Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Sensors (Basel) ; 21(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33477963

ABSTRACT

The continuous development of fifth-generation (5G) networks is the main driving force for the growth of Internet of Things (IoT) applications. It is expected that the 5G network will greatly expand the applications of the IoT, thereby promoting the operation of cellular networks, the security and network challenges of the IoT, and pushing the future of the Internet to the edge. Because the IoT can make anything in anyplace be connected together at any time, it can provide ubiquitous services. With the establishment and use of 5G wireless networks, the cellular IoT (CIoT) will be developed and applied. In order to provide more reliable CIoT applications, a reliable network topology is very important. Reaching a consensus is one of the most important issues in providing a highly reliable CIoT design. Therefore, it is necessary to reach a consensus so that even if some components in the system is abnormal, the application in the system can still execute correctly in CIoT. In this study, a protocol of consensus is discussed in CIoT with dual abnormality mode that combines dormant abnormality and malicious abnormality. The protocol proposed in this research not only allows all normal components in CIoT to reach a consensus with the minimum times of data exchange, but also allows the maximum number of dormant and malicious abnormal components in CIoT. In the meantime, the protocol can make all normal components in CIoT satisfy the constraints of reaching consensus: Termination, Agreement, and Integrity.

2.
Cancers (Basel) ; 11(9)2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31480804

ABSTRACT

The unsatisfactory real-world efficacy of the hypomethylating agent azacitidine in treating myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) has prompted us to investigate the hematological adverse events and host variables that may compromise the use of this epigenetic drug. Using the zebrafish, we found that azacitidine destroyed their myeloid precursors and impaired myeloid function by inhibiting antigen processing, allogeneic response and phagocytic activity, resulting in increased susceptibility to infection even by the normal flora E. coli. In addition, iron overload, a MDS-associated condition following repeated transfusions, exacerbated bacterial infection especially by V. vulnificus with known iron dependence. Furthermore, we show that the tp53M214K mutant zebrafish survived longer than the wild-type (WT) when challenged with bacteria following azacitidine treatment. This was attributed to the mutant's hematopoietic cells rather than its general genetic background, since the WT animals reconstituted with the tp53M214K mutant kidney marrow became more resistant to bacterial infection following treatment with azacitidine. The clinical relevance of our findings was indicated by a MDS case with severe azacitidine-induced bone marrow suppression and by the association of hyperferritinemia with bacteremia in azacitidine-treated patients, while tp53M214K-mediated resistance to azacitidine-induced myelosuppression may explain the survival advantage of malignant MDS and AML clones over their normal counterparts under azacitidine treatment. Together, we propose that myelosuppression, iron overload and TP53 mutations may represent the host variables that compromise the azacitidine efficacy.

3.
J Mol Endocrinol ; 62(3): R223-R237, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30620710

ABSTRACT

Breast cancer is a heterogeneous disease, and the complexity of breast carcinogenesis is associated with epigenetic modification. There are several major classes of epigenetic enzymes that regulate chromatin activity. This review will focus on the nine mammalian protein arginine methyltransferases (PRMTs) and the dysregulation of PRMT expression and function in breast cancer. This class of enzymes catalyse the mono- and (symmetric and asymmetric) di-methylation of arginine residues on histone and non-histone target proteins. PRMT signalling (and R methylation) drives cellular proliferation, cell invasion and metastasis, targeting (i) nuclear hormone receptor signalling, (ii) tumour suppressors, (iii) TGF-ß and EMT signalling and (iv) alternative splicing and DNA/chromatin stability, influencing the clinical and survival outcomes in breast cancer. Emerging reports suggest that PRMTs are also implicated in the development of drug/endocrine resistance providing another prospective avenue for the treatment of hormone resistance and associated metastasis. The complexity of PRMT signalling is further underscored by the degree of alternative splicing and the scope of variant isoforms (with distinct properties) within each PRMT family member. The evolution of PRMT inhibitors, and the ongoing clinical trials of PRMT inhibitors against a subgroup of solid cancers, coupled to the track record of lysine methyltransferases inhibitors in phase I/II clinical trials against cancer underscores the potential therapeutic utility of targeting PRMT epigenetic enzymes to improve survival outcomes in aggressive and metastatic breast cancer.


