Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Heart Vessels ; 37(2): 347-358, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34727208

ABSTRACT

Calcific aortic valve disease (CAVD) is the most common heart valve disease requiring intervention. Most research on CAVD has focused on inflammation, ossification, and cellular phenotype transformation. To gain a broader picture into the wide range of cellular and molecular mechanisms involved in this disease, we compared the total protein profiles between calcified and non-calcified areas from 5 human valves resected during surgery. The 1413 positively identified proteins were filtered down to 248 proteins present in both calcified and non-calcified segments of at least 3 of the 5 valves, which were then analyzed using Ingenuity Pathway Analysis. Concurrently, the top 40 differentially abundant proteins were grouped according to their biological functions and shown in interactive networks. Finally, the abundance of selected osteogenic proteins (osteopontin, osteonectin, osteocalcin, osteoprotegerin, and RANK) was quantified using ELISA and/or immunohistochemistry. The top pathways identified were complement system, acute phase response signaling, metabolism, LXR/RXR and FXR/RXR activation, actin cytoskeleton, mineral binding, nucleic acid interaction, structural extracellular matrix (ECM), and angiogenesis. There was a greater abundance of osteopontin, osteonectin, osteocalcin, osteoprotegerin, and RANK in the calcified regions than the non-calcified ones. The osteogenic proteins also formed key connections between the biological signaling pathways in the network model. In conclusion, this proteomic analysis demonstrated the involvement of multiple signaling pathways in CAVD. The interconnectedness of these pathways provides new insights for the treatment of this disease.


Subject(s)
Aortic Valve Stenosis , Calcinosis , Aortic Valve/metabolism , Aortic Valve/surgery , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/surgery , Calcinosis/metabolism , Humans , Osteogenesis/physiology , Proteome/metabolism , Proteomics
2.
Am J Obstet Gynecol ; 224(3): 278.e1-278.e14, 2021 03.
Article in English | MEDLINE | ID: mdl-32835719

ABSTRACT

BACKGROUND: Obesity is a well-known risk factor for endometrial cancer, but the mechanisms of obesity-related carcinogenesis are not well defined, particularly for premenopausal women. With the continuing obesity epidemic, increases in the incidence of endometrial cancer and a younger age of diagnosis are often attributed to a hyperestrogenic state created by hormone production in adipose tissue, but significant knowledge gaps remain. The balance of estrogen-responsive signals has not been defined in the endometrium of premenopausal women with obesity, where obesity may not create hyperestrogenism in the context of ovaries being the primary source of estrogen production. Obesity is associated with a state of low-grade, chronic inflammation that can promote tumorigenesis, and it is also known that hormonal changes alter the immune microenvironment of the endometrium. However, limited research has been conducted on endometrial immune-response changes in women who have an increased risk for cancer due to obesity. OBJECTIVE: Endometrial estrogen-regulated biomarkers, previously shown to be dysregulated in endometrial cancer, were evaluated in a cohort of premenopausal women to determine if obesity is associated with differences in the biomarker expression levels, which might reflect an altered risk of developing cancer. The expression of a multiplexed panel of immune-related genes was also evaluated for expression differences related to obesity. STUDY DESIGN: Premenopausal women with a body mass index of ≥30 kg/m2 (n=97) or a body mass index of ≤25 kg/m2 (n=33) were prospectively enrolled in this cross-sectional study, which included the assessment of serum metabolic markers and a timed endometrial biopsy for pathologic evaluation, hormone-regulated biomarker analysis, and immune response gene expression analysis. Medical and gynecologic histories were obtained. Endometrial gene expression markers were also compared across the body mass index groups in a previous cohort of premenopausal women with an inherited cancer risk (Lynch syndrome). RESULTS: In addition to known systemic metabolic differences, histologically normal endometria from women with obesity showed a decrease in gene expression of progesterone receptor (P=.0027) and the estrogen-induced genes retinaldehyde dehydrogenase 2 (P=.008), insulin-like growth factor 1 (P=.016), and survivin (P=.042) when compared with women without obesity. The endometrial biomarkers insulin-like growth factor 1, survivin, and progesterone receptor remained statistically significant in multivariate linear regression models. In contrast, women with obesity and Lynch syndrome had an increased expression of insulin-like growth factor 1 (P=.017). There were no differences in endometrial proliferation, and limited endometrial immune differences were observed. CONCLUSION: When comparing premenopausal women with and without obesity in the absence of endometrial pathology or an inherited cancer risk, the expression of the endometrial biomarkers does not reflect a local hyperestrogenic environment, but it instead reflects a decreased cancer risk profile that may be indicative of a compensated state. In describing premenopausal endometrial cancer risk, it may be insufficient to attribute a high-risk state to obesity alone; further studies are warranted to evaluate individualized biomarker profiles for differences in the hormone-responsive signals or immune response. In patients with Lynch syndrome, the endometrial biomarker profile suggests that obesity further increases the risk of developing cancer.


