Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Fluids Barriers CNS ; 15(1): 18, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29848382

ABSTRACT

BACKGROUND: In Alzheimer's disease, there are striking changes in CSF composition that relate to altered choroid plexus (CP) function. Studying CP tissue gene expression at the blood-cerebrospinal fluid barrier could provide further insight into the epithelial and stromal responses to neurodegenerative disease states. METHODS: Transcriptome-wide Affymetrix microarrays were used to determine disease-related changes in gene expression in human CP. RNA from post-mortem samples of the entire lateral ventricular choroid plexus was extracted from 6 healthy controls (Ctrl), 7 patients with advanced (Braak and Braak stage III-VI) Alzheimer's disease (AD), 4 with frontotemporal dementia (FTD) and 3 with Huntington's disease (HuD). Statistics and agglomerative clustering were accomplished with MathWorks, MatLab; and gene set annotations by comparing input sets to GeneGo ( http://www.genego.com ) and Ingenuity ( http://www.ingenuity.com ) pathway sets. Bonferroni-corrected hypergeometric p-values of < 0.1 were considered a significant overlap between sets. RESULTS: Pronounced differences in gene expression occurred in CP of advanced AD patients vs. Ctrls. Metabolic and immune-related pathways including acute phase response, cytokine, cell adhesion, interferons, and JAK-STAT as well as mTOR were significantly enriched among the genes upregulated. Methionine degradation, claudin-5 and protein translation genes were downregulated. Many gene expression changes in AD patients were observed in FTD and HuD (e.g., claudin-5, tight junction downregulation), but there were significant differences between the disease groups. In AD and HuD (but not FTD), several neuroimmune-modulating interferons were significantly enriched (e.g., in AD: IFI-TM1, IFN-AR1, IFN-AR2, and IFN-GR2). AD-associated expression changes, but not those in HuD and FTD, were enriched for upregulation of VEGF signaling and immune response proteins, e.g., interleukins. HuD and FTD patients distinctively displayed upregulated cadherin-mediated adhesion. CONCLUSIONS: Our transcript data for human CP tissue provides genomic and mechanistic insight for differential expression in AD vs. FTD vs. HuD for stromal as well as epithelial components. These choroidal transcriptome characterizations elucidate immune activation, tissue functional resiliency, and CSF metabolic homeostasis. The BCSFB undergoes harmful, but also important functional and adaptive changes in neurodegenerative diseases; accordingly, the enriched JAK-STAT and mTOR pathways, respectively, likely help the CP in adaptive transcription and epithelial repair and/or replacement when harmed by neurodegeneration pathophysiology. We anticipate that these precise CP translational data will facilitate pharmacologic/transgenic therapies to alleviate dementia.


Subject(s)
Alzheimer Disease/metabolism , Choroid Plexus/metabolism , Frontotemporal Dementia/metabolism , Huntington Disease/metabolism , Adult , Aged , Aged, 80 and over , Female , Gene Expression , Homeostasis/physiology , Humans , Male , Microarray Analysis , Middle Aged , Transcriptome
2.
Obesity (Silver Spring) ; 26(2): 332-339, 2018 02.
Article in English | MEDLINE | ID: mdl-29363287

ABSTRACT

OBJECTIVE: This study evaluated the effect of lorcaserin 10 mg twice daily (LOR BID), or with phentermine 15 mg once daily (LOR BID + PHEN QD) and 15 mg twice daily (LOR BID + PHEN BID), in conjunction with energy restriction on food cravings. METHODS: Two hundred and thirty-five patients without diabetes but with obesity or overweight and ≥ 1 comorbidity received LOR BID, LOR BID + PHEN QD, or LOR BID + PHEN BID for 12 weeks in a randomized double-blind study. The Food Craving Inventory (FCI) and the Control of Eating Questionnaire (COEQ) were administered over 12 weeks. RESULTS: The FCI total score and the subscale scores reduced from baseline in all groups. The least squares means (95% confidence intervals) for the total scores were -0.65 (-0.75 to -0.55), -0.75 (-0.84 to -0.65), and -0.84 (-0.95 to -0.74) in the LOR BID, LOR BID + PHEN QD, and LOR BID + PHEN BID groups, respectively. Cravings assessed by COEQ reduced from baseline in all groups. In general, the combination treatments were more effective than lorcaserin alone. At week 12, except for fruit juice and dairy products, general and specific cravings reduced in LOR BID + PHEN BID compared with LOR BID (P < 0.05). CONCLUSIONS: Lorcaserin in combination with phentermine improves control of food cravings during short-term energy restriction.