Subject(s)
Arginine/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Epigenesis, Genetic/genetics , Animals , Female , Humans , Methylation
4.
Endocrinology ; 158(3): 431-447, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27983866

ABSTRACT

Breast cancer is a heterogeneous disease and its complexity has hindered the development of efficacious treatments targeting all breast cancer subtypes. Many studies have linked the diversity of breast carcinogenesis and metastasis to aberrant epigenetic signaling and control. Here, we focus on the current state of the discipline and review the major epigenetic enzymes controlling chromatin structure and function in the context of breast cancer, including (1) DNA methyltransferases, (2) lysine methyltransferases and demethylases, (3) protein arginine methyltransferases, and (4) histone acetyltransferases and deacetylases. Moreover, therapeutic drugs targeting these epigenetic enzymes are rapidly emerging and/or undergoing clinical trials. Therefore, we discuss the pharmacological manipulation of epigenetic enzymes for breast cancer treatment and present new clinical and survival outcome analysis on epigenetic factors that have evaded analysis to date. Understanding and pharmacologically exploiting epigenetic regulation in breast cancer promises to be an essential aspect of next-generation drug development and adjuvant therapies targeting advanced disease and treatment-resistant tumors.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Epigenesis, Genetic , Animals , Carcinogenesis , DNA Modification Methylases/metabolism , Female , Histone Deacetylases/metabolism , Histone Demethylases/metabolism , Humans , Lysine Acetyltransferases/metabolism
5.
EBioMedicine ; 11: 101-117, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27568222

ABSTRACT

RORα is a member of the nuclear receptor (NR) superfamily and analysis of the (global) RORα-deficient mouse model revealed this NR has a role in glycemic control and fat deposition. Therefore, we generated an adipose-specific RORα 'gain of function' mouse model under the control of the fatty acid binding protein 4 (FABP4) promoter to elucidate the function of RORα in adipose tissue. The Tg-FABP4-RORα4 mice demonstrated a shift in fat distribution to non-adipose tissues when challenged with a high fat diet (HFD). Specifically, we observed a subcutaneous lipodystrophy, accompanied by hepatomegaly (fatty liver/mild portal fibrosis) and splenomegaly; in a background of decreased weight gain and total body fat after HFD. Moreover, we observed significantly higher fasting blood glucose and impaired clearance of glucose in Tg-FABP4-RORα4 mice. Genome wide expression and qPCR profiling analysis identified: (i) subcutaneous adipose specific decreases in the expression of genes involved in fatty acid biosynthesis, lipid droplet expansion and glycemic control, and (ii) the fibrosis pathway as the most significant pathway [including dysregulation of the collagen/extracellular matrix (ECM) pathways] in subcutaneous adipose and liver. The pathology presented in the Tg-FABP4-RORα4 mice is reminiscent of human metabolic disease (associated with aberrant ECM expression) highlighting the therapeutic potential of this NR.


Subject(s)
Adipose Tissue/metabolism , Adiposity/genetics , Blood Glucose , Gene Expression , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Adipose Tissue/immunology , Adipose Tissue/pathology , Adiposity/immunology , Animals , Biomarkers , Cluster Analysis , Extracellular Matrix/metabolism , Fibrosis , Gene Expression Profiling , Genotype , Glucose Tolerance Test , Hepatomegaly/genetics , Hepatomegaly/metabolism , Hepatomegaly/pathology , Humans , Insulin Resistance , Lipid Metabolism , Lipids/blood , Liver/metabolism , Liver/pathology , Mice , Mice, Transgenic , Organ Specificity , Phenotype , Splenomegaly/genetics , Splenomegaly/metabolism , Splenomegaly/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transgenes , Weight Gain
6.
EBioMedicine ; 6: 59-72, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27211549