Subject(s)
Estrogens/blood , Obesity/blood , Premenopause/blood , Adult , Biomarkers/blood , Cohort Studies , Cross-Sectional Studies , Endometrial Neoplasms/epidemiology , Endometrial Neoplasms/etiology , Endometrium/metabolism , Endometrium/pathology , Estrogens/biosynthesis , Female , Humans , Obesity/complications , Risk Factors
3.
BMC Genomics ; 20(1): 852, 2019 Nov 14.
Article in English | MEDLINE | ID: mdl-31727022

ABSTRACT

BACKGROUND: Cleft lip (CL), one of the most common congenital birth defects, shows considerable geographic and ethnic variation, with contribution of both genetic and environmental factors. Mouse genetic studies have identified several CL-associated genes. However, it remains elusive how these CL-associated genes are regulated and involved in CL. Environmental factors may regulate these genes at the post-transcriptional level through the regulation of non-coding microRNAs (miRNAs). In this study, we sought to identify miRNAs associated with CL in mice. RESULTS: Through a systematic literature review and a Mouse Genome Informatics (MGI) database search, we identified 55 genes that were associated with CL in mice. Subsequent bioinformatic analysis of these genes predicted that a total of 33 miRNAs target multiple CL-associated genes, with 20 CL-associated genes being potentially regulated by multiple miRNAs. To experimentally validate miRNA function in cell proliferation, we conducted cell proliferation/viability assays for the selected five candidate miRNAs (miR-124-3p, let-7a-5p, let-7b-5p, let-7c-5p, and let-7d-5p). Overexpression of miR-124-3p, but not of the others, inhibited cell proliferation through suppression of CL-associated genes in cultured mouse embryonic lip mesenchymal cells (MELM cells) isolated from the developing mouse lip region. By contrast, miR-124-3p knockdown had no effect on MELM cell proliferation. This miRNA-gene regulatory mechanism was mostly conserved in O9-1 cells, an established cranial neural crest cell line. Expression of miR-124-3p was low in the maxillary processes at E10.5, when lip mesenchymal cells proliferate, whereas it was greatly increased at later developmental stages, suggesting that miR-124-3p expression is suppressed during the proliferation phase in normal palate development. CONCLUSIONS: Our findings indicate that upregulated miR-124-3p inhibits cell proliferation in cultured lip cells through suppression of CL-associated genes. These results will have a significant impact, not only on our knowledge about lip morphogenesis, but also on the development of clinical approaches for the diagnosis and prevention of CL.


Subject(s)
Cleft Lip/genetics , Gene Expression Regulation , Lip/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , RNA Interference , Animals , Cell Proliferation/genetics , Cells, Cultured , Computational Biology/methods , Embryonic Development/genetics , Environment , Epigenesis, Genetic , Gene Expression Profiling , Mice , Mutation , Reproducibility of Results
4.
J Neurotrauma ; 34(6): 1175-1186, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27750479

ABSTRACT

Spinal cord injury (SCI) results in devastating changes to almost all aspects of a patient's life. In addition to a permanent loss of sensory and motor function, males also will frequently exhibit a profound loss of fertility through poorly understood mechanisms. We demonstrate that SCI causes measureable pathology in the testis both acutely (24 h) and chronically up to 1.5 years post-injury, leading to loss in sperm motility and viability. SCI has been shown in humans and rats to induce leukocytospermia, with the presence of inflammatory cytokines, anti-sperm antibodies, and reactive oxygen species found within the ejaculate. Using messenger RNA and metabolomic assessments, we describe molecular and cellular changes that occur within the testis of adult rats over an acute to chronic time period. From 24 h, 72 h, 28 days, and 90 days post-SCI, the testis reveal a distinct time course of pathological events. The testis show an acute drop in normal sexual organ processes, including testosterone production, and establishment of a pro-inflammatory environment. This is followed by a subacute initiation of an innate immune response and loss of cell cycle regulation, possibly due to apoptosis within the seminiferous tubules. At 1.5 years post-SCI, there is a chronic low level immune response as evidenced by an elevation in T cells. These data suggest that SCI elicits a wide range of pathological processes within the testes, the actions of which are not restricted to the acute phase of injury but rather extend chronically, potentially through the lifetime of the subject. The multiplicity of these pathological events suggest a single therapeutic intervention is unlikely to be successful.


