conversely, the p53 inhibitor pifithrin-α or silencing of p53 abolished the effects of α-MSH and augmented oxidative stress. These results show that p53 is an important target of the downstream MC1R signaling that reduces oxidative stress and possibly malignant transformation of melanocytes.


Subject(s)
Melanocytes/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , alpha-MSH/pharmacology , Benzothiazoles/pharmacology , Blotting, Western , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Damage , DNA Glycosylases/metabolism , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Activated Protein Kinase/metabolism , Histones/metabolism , Humans , Hydrogen Peroxide/pharmacology , Imidazoles/pharmacology , Melanocytes/metabolism , Melanocytes/radiation effects , Oxidants/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , Piperazines/pharmacology , RNA Interference , Receptor, Melanocortin, Type 1/genetics , Receptor, Melanocortin, Type 1/metabolism , Signal Transduction/radiation effects , Toluene/analogs & derivatives , Toluene/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Ultraviolet Rays
20.
J Cell Sci ; 124(Pt 24): 4286-98, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22193954

ABSTRACT

Serine-arginine-rich (SR) proteins play a key role in alternative pre-mRNA splicing in eukaryotes. We recently showed that a large SR protein called Son has unique repeat motifs that are essential for maintaining the subnuclear organization of pre-mRNA processing factors in nuclear speckles. Motif analysis of Son highlights putative RNA interaction domains that suggest a direct role for Son in pre-mRNA splicing. Here, we used in situ approaches to show that Son localizes to a reporter minigene transcription site, and that RNAi-mediated Son depletion causes exon skipping on reporter transcripts at this transcription site. A genome-wide exon microarray analysis was performed to identify human transcription and splicing targets of Son. Our data show that Son-regulated splicing encompasses all known types of alternative splicing, the most common being alternative splicing of cassette exons. We confirmed that knockdown of Son leads to exon skipping in pre-mRNAs for chromatin-modifying enzymes, including ADA, HDAC6 and SetD8. This study reports a comprehensive view of human transcription and splicing targets for Son in fundamental cellular pathways such as integrin-mediated cell adhesion, cell cycle regulation, cholesterol biosynthesis, apoptosis and epigenetic regulation of gene expression.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/physiology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/chemistry , HeLa Cells , Humans , Metaphase , Minor Histocompatibility Antigens , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , Serine-Arginine Splicing Factors , Spindle Apparatus/ultrastructure , Transcription, Genetic , Tropomyosin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...