ABSTRACT

We have previously reported that RORγ expression was decreased in ER-ve breast cancer, and increased expression improves clinical outcomes. However, the underlying RORγ dependent mechanisms that repress breast carcinogenesis have not been elucidated. Here we report that RORγ negatively regulates the oncogenic TGF-ß/EMT and mammary stem cell (MaSC) pathways, whereas RORγ positively regulates DNA-repair. We demonstrate that RORγ expression is: (i) decreased in basal-like subtype cancers, and (ii) inversely correlated with histological grade and drivers of carcinogenesis in breast cancer cohorts. Furthermore, integration of RNA-seq and ChIP-chip data reveals that RORγ regulates the expression of many genes involved in TGF-ß/EMT-signaling, DNA-repair and MaSC pathways (including the non-coding RNA, LINC00511). In accordance, pharmacological studies demonstrate that an RORγ agonist suppresses breast cancer cell viability, migration, the EMT transition (microsphere outgrowth) and mammosphere-growth. In contrast, RNA-seq demonstrates an RORγ inverse agonist induces TGF-ß/EMT-signaling. These findings suggest pharmacological modulation of RORγ activity may have utility in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA Repair , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Transforming Growth Factor beta/genetics , Benzamides/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplasm Metastasis , Piperazines/pharmacology , Propanols/pharmacology , Sequence Analysis, RNA , Signal Transduction
7.
Mol Endocrinol ; 30(6): 660-76, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27144290

ABSTRACT

Skeletal muscle remodels metabolic capacity, contractile and exercise phenotype in response to physiological demands. This adaptive remodeling response to physical activity can ameliorate/prevent diseases associated with poor diet and lifestyle. Our previous work demonstrated that skeletal muscle-specific transgenic expression of the neuron-derived orphan nuclear receptor, Nor-1 drives muscle reprogramming, improves exercise endurance, and oxidative metabolism. The current manuscript investigates the association between exercise, Nor-1 expression and the role of Nor-1 in adaptive remodeling. We demonstrate that Nor-1 expression is induced by exercise and is dependent on calcium/calcineurin signaling (in vitro and in vivo). Analysis of fatigue-resistant transgenic mice that express Nor-1 in skeletal muscle revealed increased hypertrophy and vascularization of muscle tissue. Moreover, we demonstrate that transgenic Nor-1 expression is associated with increased intracellular recycling, ie, autophagy, involving 1) increased expression of light chain 3A or LC3A-II, autophagy protein 5, and autophagy protein 12 in quadriceps femoris muscle extracts from Tg-Nor-1 (relative to Wild-type (WT) littermates); 2) decreased p62 expression indicative of increased autophagolysosome assembly; and 3) decreased mammalian target of rapamycin complex 1 activity. Transfection of LC3A-GFP-RFP chimeric plasmid demonstrated that autophagolysosome formation was significantly increased by Nor-1 expression. Furthermore, we demonstrated a single bout of exercise induced LC3A-II expression in skeletal muscle from C57BL/6 WT mice. This study, when combined with our previous studies, demonstrates that Nor-1 expression drives multiple physiological changes/pathways that are critical to the beneficial responses of muscle to exercise and provides insights into potential pharmacological manipulation of muscle reprogramming for the treatment of lifestyle induced chronic diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Physical Conditioning, Animal , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Calcineurin/metabolism , Calcium/metabolism , Cell Line , DNA-Binding Proteins/genetics , Hypertrophy , Lysosomes/drug effects , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Models, Biological , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/parasitology , Neovascularization, Physiologic/drug effects , Nerve Tissue Proteins/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Signal Transduction/drug effects , Sirolimus/pharmacology
8.
Am J Physiol Endocrinol Metab ; 308(2): E159-71, 2015 01 15.
Article in English | MEDLINE | ID: mdl-25424999