Subject(s)
Spinal Cord Injuries/complications , Testicular Diseases/etiology , Testicular Diseases/metabolism , Animals , Disease Models, Animal , Gene Expression/genetics , Male , Metabolomics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Testicular Diseases/immunology
5.
Physiol Genomics ; 48(4): 281-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26884459

ABSTRACT

The intensification and concentration of animal production operations expose workers to high levels of organic dusts in the work environment. Exposure to organic dusts is a risk factor for the development of acute and chronic respiratory symptoms and diseases. Lung epithelium plays important roles in the control of immune and inflammatory responses to environmental agents to maintain lung health. To better understand the effects of organic dust on lung inflammatory responses, we characterized the gene expression profiles of A549 alveolar and Beas2B bronchial epithelial and THP-1 monocytic cells influenced by exposure to poultry dust extract by DNA microarray analysis using Illumina Human HT-12 v4 Expression BeadChip. We found that A549 alveolar and Beas2B bronchial epithelial and THP-1 cells responded with unique changes in the gene expression profiles with regulation of genes encoding inflammatory cytokines, chemokines, and other inflammatory proteins being common to all the three cells. Significantly induced genes included IL-8, IL-6, IL-1ß, ICAM-1, CCL2, CCL5, TLR4, and PTGS2. Validation by real-time qRT-PCR, ELISA, Western immunoblotting, and immunohistochemical staining of lung sections from mice exposed to dust extract validated DNA microarray results. Pathway analysis indicated that dust extract induced changes in gene expression influenced functions related to cellular growth and proliferation, cell death and survival, and cellular development. These data show that a broad range of inflammatory mediators produced in response to poultry dust exposure can modulate lung immune and inflammatory responses. This is the first report on organic dust induced changes in expression profiles in lung epithelial and THP-1 monocytic cells.


Subject(s)
Chemokines/genetics , Cytokines/genetics , Dust , Gene Expression Profiling/methods , Lung/cytology , Animals , Chemokines/immunology , Cytokines/immunology , Epithelial Cells/drug effects , Female , Humans , Inflammation Mediators/metabolism , Lung/immunology , Mice, Inbred C57BL , Pneumonia/genetics , Pneumonia/immunology , Poultry
6.
BMC Genomics ; 16: 984, 2015 Nov 21.
Article in English | MEDLINE | ID: mdl-26589571

ABSTRACT

BACKGROUND: Although extensive studies have investigated radiation-induced injuries in particular gastrointestinal (GI) segments, a systematic comparison among the different segments on the basis of mode, magnitude and mechanism has not been reported. Here, a comparative study of segment-specific molecular and cellular responses was performed on jejunum, ileum and colon obtained at three time points (4, 7 and 12 days after irradiation) from non-human primate (Rhesus macaque) models exposed to 6.7 Gy or 7.4 Gy total body irradiation (TBI). RESULTS: Pathway analysis on the gene expression profiles identified radiation-induced time-, dose- and segment-dependent activation of tumor necrosis factor α (TNFα) cascade, tight junction, apoptosis, cell cycle control/DNA damage repair and coagulation system signaling. Activation of these signaling pathways suggests that colon sustained the severest mucosal barrier disruption and inflammation, and jejunum the greatest DNA damage, apoptosis and endothelial dysfunction. These more pronounced alterations correlate with the high incidence of macroscopic pathologies that are observed in the colon after TBI. Compared to colon and jejunum, ileum was resistant to radiation injury. In addition to the identification a marked increase of TNFα cascade, this study also identified radiation induced strikingly up-regulated tight junction gene CLDN2 (196-fold after 7.4-Gy TBI), matrix degradation genes such as MMP7 (increased 11- and 41-fold after 6.7-Gy and 7.4-Gy TBI), and anoikis mediated gene EDA2R that mediate mucosal shedding and barrier disruption. CONCLUSIONS: This is the first systematic comparative study of the molecular and cellular responses to radiation injury in jejunum, ileum and colon. The strongest activation of TNFα cascades and the striking up-regulation of its down-stream matrix-dissociated genes suggest that TNFα modulation could be a target for mitigating radiation-induced mucosal barrier disruption.


Subject(s)
Colon/metabolism , Gene Expression Profiling , Gene Expression Regulation/radiation effects , Ileum/metabolism , Jejunum/metabolism , Transcriptome , Whole-Body Irradiation , Animals , Anoikis/genetics , Apoptosis/genetics , Cell Cycle , Cluster Analysis , Colon/immunology , Ileum/immunology , Immunity, Mucosal/genetics , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/radiation effects , Jejunum/immunology , Macaca mulatta , Male , Radiation Dosage , Radiation Injuries, Experimental , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/radiation effects , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/metabolism
7.
Gynecol Oncol ; 133(1): 83-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24680596