Subject(s)
Benzazepines/therapeutic use , Craving/drug effects , Obesity/drug therapy , Overweight/drug therapy , Phentermine/therapeutic use , Adolescent , Adult , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Benzazepines/pharmacology , Female , Humans , Male , Middle Aged , Phentermine/pharmacology , Young Adult
3.
Endocr Pract ; 24(2): 143-149, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29106816

ABSTRACT

OBJECTIVE: Assess efficacy, hypoglycemia, and weight gain in patients with type 2 diabetes (T2D) treated with insulin glargine 300 U/mL (Gla-300) or 100 U/mL (Gla-100) across different age groups. METHODS: Pooled data were generated for patients randomized to Gla-300 or Gla-100 in the EDITION 2 (NCT01499095) and 3 (NCT01676220) studies. In 4 age groups (<55, ≥55 to <60, ≥60 to <65, ≥65 years), glycated hemoglobin A1C (A1C), percentage of patients reaching A1C <7.5% (58 mmol/mol), weight change, confirmed hypoglycemia (blood glucose ≤70 mg/dL), and/or severe hypoglycemia (events requiring third-party assistance) were analyzed with descriptive statistics and logistic, binomial, and analysis of covariance regression modeling. RESULTS: A1C reductions from baseline and proportions of patients at target were similar for Gla-300 and Gla-100 across all age groups at 6 and 12 months, but hypoglycemia incidence and event rate were lower with Gla-300 at 6 (both P<.001) and 12 months ( P<.001 and P = .005, respectively). Patients on Gla-300 gained less weight than those on Gla-100 at 6 ( P = .027) and 12 months ( P = .021). Changes in weight and daily weight-adjusted insulin dose decreased with increasing age at 6 ( P<.001 and P = .017, respectively) and 12 months ( P<.001 and P = .011, respectively). CONCLUSION: Older patients with T2D may benefit from treatment with Gla-300, which is associated with a lower hypoglycemia rate and less weight gain with similar efficacy compared with Gla-100. ABBREVIATIONS: A1C = glycated hemoglobin A1C BMI = body mass index Gla-100 = insulin glargine 100 U/mL Gla-300 = insulin glargine 300 U/mL OAD = oral antidiabetes drug T2D = type 2 diabetes.


Subject(s)
Aging , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemia/chemically induced , Insulin Glargine/administration & dosage , Insulin Glargine/adverse effects , Weight Gain/drug effects , Adult , Aged , Aged, 80 and over , Aging/blood , Aging/drug effects , Aging/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/epidemiology , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Hypoglycemia/epidemiology , Incidence , Male , Middle Aged , Young Adult
4.
Sleep ; 40(6)2017 06 01.
Article in English | MEDLINE | ID: mdl-28419375

ABSTRACT

Study objective: To assess differences in gene expression in cholinergic basal forebrain cells between sleeping and sleep-deprived mice sacrificed at the same time of day. Methods: Tg(ChAT-eGFP)86Gsat mice expressing enhanced green fluorescent protein (eGFP) under control of the choline acetyltransferase (Chat) promoter were utilized to guide laser capture of cholinergic cells in basal forebrain. Messenger RNA expression levels in these cells were profiled using microarrays. Gene expression in eGFP(+) neurons was compared (1) to that in eGFP(-) neurons and to adjacent white matter, (2) between 7:00 am (lights on) and 7:00 pm (lights off), (3) between sleep-deprived and sleeping animals at 0, 3, 6, and 9 hours from lights on. Results: There was a marked enrichment of ChAT and other markers of cholinergic neurons in eGFP(+) cells. Comparison of gene expression in these eGFP(+) neurons between 7:00 am and 7:00 pm revealed expected differences in the expression of clock genes (Arntl2, Per1, Per2, Dbp, Nr1d1) as well as mGluR3. Comparison of expression between spontaneous sleep and sleep-deprived groups sacrificed at the same time of day revealed a number of transcripts (n = 55) that had higher expression in sleep deprivation compared to sleep. Genes upregulated in sleep deprivation predominantly were from the protein folding pathway (25 transcripts, including chaperones). Among 42 transcripts upregulated in sleep was the cold-inducible RNA-binding protein. Conclusions: Cholinergic cell signatures were characterized. Whether the identified genes are changing as a consequence of differences in behavioral state or as part of the molecular regulatory mechanism remains to be determined.