ABSTRACT

The Rar-related orphan receptor-α (Rorα) is a nuclear receptor that regulates adiposity and is a potential regulator of energy homeostasis. We have demonstrated that the Rorα-deficient staggerer (sg/sg) mice display a lean and obesity-resistant phenotype. Adaptive Ucp1-dependent thermogenesis in beige/brite and brown adipose tissue serves as a mechanism to increase energy expenditure and resist obesity. DEXA and MRI analysis demonstrated significantly decreased total fat mass and fat/lean mass tissue ratio in male chow-fed sg/sg mice relative to wt mice. In addition, we observed increased Ucp1 expression in brown adipose and subcutaneous white adipose tissue but not in visceral adipose tissue from Rorα-deficient mice. Moreover, this was associated with significant increases in the expression of the mRNAs encoding the thermogenic genes (i.e., markers of brown and beige adipose) Pparα, Errα, Dio2, Acot11/Bfit, Cpt1ß, and Cidea in the subcutaneous adipose in the sg/sg relative to WT mice. These changes in thermogenic gene expression involved the significantly increased expression of the (cell-fate controlling) histone-lysine N-methyltransferase 1 (Ehmt1), which stabilizes the Prdm16 transcriptional complex. Moreover, primary brown adipocytes from sg/sg mice displayed a higher metabolic rate, and further analysis was consistent with increased uncoupling. Finally, core body temperature analysis and infrared thermography demonstrated that the sg/sg mice maintained greater thermal control and cold tolerance relative to the WT littermates. We suggest that enhanced Ucp1 and thermogenic gene expression/activity may be an important contributor to the lean, obesity-resistant phenotype in Rorα-deficient mice.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Gene Expression Regulation/physiology , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Obesity/metabolism , Thermogenesis/physiology , Absorptiometry, Photon , Animals , Body Composition/physiology , Body Temperature/physiology , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Immunohistochemistry , Magnetic Resonance Imaging , Male , Mice , Mice, Neurologic Mutants , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Thermogenesis/genetics , Transcription Factors/metabolism , Uncoupling Protein 1
9.
IUBMB Life ; 65(8): 657-64, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23846999

ABSTRACT

Glycogen is an energy storage depot for the mammalian species. This review focuses on recent developments that have identified the role of nuclear hormone receptor (NR) signaling and epigenomic control in the regulation of important genes that modulate glycogen metabolism. Specifically, new studies have revealed that the NR4A subgroup (of the NR superfamily) are strikingly sensitive to beta-adrenergic stimulation in skeletal muscle, and transgenic studies in mice have revealed the expression of these NRs affects endurance and glycogen levels in muscle. Furthermore, other studies have demonstrated that one of the NR coregulator class of enzymes that mediate chromatin remodeling, the histone methyltransferases (for example, protein arginine methyltransferase 4) regulates the expression of several genes involved in glycogen metabolism and glycogen storage diseases in skeletal muscle. Importantly, NRs and histone methyltransferases, have the potential to be pharmacologically exploited and may provide novel targets in the quest to treat disorders of glycogen storage.


Subject(s)
Epigenomics , Glycogen/metabolism , Muscle, Skeletal/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Glycogen Storage Disease/physiopathology , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Protein-Arginine N-Methyltransferases/metabolism
10.
Biochem J ; 444(2): 323-31, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22428544