ABSTRACT

OBJECTIVE: Obesity-associated hyperestrogenism and hyperinsulinemia contribute significantly to the pathogenesis of endometrial cancer. We recently demonstrated that metformin, a drug long used for treatment of type 2 diabetes, attenuates both insulin- and estrogen-mediated proliferative signaling in the obese rat endometrium. In this study, we sought to identify tissue biomarkers that may prove clinically useful to predict tissue response for both prevention and therapeutic studies. We identified CGRRF1 (cell growth regulator with ring finger domain 1) as a novel metformin-responsive gene and characterized its possible role in endometrial cancer prevention. METHODS: CGRRF1 mRNA expression was evaluated by RT-qPCR in the endometrium of obese and lean rats, and also in normal and malignant human endometrium. CGRRF1 levels were genetically manipulated in endometrial cancer cells, and its effects on proliferation and apoptosis were evaluated by MTT and Western blot. RESULTS: CGRRF1 is significantly induced by metformin treatment in the obese rat endometrium. In vitro studies demonstrate that overexpression of CGRRF1 inhibits endometrial cancer cell proliferation. Analysis of human endometrial tumors reveals that CGRRF1 expression is significantly lower in hyperplasia, Grade 1, Grade 2, Grade 3, MMMT, and UPSC endometrial tumors compared to normal human endometrium (p<0.05), suggesting that loss of CGRRF1 is associated with the presence of disease. CONCLUSION: CGRRF1 represents a novel, reproducible tissue marker of metformin response in the obese endometrium. Furthermore, our preliminary data suggests that up-regulation of CGRRF1 expression may prove clinically useful in the prevention or treatment of endometrial cancer.


Subject(s)
Endometrium/drug effects , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/drug effects , Metformin/pharmacology , Obesity/metabolism , RNA, Messenger/analysis , Animals , Apoptosis/drug effects , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Endometrial Hyperplasia/genetics , Endometrial Hyperplasia/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Rats , Rats, Zucker , Reverse Transcriptase Polymerase Chain Reaction
8.
J Biol Chem ; 288(50): 35940-51, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24163369

ABSTRACT

Genome-scale mapping suggests that the function of DNA methylation varies with genomic context beyond transcriptional repression. However, the use of DNA-demethylating agents (e.g. 5-aza-2'-deoxycytidine (5aza-dC)) to study epigenetic regulation often focuses on gene activation and ignores repression elicited by 5aza-dC. Here, we show that repression of NEK2, which encodes the never in mitosis A (NIMA)-related kinase, by 5aza-dC is context-specific as NEK2 transcript levels were reduced in HCT116 colon cancer cells but not in isogenic p53(-/-) cells. Bisulfite sequencing showed that DNA methylation was restricted to the distal region of the NEK2 promoter. Demethylation by 5aza-dC was associated with increased accessibility to micrococcal nuclease, i.e. nucleosome depletion. Conversely, methyltransferase accessibility protocol for individual templates (MAPit) methylation footprinting showed that nucleosome occupancy and DNA methylation at the distal promoter were significantly increased in p53(-/-) cells, suggesting dynamic regulation of chromatin structure at this region by p53 in HCT116 cells. Stabilization of endogenous p53 by doxorubicin or ectopic expression of p53, but not a p53 DNA-binding mutant, decreased NEK2 expression. Chromatin immunoprecipitation demonstrated direct and specific association of p53 with the distal NEK2 promoter, which was enhanced by doxorubicin. Luciferase reporters confirmed that this region is required for p53-mediated repression of NEK2 promoter activity. Lastly, modulation of p53 abundance altered nucleosome occupancy and DNA methylation at its binding region. These results identify NEK2 as a novel p53-repressed gene, illustrate that its repression by 5aza-dC is specific and associated with nucleosome reorganization, and provide evidence that identification of partially methylated regions can reveal novel p53 target genes.


Subject(s)
DNA Methylation , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Protein p53/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Base Sequence , Binding Sites , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , DNA/metabolism , DNA Methylation/drug effects , Decitabine , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HCT116 Cells , Humans , NIMA-Related Kinases , Nucleosomes/drug effects , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
9.
Cancer Prev Res (Phila) ; 6(8): 774-81, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23639481

ABSTRACT

Women with Lynch syndrome have a 40% to 60% lifetime risk for developing endometrial cancer, a cancer associated with estrogen imbalance. The molecular basis for endometrial-specific tumorigenesis is unclear. Progestins inhibit estrogen-driven proliferation, and epidemiologic studies have shown that progestin-containing oral contraceptives (OCP) reduce the risk of endometrial cancer by 50% in women at general population risk. It is unknown whether they are effective in women with Lynch syndrome. Asymptomatic women ages 25 to 50 with Lynch syndrome were randomized to receive the progestin compounds Depo-Provera (depo-MPA) or OCP for three months. An endometrial biopsy and transvaginal ultrasound were conducted before and after treatment. Endometrial proliferation was evaluated as the primary endpoint. Histology and a panel of surrogate endpoint biomarkers were evaluated for each endometrial biopsy as secondary endpoints. A total of 51 women were enrolled, and 46 completed treatment. Two of the 51 women had complex hyperplasia with atypia at the baseline endometrial biopsy and were excluded from the study. Overall, both depo-MPA and OCP induced a dramatic decrease in endometrial epithelial proliferation and microscopic changes in the endometrium characteristic of progestin action. Transvaginal ultrasound measurement of endometrial stripe was not a useful measure of endometrial response or baseline hyperplasia. These results show that women with Lynch syndrome do show an endometrial response to short-term exogenous progestins, suggesting that OCP and depo-MPA may be reasonable chemopreventive agents in this high-risk patient population.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Biomarkers, Tumor/metabolism , Colorectal Neoplasms, Hereditary Nonpolyposis/complications , Contraceptives, Oral/therapeutic use , Endometrial Neoplasms/prevention & control , Medroxyprogesterone Acetate/therapeutic use , Adult , Biomarkers, Tumor/genetics , Endometrial Neoplasms/etiology , Endometrial Neoplasms/metabolism , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Middle Aged , Mutation/genetics , Prognosis , Prospective Studies
10.
PLoS One ; 8(1): e53711, 2013.
Article in English | MEDLINE | ID: mdl-23341980