Subject(s)
Basal Forebrain/cytology , Cholinergic Neurons/metabolism , Gene Expression Profiling , Sleep Deprivation/metabolism , Sleep/genetics , Wakefulness/genetics , Acetylcholine/metabolism , Animals , CLOCK Proteins/genetics , Choline O-Acetyltransferase/genetics , Male , Mice , Protein Folding , Receptors, Metabotropic Glutamate/genetics , Sleep Deprivation/pathology
5.
Diabetes Obes Metab ; 19(2): 275-283, 2017 02.
Article in English | MEDLINE | ID: mdl-27767249

ABSTRACT

AIMS: To explore the treatment outcomes in adult patients with type 2 diabetes (T2D) enrolled in the GetGoal trials of lixisenatide (LIXI), and the predictive effects of baseline characteristics on outcomes. METHODS: This study was a pooled analysis of patient-level data from the LIXI GetGoal studies comparing LIXI and placebo. Patients were divided into baseline therapy groups: those receiving oral antidiabetes drugs (OADs) at baseline (n = 2760) or those receiving basal insulin at baseline (n = 1198). RESULTS: Compared with placebo, LIXI treatment led to significantly greater reductions in glycated haemoglobin (HbA1c), and greater achievement of the composite endpoint of HbA1c <7.0% (53 mmol/mol) with no symptomatic hypoglycaemia and no weight gain in either the OAD (34% vs 18%; P < .0001) or the basal insulin groups (19% vs 10%; P < .0001). Treatment with LIXI was associated with a greater percentage of patients experiencing a symptomatic hypoglycaemic event compared with placebo in both the OAD (5% vs 3%; P = .0098) and basal insulin groups (27% vs 17%; P < .0001). In assessing baseline factors that were predictors of treatment outcomes, only baseline HbA1c and LIXI treatment were strong predictors of outcomes in both the OAD and basal insulin groups. No other baseline characteristic had such a large or consistent clinically relevant predictive effect across treatment outcomes. CONCLUSIONS: The results from this study show that irrespective of baseline characteristics, LIXI treatment, as an add-on to OAD or basal insulin therapy, is effective in reducing HbA1c and achieving composite endpoints.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , Peptides/therapeutic use , Adult , Aged , Blood Glucose/metabolism , Clinical Trials, Phase III as Topic , Diabetes Mellitus, Type 2/metabolism , Drug Therapy, Combination , Female , Glycated Hemoglobin/metabolism , Humans , Hypoglycemia/chemically induced , Male , Middle Aged , Treatment Outcome , Weight Gain
6.
J Diabetes Complications ; 30(7): 1385-92, 2016.
Article in English | MEDLINE | ID: mdl-27267268

ABSTRACT

AIMS: To evaluate the impact of ß-cell function on the efficacy of lixisenatide, a once-daily prandial glucagon-like peptide-1 receptor agonist, in patients with type 2 diabetes (T2D). MATERIALS AND METHODS: In this post hoc analysis, patients from the Phase 3 GetGoal-M and GetGoal-S clinical trials randomized to lixisenatide 20µg once daily were stratified into quartiles by baseline ß-cell function, as measured by the secretory units of islet in transplantation (SUIT) index. RESULTS: Patients (N=437) were distributed evenly among SUIT index quartiles 1 to 4 (lowest to highest ß-cell function). Clinical outcomes improved from baseline across all SUIT quartiles; mean changes at week 24 were: glycated hemoglobin (HbA1c; % [mmol/mol]), -0.99 (-10.8), -0.87 (-9.5), -0.86 (-9.4), -0.83 (-9.1); and postprandial plasma glucose (PPG; mmol/L), -7.9, -5.6, -5.5, -4.3 (overall effect P<0.0001). Furthermore, postprandial glucagon was reduced in all SUIT quartiles, while insulinogenic index improved only in patients with higher baseline SUIT (overall effect P=0.0286). No severe symptomatic hypoglycemic events were reported. CONCLUSIONS: Lixisenatide treatment resulted in reductions in HbA1c and PPG levels across all SUIT quartiles. This suggests that non-insulin-related actions of lixisenatide contribute to improved glycemic control in T2D.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Insulin-Secreting Cells/drug effects , Peptides/therapeutic use , Aged , Female , Glycated Hemoglobin/analysis , Humans , Male , Middle Aged
7.
Sleep ; 37(10): 1589-600, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25197809