ABSTRACT

CARM1 (co-activator-associated arginine methyltransferase 1)/PRMT4 (protein arginine methyltransferase 4), functions as a co-activator for transcription factors that are regulators of muscle fibre type and oxidative metabolism, including PGC (peroxisome-proliferator-activated receptor γ co-activator)-1α and MEF2 (myocyte enhancer factor 2). We observed significantly higher Prmt4 mRNA expression in comparison with Prmt1-Prmt6 mRNA expression in mouse muscle (in vitro and in vivo). Transfection of Prmt4 siRNA (small interfering RNA) into mouse skeletal muscle C2C12 cells attenuated PRMT4 mRNA and protein expression. We subsequently performed additional qPCR (quantitative PCR) analysis (in the context of metabolism) to examine the effect of Prmt4 siRNA expression on >200 critical genes that control (and are involved in) lipid, glucose and energy homoeostasis, and circadian rhythm. This analysis revealed a strikingly specific metabolic expression footprint, and revealed that PRMT4 is necessary for the expression of genes involved in glycogen metabolism in skeletal muscle cells. Prmt4 siRNA expression selectively suppressed the mRNAs encoding Gys1 (glycogen synthase 1), Pgam2 (muscle phosphoglycerate mutase 2) and Pygm (muscle glycogen phosphorylase). Significantly, PGAM, PYGM and GYS1 deficiency in humans causes glycogen storage diseases type X, type V/McArdle's disease and type 0 respectively. Attenuation of PRMT4 was also associated with decreased expression of the mRNAs encoding AMPK (AMP-activated protein kinase) α2/γ3 (Prkaa2 and Prkag3) and p38 MAPK (mitogen-activated protein kinase), previously implicated in Wolff-Parkinson-White syndrome and Pompe Disease (glycogen storage disease type II). Furthermore, stable transfection of two PRMT4-site-specific (methyltransferase deficient) mutants (CARM1/PRMT4 VLD and CARM1E267Q) significantly repressed the expression of Gys1, Pgam2 and AMPKγ3. Finally, in concordance, we observed increased and decreased glycogen levels in PRMT4 (native)- and VLD (methylation deficient mutant)-transfected skeletal muscle cells respectively. This demonstrated that PRMT4 expression and the associated methyltransferase activity is necessary for the gene expression programme involved in glycogen metabolism and human glycogen storage diseases.


Subject(s)
Gene Expression Regulation/genetics , Glycogen/biosynthesis , Glycogen/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Protein-Arginine N-Methyltransferases/physiology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Protein-Arginine N-Methyltransferases/biosynthesis , Protein-Arginine N-Methyltransferases/genetics
11.
Physiol Genomics ; 43(4): 213-27, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21119012

ABSTRACT

We demonstrate that chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) mRNA is more abundantly expressed (than COUP-TFI mRNA) in skeletal muscle C2C12 cells and in (type I and II) skeletal muscle tissue from C57BL/10 mice. Consequently, we have utilized the ABI TaqMan Low Density Array (TLDA) platform to analyze gene expression changes specifically attributable to ectopic COUP-TFII (relative to vector only) expression in muscle cells. Utilizing a TLDA-based platform and 5 internal controls, we analyze the entire NR superfamily, 96 critical metabolic genes, and 48 important myogenic regulatory genes on the TLDA platform utilizing 5 internal controls. The low density arrays were analyzed by rigorous statistical analysis (with Genorm normalization, Bioconductor R, and the Empirical Bayes statistic) using the (integromics) statminer software. In addition, we validated the differentially expressed patho-physiologically relevant gene (identified on the TLDA platform) glucose transporter type 4 (Glut4). We demonstrated that COUP-TFII expression increased the steady state levels of Glut4 mRNA and protein, while ectopic expression of truncated COUP-TFII lacking helix 12 (COUP-TFΔH12) reduced Glut4 mRNA expression in C2C12 cells. Moreover, COUP-TFII expression trans-activated the Glut4 promoter (-997/+3), and ChIP analysis identified selective recruitment of COUP-TFII to a region encompassing a highly conserved SP1 binding site (in mouse, rat, and human) at nt positions -131/-118. Mutation of the SpI site ablated COUP-TFII mediated trans-activation of the Glut4 promoter. In conclusion, this study demonstrates that in skeletal muscle cells, COUP-TFII regulates several nuclear hormone receptors, and critical metabolic and muscle specific genes.