ABSTRACT

BACKGROUND: The intestinal mucosa is the compartment that sustains the most severe injury in response to radiation and is therefore of primary interest. The use of whole gut extracts for analysis of gene expression may confound important changes in the mucosa. On the other hand, laser capture microdissection (LCM) is hampered by the unstable nature of RNA and by a more complicated collection process. This study assessed, in parallel samples from a validated radiation model, the indications for use of LCM for intestinal gene expression analysis. METHODOLOGY/PRINCIPAL FINDINGS: RNA was extracted from mouse whole intestine and from mucosa by LCM at baseline and 4 h, 24 h, and 3.5 d after total body irradiation and subjected to microarray analysis. Among mucosal genes that were altered > = 2-fold, less than 7% were present in the whole gut at 4 and 24 h, and 25% at 3.5 d. As expected, pathway analysis of mucosal LCM samples showed that radiation activated the coagulation system, lymphocyte apoptosis, and tight junction signaling, and caused extensive up-regulation of cell cycle and DNA damage repair pathways. Using similar stringent criteria, regulation of these pathways, with exception of the p53 pathway, was undetectable in the whole gut. Radiation induced a dramatic increase of caspase14 and ectodysplasin A2 receptor (Eda2r), a TNFα receptor, in both types of samples. CONCLUSIONS/SIGNIFICANCE: LCM-isolated mucosal specimens should be used to study cellular injury, cell cycle control, and DNA damage repair pathways. The remarkable increase of caspase14 and Eda2r suggests a novel role for these genes in regulating intestinal radiation injury. Comparative gene expression data from complex tissues should be interpreted with caution.


Subject(s)
Intestinal Mucosa/radiation effects , Intestine, Small/radiation effects , Laser Capture Microdissection/methods , Animals , Apoptosis/radiation effects , Caspase 14/metabolism , Cell Cycle/radiation effects , Cytokines/metabolism , DNA Repair/radiation effects , Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Intestine, Small/enzymology , Intestine, Small/metabolism , Male , Mice , Oligonucleotide Array Sequence Analysis , Signal Transduction/radiation effects , Time Factors , Transcriptome/radiation effects
11.
Exp Mol Pathol ; 94(1): 289-300, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22609242

ABSTRACT

It is now known that there are at least two basic patterns of cell injury progressing to cell death: cell injury with swelling, known as oncosis, and cell injury with shrinkage, known as apoptosis. Both types of cell death are "programmed" in the sense that the genetic information and many of the enzymes and other factors pre-exist in the cell. Previous investigation has pointed to cardiomyocyte ischemic injury evolving as the oncotic pattern of injury, although apoptosis has also been implicated. This study was designed, using a unique cell model system, to gain insight into the molecular events of anticancer agent-induced cardiomyocyte injury. Cardiomyocytes exposed for 2 h to 1.5 µg/ml sanguinarine consistently displayed the morphology of apoptosis in over 80% of cells, whereas a higher dose of 25 µg/ml at 2 h yielded the pattern of oncosis in over 90% of cells. Microarray analysis revealed altered expression of 2514 probes in sanguinarine-induced oncosis and 1643 probes in apoptosis at a level of significance of p<0.001. Some of the inductions such as perforin were found to be higher than 11-fold in oncosis. When perforin was blocked by perforin-specific siRNA we found a reduction in oncotic cell death. These results strengthen the notion that oncosis is not representative of nonspecific necrosis, but constitutes a genetically controlled form of "programmed cell death"; and also that oncosis might represent a pathogenetic mechanism of cardiomyocyte injury. This is also the first demonstration of the involvement of perforin in cardiomyocyte oncosis.