ABSTRACT

STUDY OBJECTIVES: To address whether changes in gene expression in blood cells with sleep loss are different in individuals resistant and sensitive to sleep deprivation. DESIGN: Blood draws every 4 h during a 3-day study: 24-h normal baseline, 38 h of continuous wakefulness and subsequent recovery sleep, for a total of 19 time-points per subject, with every 2-h psychomotor vigilance task (PVT) assessment when awake. SETTING: Sleep laboratory. PARTICIPANTS: Fourteen subjects who were previously identified as behaviorally resistant (n = 7) or sensitive (n = 7) to sleep deprivation by PVT. INTERVENTION: Thirty-eight hours of continuous wakefulness. MEASUREMENTS AND RESULTS: We found 4,481 unique genes with a significant 24-h diurnal rhythm during a normal sleep-wake cycle in blood (false discovery rate [FDR] < 5%). Biological pathways were enriched for biosynthetic processes during sleep. After accounting for circadian effects, two genes (SREBF1 and CPT1A, both involved in lipid metabolism) exhibited small, but significant, linear changes in expression with the duration of sleep deprivation (FDR < 5%). The main change with sleep deprivation was a reduction in the amplitude of the diurnal rhythm of expression of normally cycling probe sets. This reduction was noticeably higher in behaviorally resistant subjects than sensitive subjects, at any given P value. Furthermore, blood cell type enrichment analysis showed that the expression pattern difference between sensitive and resistant subjects is mainly found in cells of myeloid origin, such as monocytes. CONCLUSION: Individual differences in behavioral effects of sleep deprivation are associated with differences in diurnal amplitude of gene expression for genes that show circadian rhythmicity.


Subject(s)
Circadian Rhythm/physiology , Sleep Deprivation/blood , Sleep Deprivation/genetics , Adult , Attention/physiology , Biomarkers , Female , Gene Expression Profiling , Humans , Individuality , Male , Psychomotor Performance , Sleep/physiology , Sleep Deprivation/physiopathology , Time Factors , Wakefulness/physiology
8.
PLoS One ; 8(11): e79067, 2013.
Article in English | MEDLINE | ID: mdl-24223881

ABSTRACT

Previously, we showed that transient inhibition of TGF- ß1 resulted in correction of key aspects of diabetes-induced CD34(+) cell dysfunction. In this report, we examine the effect of transient inhibition of plasminogen activator inhibitor-1 (PAI-1), a major gene target of TGF-ß1 activation. Using gene array studies, we examined CD34(+) cells isolated from a cohort of longstanding diabetic individuals, free of microvascular complications despite suboptimal glycemic control, and found that the cells exhibited reduced transcripts of both TGF-ß1 and PAI-1 compared to age, sex, and degree of glycemic control-matched diabetic individuals with microvascular complications. CD34(+) cells from diabetic subjects with microvascular complications consistently exhibited higher PAI-1 mRNA than age-matched non-diabetic controls. TGF- ß1 phosphorodiamidate morpholino oligo (PMO) reduced PAI-1 mRNA in diabetic (p<0.01) and non-diabetic (p=0.05) CD34(+) cells. To reduce PAI-1 in human CD34(+) cells, we utilized PAI-1 siRNA, lentivirus expressing PAI-1 shRNA or PAI-1 PMO. We found that inhibition of PAI-1 promoted CD34(+) cell proliferation and migration in vitro, likely through increased PI3(K) activity and increased cGMP production. Using a retinal ischemia reperfusion injury model in mice, we observed that recruitment of diabetic CD34(+) cells to injured acellular retinal capillaries was greater after PAI-1-PMO treatment compared with control PMO-treated cells. Targeting PAI-1 offers a promising therapeutic strategy for restoring vascular reparative function in defective diabetic progenitors.