Subject(s)
COUP Transcription Factor II/metabolism , Gene Expression Regulation, Developmental , Muscle Cells/metabolism , Muscle Development/genetics , Muscle, Skeletal/cytology , Animals , COUP Transcription Factor II/genetics , Cell Line , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Male , Mice , Muscle Fibers, Skeletal/metabolism , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Transcriptional Activation/genetics
12.
Mol Cell Endocrinol ; 315(1-2): 146-52, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19631715

ABSTRACT

Estrogen-related receptors (ERRs) are constitutively active orphan nuclear receptors. Natural ligands have not been identified, however, recent reports have demonstrated the synthetic phenolic acyl hydrazone, GSK4716, functions as a selective ERRbeta/gamma agonist. We demonstrate that ERRbeta is transiently induced, and ERRgamma is dramatically induced (and accumulates) in a differentiation-dependent manner in skeletal muscle cells. Treatment of differentiated skeletal muscle cells with the ERRbeta/gamma agonist (GSK4716) produced a significant increase in the expression of GRalpha (isoform D) protein. Quantitative RT-PCR (Q-RT-PCR) analysis after treatment with GSK4716, revealed induction of the mRNAs encoding the glucocorticoid receptor (GR), 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1), the enzyme that converts inactive cortisone to cortisol and hexose-6-phosphate dehydrogenase expression (H6PDH) that stimulates oxoreduction by 11beta-HSD1. Candidate based expression profiling also demonstrated the mRNAs encoding characterized GR target genes, including C/EBP, ApoD and Monoamine oxidase-A (MAO-A) are induced in GSK4716 treated cells. In concordance with these observations, siRNA-mediated suppression of the mRNA encoding ERRgamma (but not ERRalpha and beta) attenuated the expression of mRNAs encoding GR, 11betaHSD1 and GR target gene(s). Similarly, treatment with the ERRgamma (and ERalpha) antagonist diethylstilbestrol (DES) suppressed glucocorticoid responsive gene expression in skeletal muscle cells. Interestingly, we observed that GSK4716 trans-activated GRE-TK-LUC in a GR-dependent manner. This study highlights the regulatory crosstalk between ERRgamma and GR signaling in skeletal muscle cells, and suggests the ERRgamma agonist modulates the expression of critical genes that control GR signaling and glucocorticoid sensitive gene expression.


Subject(s)
Gene Expression Regulation/drug effects , Glucocorticoids/metabolism , Hydrazines/pharmacology , Muscle, Skeletal , Receptors, Estrogen/agonists , Receptors, Estrogen/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Diethylstilbestrol/pharmacology , Estrogens, Non-Steroidal/pharmacology , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , RNA, Small Interfering/metabolism , Receptors, Estrogen/genetics , Receptors, Glucocorticoid/genetics , Signal Transduction/physiology
13.
Mol Cell Endocrinol ; 309(1-2): 101-8, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19465082

ABSTRACT

The nuclear hormone receptor (NR) 4A subgroup of orphan nuclear receptors includes three members, Nur77 (NR4A1), Nurr1 (NR4A2) and Nor-1 (NR4A3). Previously we have identified the rapid and robust (in vitro and in vivo) induction of the NR4A subgroup following beta-adrenergic stimulation in mouse skeletal muscle. This was concomitant with changes in the expression of genes involved in the regulation of nutrient metabolism. We have isolated mouse tissue of cardiovascular, endocrine and gastrointestinal origin at 1, 4, 8 and 24h after a single intraperitoneal injection of the beta-adrenergic agonist, isoprenaline. We similarly identified the significant induction (between 1 and 4h) of the NR4A genes in many of these tissues. Moreover, we have utilized TaqMan((R)) Low Density Arrays to determine the beta-adrenergic-sensitive metabolic gene expression in liver, white adipose and heart. In summary, cross-talk between beta-adrenergic and NR4A signaling occurs in several tissues, and is accompanied by modulation of metabolic gene expression.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Organ Specificity/genetics , Receptors, Adrenergic, beta/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Glucose/metabolism , Isoproterenol/pharmacology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1 , Nuclear Receptor Subfamily 4, Group A, Member 2 , Organ Specificity/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism
14.
J Biol Chem ; 283(26): 18411-21, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18441015