Subject(s)
Apoptosis , Cell Death , Myocytes, Cardiac/physiology , Perforin/genetics , Animals , Apoptosis/genetics , Benzophenanthridines/pharmacology , Cell Death/genetics , Cell Line , Embryonic Stem Cells , Isoquinolines/pharmacology , Mice , Myocytes, Cardiac/drug effects , RNA Interference , RNA, Small Interfering
12.
Chem Biol ; 19(9): 1126-41, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22999881

ABSTRACT

We compared transcriptomes of terminally differentiated mouse 3T3-L1 and human adipocytes to identify cell-specific differences. Gene expression and high content analysis (HCA) data identified the androgen receptor (AR) as both expressed and functional, exclusively during early human adipocyte differentiation. The AR agonist dihydrotestosterone (DHT) inhibited human adipocyte maturation by downregulation of adipocyte marker genes, but not in 3T3-L1. It is interesting that AR induction corresponded with dexamethasone activation of the glucocorticoid receptor (GR); however, when exposed to the differentiation cocktail required for adipocyte maturation, AR adopted an antagonist conformation and was transcriptionally repressed. To further explore effectors within the cocktail, we applied an image-based support vector machine (SVM) classification scheme to show that adipocyte differentiation components inhibit AR action. The results demonstrate human adipocyte differentiation, via GR activation, upregulates AR but also inhibits AR transcriptional activity.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Androgen Receptor Antagonists/pharmacology , Glucocorticoids/pharmacology , Receptors, Androgen/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Androgen Receptor Antagonists/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dihydrotestosterone/pharmacology , Glucocorticoids/metabolism , Humans , Mice , Receptors, Glucocorticoid/metabolism , Structure-Activity Relationship , Support Vector Machine , Transcription, Genetic/drug effects
13.
Genes Nutr ; 7(1): 75-81, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21516479

ABSTRACT

Among the eight naturally occurring vitamin E analogs, γ-tocotrienol (GT3) is a particularly potent radioprophylactic agent in vivo. Moreover, GT3 protects endothelial cells from radiation injury not only by virtue of its antioxidant properties but also by inhibition of 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase and by improving the availability of the nitric oxide synthase cofactor tetrahydrobiopterin. Nevertheless, the precise mechanisms underlying the superior radioprotective properties of GT3 compared with other tocols are not known. This study, therefore, examined the differences in gene expression profiles between GT3 and its tocopherol counterpart, γ-tocopherol, as well as between GT3 and α-tocopherol in human endothelial cells. Cells were treated with vehicle or the appropriate tocol for 24 h, after which total RNA was isolated and genome-wide gene expression profiles were obtained using the Illumina platform. GT3 was far more potent in inducing gene-expression changes than α-tocopherol or γ-tocopherol. In particular, GT3 induced multiple changes in pathways known to be of importance in the cellular response to radiation exposure. Affected GO functional clusters included response to oxidative stress, response to DNA damage stimuli, cell cycle phase, regulation of cell death, regulation of cell proliferation, hematopoiesis, and blood vessel development. These results form the basis for further studies to determine the exact importance of differentially affected GO functional clusters in endothelial radioprotection by GT3.

14.
HPB (Oxford) ; 13(6): 369-76, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21609368

ABSTRACT

BACKGROUND: Epigenetics is a rapidly evolving field of genetic study applicable to nearly every aspect of genome-related research. The importance of epigenetics has been recognised in human hepatocellular carcinoma (HCC). Changes in DNA methylation patterns, including global hypomethylation and promoter hypermethylation, are thought to be early events in hepatocarcinogenesis. OBJECTIVES: This review aimed to summarise the role of epigenetics in HCC, to describe the mechanisms of epigenetic changes in HCC and to examine the clinical relevance of epigenetics in HCC. METHODS: This review examines the role of CpG-rich regions and DNA methylation, and describes an epigenetic model of cancer, tumour type-specific methylation, the relationships among methylation, cirrhosis and hepatocarcinogenesis, and the role of DNA methylation in HCC. The clinical implications of epigenetics in HCC are discussed. RESULTS: A multivariate predictor model based on traditional clinical factors and DNA methylation profile may have important applications in the early detection of neoplastic transformation in populations at high risk for HCC. CpG methylation may be valuable in HCC prognostics. DNA methylation profiles may enable clinical prediction in pre-therapy patient biopsies, paraffin-embedded samples or plasma DNA. CONCLUSIONS: Epigenetic changes and profiles may correlate to the biological behaviour of tumours and clinical outcome of HCC patients. The use of DNA methylation profiles as a surrogate biomarker remains an active area of clinical cancer research.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , DNA Methylation , Genetic Markers , Liver Neoplasms/genetics , Animals , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Transformation, Neoplastic/genetics , CpG Islands , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genetic Testing , Humans , Liver Cirrhosis/genetics , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Phenotype , Predictive Value of Tests , Prognosis
15.
Physiol Genomics ; 43(7): 325-45, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21224422

ABSTRACT

There is currently much interest in clinical applications of therapeutic hypothermia. Hypothermia can be a consequence of hypometabolism. We have recently established a procedure for the induction of a reversible deep hypometabolic state in mice using 5'-adenosine monophosphate (5'-AMP) in conjunction with moderate ambient temperature. The current study aims at investigating the impact of this technology at the gene expression level in a major metabolic organ, the liver. Our findings reveal that expression levels of the majority of genes in liver are not significantly altered by deep hypometabolism. However, among those affected by hypometabolism, more genes are differentially upregulated than downregulated both in a deep hypometabolic state and in the early arousal state. These altered gene expression levels during 5'-AMP induced hypometabolism are largely restored to normal levels within 2 days of the treatment. Our data also suggest that temporal control of circadian genes is largely stalled during deep hypometabolism.