Subject(s)
Antigens, CD34/metabolism , Diabetes Mellitus/genetics , Leukocytes, Mononuclear/metabolism , Plasminogen Activator Inhibitor 1/genetics , Adult , Animals , Cell Movement/genetics , Cell Proliferation , Cells, Cultured , Cohort Studies , Cyclic GMP/metabolism , Diabetes Mellitus/blood , Diabetes Mellitus/metabolism , Diabetic Angiopathies/blood , Diabetic Angiopathies/genetics , Diabetic Angiopathies/metabolism , Humans , Leukocytes, Mononuclear/cytology , Mice , Middle Aged , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Plasminogen Activator Inhibitor 1/blood , RNA Interference , Reperfusion Injury/metabolism , Retinal Vessels/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/genetics , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/genetics
10.
Hum Mol Genet ; 21(1): 163-74, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21972245

ABSTRACT

Mutations in the leucine-rich repeat kinase 2 gene (LRRK2) are the most common genetic cause of Parkinson's disease (PD) and cause both autosomal dominant familial and sporadic PD. Currently, the physiological and pathogenic activities of LRRK2 are poorly understood. To decipher the biological functions of LRRK2, including the genes and pathways modulated by LRRK2 kinase activity in vivo, we assayed genome-wide mRNA expression in the brain and peripheral tissues from LRRK2 knockout (KO) and kinase hyperactive G2019S (G2019S) transgenic mice. Subtle but significant differences in mRNA expression were observed relative to wild-type (WT) controls in the cortex, striatum and kidney of KO animals, but only in the striatum in the G2019S model. In contrast, robust, consistent and highly significant differences were identified by the direct comparison of KO and G2019S profiles in the cortex, striatum, kidney and muscle, indicating opposite effects on mRNA expression by the two models relative to WT. Ribosomal and glycolytic biological functions were consistently and significantly up-regulated in LRRK2 G2019S compared with LRRK2 KO tissues. Genes involved in membrane-bound organelles, oxidative phosphorylation, mRNA processing and the endoplasmic reticulum were down-regulated in LRRK2 G2019S mice compared with KO. We confirmed the expression patterns of 35 LRRK2-regulated genes using quantitative reverse transcription polymerase chain reaction. These findings provide the first description of the transcriptional responses to genetically modified LRRK2 activity and provide preclinical target engagement and/or pharmacodynamic biomarker strategies for LRRK2 and may inform future therapeutic strategies for LRRK2-associated PD.


Subject(s)
Parkinson Disease/enzymology , Parkinson Disease/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic , Animals , Brain/enzymology , Female , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics
11.
Cancer Res ; 70(23): 9991-10001, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21118961

ABSTRACT

Identifying the functions of proteins, which associate with specific subnuclear structures, is critical to understanding eukaryotic nuclear dynamics. Sp100 is a prototypical protein of ND10/PML nuclear bodies, which colocalizes with Daxx and the proto-oncogenic PML. Sp100 isoforms contain SAND, PHD, Bromo, and HMG domains and are highly sumoylated, all characteristics suggestive of a role in chromatin-mediated gene regulation. A role for Sp100 in oncogenesis has not been defined previously. Using selective Sp100 isoform-knockdown approaches, we show that normal human diploid fibroblasts with reduced Sp100 levels rapidly senesce. Subsequently, small rapidly dividing Sp100 minus cells emerge from the senescing fibroblasts and are found to be highly tumorigenic in nude mice. The derivation of these tumorigenic cells from the parental fibroblasts is confirmed by microsatellite analysis. The small rapidly dividing Sp100 minus cells now also lack ND10/PML bodies, and exhibit genomic instability and p53 cytoplasmic sequestration. They have also activated MYC, RAS, and TERT pathways and express mesenchymal to epithelial transdifferentiation (MET) markers. Reintroduction of expression of only the Sp100A isoform is sufficient to maintain senescence and to inhibit emergence of the highly tumorigenic cells. Global transcriptome studies, quantitative PCR, and protein studies, as well as immunolocalization studies during the course of the transformation, reveal that a transient expression of stem cell markers precedes the malignant transformation. These results identify a role for Sp100 as a tumor suppressor in addition to its role in maintaining ND10/PML bodies and in the epigenetic regulation of gene expression.