ABSTRACT

Homozygous staggerer mice (sg/sg) display decreased and dysfunctional retinoic acid receptor-related orphan receptor alpha (RORalpha) expression. We observed decreases in serum (and liver) triglycerides and total and high density lipoprotein serum cholesterol in sg/sg mice. Moreover, the sg/sg mice were characterized by reduced adiposity (associated with decreased fat pad mass and adipocyte size). Candidate-based expression profiling demonstrated that the dyslipidemia in sg/sg mice is associated with decreased hepatic expression of SREBP-1c, and the reverse cholesterol transporters, ABCA1 and ABCG1. This is consistent with the reduced serum lipids. The molecular mechanism did not involve aberrant expression of LXR and/or ChREBP. However, ChIP and transfection analyses revealed that RORalpha is recruited to and regulates the activity of the SREBP-1c promoter. Furthermore, the lean phenotype in sg/sg mice is also characterized by significantly increased expression of PGC-1alpha, PGC-1beta, and lipin1 mRNA in liver and white and brown adipose tissue from sg/sg mice. In addition, we observed a significant 4-fold increase in beta(2)-adrenergic receptor mRNA in brown adipose tissue. Finally, dysfunctional RORalpha expression protects against diet-induced obesity. Following a 10-week high fat diet, wild-type but not sg/sg mice exhibited a approximately 20% weight gain, increased hepatic triglycerides, and notable white and brown adipose tissue accumulation. In summary, these changes in gene expression (that modulate lipid homeostasis) in metabolic tissues are involved in decreased adiposity and resistance to diet-induced obesity in the sg/sg mice, despite hyperphagia. In conclusion, we suggest this orphan nuclear receptor is a key modulator of fat accumulation and that selective ROR modulators may have utility in the treatment of obesity.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , Lipids/chemistry , Obesity/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Animal Feed , Animals , COS Cells , Chlorocebus aethiops , Heterozygote , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 1 , Triglycerides/chemistry
15.
Int J Med Inform ; 76(1): 66-77, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16901749

ABSTRACT

OBJECTIVE: This paper presents a revised technology acceptance model to examine what determines mobile healthcare systems (MHS) acceptance by healthcare professionals. METHOD: Conformation factor analysis was performed to test the reliability and validity of the measurement model. The structural equation modeling technique was used to evaluate the causal model. RESULTS: The results indicated that compatibility, perceived usefulness and perceived ease of use significantly affected healthcare professional behavioral intent. MHS self-efficacy had strong indirect impact on healthcare professional behavioral intent through the mediators of perceived usefulness and perceived ease of use. Yet, the hypotheses for technical support and training effects on the perceived usefulness and perceived ease of use were not supported. CONCLUSION: This paper provides initial insights into factors that are likely to be significant antecedents of planning and implementing mobile healthcare to enhance professionals' MHS acceptance. The proposed model variables explained 70% of the variance in behavioral intention to use MHS; further study is needed to explore extra significant antecedents of new IT/IS acceptance for mobile healthcare. Such as privacy and security issue, system and information quality, limitations of mobile devices; the above may be other interesting factors for implementing mobile healthcare and could be conducted by qualitative research.


Subject(s)
Attitude of Health Personnel , Attitude to Computers , Computers, Handheld , Diffusion of Innovation , Health Care Sector/organization & administration , Models, Organizational , Telemedicine/organization & administration , Point-of-Care Systems/organization & administration , Taiwan
16.
Pharmacogenomics ; 7(7): 1017-24, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17054412