Subject(s)
Adenosine Monophosphate/pharmacology , Liver/drug effects , Liver/metabolism , Animals , Female , Hypothermia/chemically induced , Hypothermia/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/metabolism
16.
J Endocrinol ; 207(2): 237-43, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20798131

ABSTRACT

Survivin (BIRC5) is a cell survival gene that is overexpressed in endometrial cancer and has been implicated to have a physiological role in normal endometrial function. To determine whether survivin gene expression is regulated by reproductive steroid hormones in the human endometrium, RNA was prepared from normal cycling women in the proliferative and secretory phases of the menstrual cycle. RNA was also isolated from 21 endometrial biopsies from premenopausal women at baseline and following 3 months of treatment with depot medroxyprogesterone acetate. Finally, RNA was isolated from endometrial biopsies from ten healthy postmenopausal women participating in a clinical trial of estrogen replacement therapy at baseline and following 6 months of treatment with conjugated equine estrogen. Quantitative RT-PCR analysis was used to determine survivin, insulin-like growth factor binding protein 1 (IGFBP1), Ki67, and IGF1 gene expression levels. Survivin gene expression was highest in the proliferative phase of the menstrual cycle and showed a statistically significant 4-fold increase in expression following chronic treatment with estrogens; this was strongly correlated with increased Ki67, a marker of proliferation. Survivin gene expression decreased 4.6-fold following chronic progestin treatment in the human endometrium. These data suggest that survivin transcript is regulated by estrogens and progestins in the disease-free human endometrium. The data also suggest that survivin transcript may be used as a biomarker of estrogen and progestin treatment efficacy, but validation studies must be conducted to support this conclusion.


Subject(s)
Endometrium/drug effects , Endometrium/metabolism , Estrogens/pharmacology , Microtubule-Associated Proteins/metabolism , Progestins/pharmacology , Adult , Female , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Middle Aged , Postmenopause , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survivin , Tissue Culture Techniques
17.
Anal Biochem ; 401(2): 288-94, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20227380

ABSTRACT

Wnts are secreted lipid-modified glycoproteins that carry out various signaling functions during development and in adult tissue. Wnt signaling is mediated by frizzled receptors (Fzds) at the cell surface and can be modulated by the secreted frizzled-related proteins (SFRPs) and other molecular antagonists. Abnormal Wnt signaling has been implicated in several diseases. However, due to the complexity of the Wnt signal and the lack of knowledge pertaining to the binding properties of different Wnt ligands, no therapeutic agents that target this pathway exist. Using a novel enzyme-linked immunosorbent assay (ELISA)-based technique, we were able to determine the first measurements of binding affinity for specific Wnt interactions. This study shows that purified Wnt3a, Wnt7a, and Wnt5a have different binding specificities for Fzds and SFRPs.


Subject(s)
Enzyme-Linked Immunosorbent Assay/methods , Frizzled Receptors/metabolism , Wnt Proteins/metabolism , Cell Line, Tumor , Endometrial Neoplasms/metabolism , Female , Humans , MAP Kinase Kinase 4/metabolism , Protein Binding , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Wnt3 Protein , Wnt3A Protein , beta Catenin/metabolism
18.
Radiat Res ; 173(4): 469-78, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20334519

ABSTRACT

The intestinal immune system is the largest in the body. This study analyzed changes in intestinal immune cell populations, cytokine protein levels, and transcript profiles after total-body irradiation (TBI) in CD2F1 mice. A single dose of 8.0 Gy gamma radiation caused negligible 30-day lethality but induced significant histological damage in jejunal mucosa that was maximal at 3.5 days and that had seemingly recovered by day 21 after irradiation. These changes were accompanied by decreased numbers of mucosal macrophages, neutrophils, and B and T lymphocytes, mostly coinciding with similar reductions in peripheral blood cell counts. Recovery of mucosal macrophages occurred within 1 week, whereas mucosal granulocytes and lymphocytes remained low until 3 weeks after TBI. Maximal suppression of T-helper cell (T(H))-related transcripts occurred at 3.5 days, but there was no obvious T(H)1 or T(H)2 bias. Genome-wide transcriptional profiling revealed a preponderance of differentially regulated genes involved in cell cycle control, cell death and DNA repair between 4 h and 3.5 days after irradiation. Genes involved in tissue recovery predominated from day 7 onward. We conclude that the intestinal immune system undergoes profound changes after sublethal TBI and that these changes likely contribute to postirradiation pathophysiological manifestations.


Subject(s)
Cytokines/immunology , Immunity, Innate/immunology , Immunity, Innate/radiation effects , Intestinal Mucosa/immunology , Intestinal Mucosa/radiation effects , Leukocytes/immunology , Leukocytes/radiation effects , Animals , Dose-Response Relationship, Radiation , Intestinal Mucosa/cytology , Male , Mice , Radiation Dosage , Survival Rate , Whole-Body Irradiation
19.
J Lipid Res ; 51(7): 1704-18, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20173184

ABSTRACT

LDL mediates transfection with plasmid DNA in a variety of cell types in vitro and in several tissues in vivo in the rat. The transfection capacity of LDL is based on apo B100, as arginine/lysine clusters, suggestive of nucleic acid-binding domains and nuclear localization signal sequences, are present throughout the molecule. Apo E may also contribute to this capacity because of its similarity to the Dengue virus capsid proteins and its ability to bind DNA. Synthetic peptides representing two apo B100 regions with prominent Arg/Lys clusters were shown to bind DNA. Region 1 (0014Lys-Ser0160) shares sequence motifs present in DNA binding domains of Interferon Regulatory Factors and Flaviviridae capsid/core proteins. It also contains a close analog of the B/E receptor ligand of apo E. Region 1 peptides, B1-1 (0014Lys-Glu0054) and B1-2 (0055Leu-Ala0096), mediate transfection of HeLa cells but are cytotoxic. Region 2 (3313Asp-Thr3431), containing the known B/E receptor ligand, shares analog motifs with the human herpesvirus 5 immediate-early transcriptional regulator (UL122) and Flaviviridae NS3 helicases. Region 2 peptides, B2-1 (3313Asp-Glu3355), and B2-2 (3356Gly-Thr3431) are ineffective in cell transfection and are noncytotoxic. These results confirm the role of LDL as a natural transfection vector in vivo, a capacity imparted by the apo B100, and suggest a basis for Flaviviridae cell entry.


Subject(s)
Apolipoprotein B-100 , DNA/metabolism , Lipoproteins, LDL/metabolism , Transfection/methods , Viral Proteins , Amino Acid Sequence , Animals , Apolipoprotein B-100/genetics , Apolipoprotein B-100/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Cell Line , DNA/genetics , Female , Flaviviridae/genetics , Flaviviridae/metabolism , Genes, Reporter , Humans , Lipoproteins, LDL/genetics , Molecular Sequence Data , Peptides/genetics , Peptides/metabolism , Rats , Rats, Sprague-Dawley , Tissue Distribution , Viral Proteins/genetics , Viral Proteins/metabolism
20.
Cancer Biol Ther ; 8(22): 2126-35, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19755863

ABSTRACT

Identification of biomarkers potentially provides prognostic information that can help guide clinical decision-making. Given the relationship between estrogen exposure and endometrial cancer, especially low grade endometrioid carcinoma, we hypothesized that high expression of genes induced by estrogen would identify low risk endometrioid endometrial cancers. cDNA microarray and qRT-PCR verification were used to identify six genes that are highly induced by estrogen in the endometrium. These estrogen-induced biomarkers were quantified in 72 endometrial carcinomas by qRT-PCR. Unsupervised cluster analysis was performed, with expression data correlated to tumor characteristics. Time to recurrence by cluster was analyzed using the Kaplan-Meier method. A receiver operating characteristic (ROC) curve was generated to determine the potential clinical utility of the biomarker panel to predict prognosis. Expression of all genes was higher in endometrioid carcinomas compared to non-endometrioid carcinomas. Unsupervised cluster analysis revealed two distinct groups based on gene expression. The high expression cluster was characterized by lower age, higher BMI, and low grade endometrioid histology. The low expression cluster had a recurrence rate 4.35 times higher than the high expression cluster. ROC analysis allowed for the prediction of stage and grade with a false negative rate of 4.8% based on level of gene expression in endometrioid tumors. We have therefore identified a panel of estrogen-induced genes that have potential utility in predicting endometrial cancer stage and recurrence risk. This proof-of-concept study demonstrates that biomarker analysis may play a role in clinical decision making for the therapy of women with endometrial cancer.


Subject(s)
Carcinoma, Endometrioid/genetics , Endometrial Neoplasms/genetics , Equilin/analogs & derivatives , Estrogen Replacement Therapy , Estrogens, Conjugated (USP)/pharmacology , Estrone/analogs & derivatives , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genetic Association Studies , Neoplasm Proteins/genetics , Adult , Age Factors , Aged , Aged, 80 and over , Biomarkers, Tumor , Body Mass Index , Carcinoma, Endometrioid/epidemiology , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Cluster Analysis , Endometrial Neoplasms/epidemiology , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Equilin/administration & dosage , Equilin/adverse effects , Equilin/pharmacology , Estrogen Replacement Therapy/adverse effects , Estrogens, Conjugated (USP)/adverse effects , Estrogens, Conjugated (USP)/therapeutic use , Estrone/administration & dosage , Estrone/adverse effects , Estrone/pharmacology , Female , Humans , Middle Aged , Neoplasm Proteins/biosynthesis , Oligonucleotide Array Sequence Analysis , Prognosis , ROC Curve , Randomized Controlled Trials as Topic , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...