Subject(s)
Antigens, Nuclear/genetics , Autoantigens/genetics , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Tumor Suppressor Proteins/genetics , Animals , Antigens, Nuclear/metabolism , Autoantigens/metabolism , Blotting, Western , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Cellular Senescence/genetics , Epithelial-Mesenchymal Transition/genetics , Fibroblasts/cytology , Gene Expression Profiling , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Promyelocytic Leukemia Protein , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Transplantation, Heterologous , Tumor Suppressor Proteins/metabolism , ras Proteins/metabolism
12.
Sleep ; 33(7): 889-900, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20614849

ABSTRACT

STUDY OBJECTIVES: Increases in ATP production machinery have been described in brain after 3 h of sleep deprivation. Whether this is sustained with longer durations of extended wakefulness is unknown. We hypothesized that energy depletion could be a mechanism leading to difficulty maintaining wakefulness and assessed changes in components of the electron transport chain. DESIGN: Protein levels of key subunits of complexes IV and V of the electron transport chain (COXI, COXIV, ATP5B) and uncoupling protein 2 (UCP2) in isolated mitochondria by Westerns in mouse cerebral cortex after 3 and 12 h of sleep deprivation were compared to that in control mice. Activity of complex IV enzyme and relevant transcription factors-Nrf1, Nrf2 (Gabp), and phosphorylation of AMP-dependent kinase (AMPK)-were also assessed. PARTICIPANTS: 8-10 week old C57BL/6J male mice (n = 91). INTERVENTIONS: 3, 6, and 12 h of sleep deprivation. MEASUREMENTS AND RESULTS: After both 3 and 12 h of sleep deprivation, complex IV proteins and enzyme activity were significantly increased. The complex V catalytic subunit was significantly increased after 12 h of sleep deprivation only. Increased levels of UCP2 protein after 12 h of sleep deprivation suggests that there might be alterations in the ATP/AMP ratio as wakefulness is extended. That phosphorylation of AMPK is increased after 6 h of sleep deprivation supports this assertion. The increase in Nrf1 and Nrf2 (Gabp) mRNA after 6 h of sleep deprivation provides a mechanism by which there is up-regulation of key proteins. CONCLUSIONS: There are complex dynamic changes in brain energy regulation with extended wakefulness.


Subject(s)
Cerebral Cortex/metabolism , Energy Metabolism , Sleep Deprivation/metabolism , Wakefulness , Animals , Blotting, Western , Cyclooxygenase 1/metabolism , Disease Models, Animal , Electron Transport Complex IV/metabolism , Ion Channels/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Time Factors , Transcription Factors/metabolism , Uncoupling Protein 2
13.
Cancer Res ; 69(24): 9202-10, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19951989

ABSTRACT

Early diagnosis of lung cancer followed by surgery presently is the most effective treatment for non-small cell lung cancer (NSCLC). An accurate, minimally invasive test that could detect early disease would permit timely intervention and potentially reduce mortality. Recent studies have shown that the peripheral blood can carry information related to the presence of disease, including prognostic information and information on therapeutic response. We have analyzed gene expression in peripheral blood mononuclear cell samples including 137 patients with NSCLC tumors and 91 patient controls with nonmalignant lung conditions, including histologically diagnosed benign nodules. Subjects were primarily smokers and former smokers. We have identified a 29-gene signature that separates these two patient classes with 86% accuracy (91% sensitivity, 80% specificity). Accuracy in an independent validation set, including samples from a new location, was 78% (sensitivity of 76% and specificity of 82%). An analysis of this NSCLC gene signature in 18 NSCLCs taken presurgery, with matched samples from 2 to 5 months postsurgery, showed that in 78% of cases, the signature was reduced postsurgery and disappeared entirely in 33%. Our results show the feasibility of using peripheral blood gene expression signatures to identify early-stage NSCLC in at-risk populations.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Leukocytes, Mononuclear/physiology , Lung Diseases/diagnosis , Lung Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Diagnosis, Differential , Early Detection of Cancer/methods , Female , Gene Expression Profiling , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lung Diseases/blood , Lung Diseases/genetics , Lung Diseases/immunology , Lung Neoplasms/blood , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Male , Middle Aged , Smoking/adverse effects
14.
BMC Syst Biol ; 3: 73, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19615063

ABSTRACT

BACKGROUND: The TGF-beta/SMAD pathway is part of a broader signaling network in which crosstalk between pathways occurs. While the molecular mechanisms of TGF-beta/SMAD signaling pathway have been studied in detail, the global networks downstream of SMAD remain largely unknown. The regulatory effect of SMAD complex likely depends on transcriptional modules, in which the SMAD binding elements and partner transcription factor binding sites (SMAD modules) are present in specific context. RESULTS: To address this question and develop a computational model for SMAD modules, we simultaneously performed chromatin immunoprecipitation followed by microarray analysis (ChIP-chip) and mRNA expression profiling to identify TGF-beta/SMAD regulated and synchronously coexpressed gene sets in ovarian surface epithelium. Intersecting the ChIP-chip and gene expression data yielded 150 direct targets, of which 141 were grouped into 3 co-expressed gene sets (sustained up-regulated, transient up-regulated and down-regulated), based on their temporal changes in expression after TGF-beta activation. We developed a data-mining method driven by the Random Forest algorithm to model SMAD transcriptional modules in the target sequences. The predicted SMAD modules contain SMAD binding element and up to 2 of 7 other transcription factor binding sites (E2F, P53, LEF1, ELK1, COUPTF, PAX4 and DR1). CONCLUSION: Together, the computational results further the understanding of the interactions between SMAD and other transcription factors at specific target promoters, and provide the basis for more targeted experimental verification of the co-regulatory modules.


Subject(s)
Chromatin/metabolism , Smad Proteins/metabolism , Algorithms , Animals , Base Sequence , Cell Line , Gene Expression Profiling , Genome , Humans , Immunoprecipitation , Mice , Models, Biological , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Rats , Reproducibility of Results , Sequence Analysis, DNA , Smad Proteins/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
15.
Neurobiol Aging ; 27(2): 351-60, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16399217

ABSTRACT

The impact of age on the enzymatic activities of adenosine metabolic enzymes, i.e., adenosine deaminase, adenosine kinase, cytosolic- and ecto-5'-nucleotidase have been assessed in the brain sleep/wake regulatory areas of young, intermediate and old rats (2, 12 and 24 months, respectively). There were significant spatial differences in the distribution of enzymes of adenosine metabolism in the brain. Age did not impact on the enzymatic activity of adenosine deaminase. Adenosine kinase activity increased significantly in the cerebral cortex of old animals. However, there were no differences in the activity of adenosine kinase between young and intermediate aged rats. The largest age-related changes were in the activity of cytosolic- and ecto-5'-nucleotidase and there was a significant age-related increase in the activity of these enzymes in the sleep/wake regulatory areas. In addition, the activity of cytosolic- and ecto-5'-nucleotidase increased in the cerebral cortex of old and intermediate age rats when compared to young animals. An increase in the enzymatic activities in the cerebral cortex of adenosine kinase and 5'-nucleotideases was accompanied by an increase in the level of their mRNA. An increase in the activity of 5'-nucleotideases with age likely leads to an increase in adenosine levels in the brain.


Subject(s)
Adenosine/metabolism , Aging/physiology , Brain/enzymology , Sleep/physiology , Wakefulness/physiology , 5'-Nucleotidase/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Adenosine Kinase/metabolism , Animals , Brain/anatomy & histology , Male , RNA, Messenger/biosynthesis , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction/methods
16.
Sleep ; 28(1): 21-7, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15700717

ABSTRACT

STUDY OBJECTIVES: Increased mRNA level of subunit 1 cytochrome c oxidase (COXI) during wakefulness and after short-term sleep deprivation has been described in brain. We hypothesized that this might contribute to increased activity of cytochrome oxidase (COX) enzyme during wakefulness, as part of the mechanisms to provide sufficient amounts of adenosine triphosphate to meet increased neuronal energy demands. DESIGN: COX activity was measured in isolated mitochondria from different brain regions in groups of rats with 3 hours of spontaneous sleep, 3 hours of spontaneous wake, and 3 hours of sleep deprivation. The group with 3 hours of spontaneous wake was added to delineate the circadian component of changes in the enzyme activity. Northern blot analysis was performed to examine the mRNA levels of 2 subunits of the enzyme COXI and COXIV, encoded by mitochondrial and nuclear DNA, respectively. SETTING: Laboratory of Biochemistry, Department of Animal Biology, and Center for Sleep and Respiratory Neurobiology, University of Pennsylvania. PARTICIPANTS: 2-month-old male Fischer rats (N = 21) implanted for polygraphic recording. MEASUREMENTS AND RESULTS: For COX activity, there was a main effect by analysis of variance of experimental group (P < .0001) with significant increases in COX activity in wake and sleep-deprived groups as compared to the sleep group. A main effect of brain region was also significant (P < .001). There was no difference between brain regions in the degree of increase in enzyme activity in wakefulness. Both COXI and COXIV mRNA were increased with wakefulness as compared to sleep. CONCLUSIONS: There is an increase in COX activity after both 3 hours of spontaneous wake and 3 hours of sleep deprivation as compared with 3 hours of spontaneous sleep in diverse brain regions, which could be, in part, explained by the increased levels of bigenomic transcripts of the enzyme. This likely contributes to increased adenosine triphosphate production during wakefulness. ABBREVIATIONS: ADP, adenosine diphosphate; ATP, adenosine triphosphate; COXI, cytochrome c oxidase subunit 1 mRNA; COX, cytochrome c oxidase (protein); CREB, cyclic AMP response element binding protein; DNA, deoxyribonucleic acid; EDTA, ethylenediaminetetraacetic acid; EEG, electroencephalography; EMG, electromyography; GABP, GA binding protein; HEPES, 4-(2-hydroxyethyl)piperazine-1-ethanesulfonic acid; mRNA, messenger ribonucleic acid; NADH, nicotinamid adenine dinucleotide, reduced; NDII, NADH dehydrogenase subunit 2 mRNA; NRF, nuclear respiratory factor.


Subject(s)
Brain/enzymology , Electron Transport Complex IV/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Blotting, Northern , Brain/cytology , Brain/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , DNA, Complementary/metabolism , Electroencephalography , Electromyography , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Mitochondria/enzymology , RNA, Messenger/metabolism , Rats , Rats, Inbred F344
17.
J Neurochem ; 85(2): 348-57, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12675911

ABSTRACT

Adenosine plays a role in promoting sleep, an effect that is thought to be mediated in the basal forebrain. Adenosine levels vary in this region with prolonged wakefulness in a unique way. The basis for this is unknown. We examined, in rats, the activity of the major metabolic enzymes for adenosine - adenosine deaminase, adenosine kinase, ecto- and cytosolic 5'-nucleotidase - in sleep/wake regulatory regions as well as cerebral cortex, and how the activity varies across the day and with sleep deprivation. There were robust spatial differences for the activity of adenosine deaminase, adenosine kinase, and cytosolic and ecto-5'-nucleotidase. However, the basal forebrain was not different from other sleep/wake regulatory regions apart from the tuberomammillary nucleus. All adenosine metabolic enzymes exhibited diurnal variations in their activity, albeit not in all brain regions. Activity of adenosine deaminase increased during the active period in the ventrolateral pre-optic area but decreased significantly in the basal forebrain. Enzymatic activity of adenosine kinase and cytosolic-5'-nucleotidase was higher during the active period in all brain regions tested. However, the activity of ecto-5'-nucleotidase was augmented during the active period only in the cerebral cortex. This diurnal variation may play a role in the regulation of adenosine in relationship to sleep and wakefulness across the day. In contrast, we found no changes specifically with sleep deprivation in the activity of any enzyme in any brain region. Thus, changes in adenosine with sleep deprivation are not a consequence of alterations in adenosine enzyme activity.


Subject(s)
Adenosine/metabolism , Brain/enzymology , Circadian Rhythm/physiology , Enzymes/metabolism , Sleep Deprivation/enzymology , 5'-Nucleotidase/metabolism , Adenosine Deaminase/metabolism , Adenosine Kinase/metabolism , Animals , Male , Nucleotidases/metabolism , Rats , Rats, Inbred F344 , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...