ABSTRACT

INTRODUCTION: Interferon taken alone or in combination with ribavirin can be used for the treatment of persons with chronic hepatitis C. It is highly desirable, both clinically and economically, to establish tools to distinguish responders from nonresponders and to predict possible outcomes of the treatments. In this work, our goal is to develop a prediction model resulting from the analysis of chronic hepatitis C patients' single nucleotide polymorphisms, viral genotype, viral load, age and gender, to predict the responsiveness of interferon combination treatment. MATERIALS AND METHODS: We collected blood samples from 523 chronic hepatitis C patients that had received interferon and ribavirin combination therapy. Based on the current treatment strategy for chronic hepatitis C patients, we focused our search for candidate genes involved in pathways related to interferon signaling and immunomodulation. With artificial neural network algorithms, we then developed pattern recognition methodologies to achieve predictions among the patients. The artificial neural network model was trained by an algorithm with an adaptive momentum and learning rate. RESULTS: There were seven single nucleotide polymorphisms selected from six candidate genes including adenosine deaminase-RNA-specific, caspase 5, interferon consensus sequence binding protein 1, interferon-induced protein 44, phosphoinositide-3-kinase catalytic gamma polypeptide and transporter 2 ATP-binding cassette subfamily B genes. We further applied the artificial neural network algorithms with these seven single nucleotide polymorphisms, viral genotype, viral load, age and gender information to build tools for predicting the responsiveness of interferon. Based on the fivefold cross-validation method to evaluate the performance, the model achieved a high success rate of prediction. CONCLUSION: We demonstrated that a trained artificial neural network model is a promising method for providing the inference from factors such as single nucleotide polymorphisms, viral genotype, viral load, age and gender to the responsiveness of interferon.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis C, Chronic/drug therapy , Interferons/therapeutic use , Neural Networks, Computer , Aging/physiology , Algorithms , Drug Therapy, Combination , Forecasting , Genotype , Hepacivirus/genetics , Humans , Immunologic Factors/pharmacology , Polymorphism, Single Nucleotide , Reproducibility of Results , Ribavirin/therapeutic use , Sex Characteristics , Signal Transduction/drug effects , Viral Load
17.
Cell Signal ; 17(7): 847-55, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15763427

ABSTRACT

Methylglyoxal (2-oxopropanal), a physiological glucose metabolite, is a highly reactive dicarbonyl compound that can induce stress in cells and cause apoptotic cell death. This study examines the early signaling effects of methylglyxal on renal cells. It was found that methylglyoxal caused a slow and sustained rise of intracellular Ca2+ concentration ([Ca2+]i) in a concentration-dependent manner (EC50=1.8 mM). Methylglyoxal also induced a [Ca2+]i rise when extracellular Ca2+ was removed, but the magnitude was reduced by 80%. Depletion of intracellular Ca2+ stores with thapsigargin (TG), an endoplasmic reticulum (ER) Ca2+ pump inhibitor, did not affect methylglyoxal's effect. In Ca2+-free medium, the methylglyoxal-induced [Ca2+]i rise was abolished by depleting stored Ca2+ with carbonylcyanide m-chlorophenylhydrazone (CCCP; a mitochondrial uncoupler). Methylglyoxal-caused [Ca2+]i rise in the Ca2+-containing medium was not affected by modulation of protein kinase C activity, presence of voltage-gated Ca2+ channel blockers, or preincubation with thiol-containing antioxidants. U73122, an inhibitor of phospholipase C, abolished ATP (but not methylglyoxal)-induced [Ca2+]i rise. Furthermore, the [Ca2+]i-elevating effect of methylglyoxal was cell type-dependent, because methylglyoxal failed to cause [Ca2+]i rises in CHO-K1, neutrophils, or platelets. Pretreatment with methylglyoxal for 0-24 h decreased cell viability in a concentration- and time-dependent manner. Meanwhile, methylglyoxal-induced cell death involved apoptotic and necrotic events, the former being the dominant. These findings suggest that methylglyoxal induced a significant rise in [Ca2+]i in Madin-Darby canine kidney (MDCK) renal tubular cells by stimulating both extracellular Ca2+ influx and CCCP-sensitive intracellular Ca2+ release via as yet unidentified mechanisms. The cell type-specific Ca2+ signaling may play an important role in the early process of cytotoxic action of methylglyoxal.


Subject(s)
Calcium/metabolism , Kidney Tubules/cytology , Pyruvaldehyde/metabolism , Animals , Apoptosis , CHO Cells , Calcium Signaling , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Dogs , Dose-Response Relationship, Drug , Fluorescent Dyes , Fura-2 , Humans , Intracellular Fluid/metabolism , Kidney Tubules/metabolism , Necrosis , Neutrophils/drug effects , Neutrophils/metabolism , Pyruvaldehyde/toxicity , Rabbits , Signal Transduction , Time Factors , Type C Phospholipases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL