Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Oct 2.
Published in final edited form as: Endocrinology. 2007 Feb 15;148(5):2209–2225. doi: 10.1210/en.2006-0790

A Microarray Analysis of the Temporal Response of Liver to Methylprednisolone: A Comparative Analysis of Two Dosing Regimens

Richard R Almon 1, Debra C DuBois 1, William J Jusko 1
PMCID: PMC4183266  NIHMSID: NIHMS630857  PMID: 17303664

Abstract

Microarray analyses were performed on livers from adrenalectomized male Wistar rats chronically infused with methylprednisolone (MPL) (0.3 mg/kg·h) using Alzet mini-osmotic pumps for periods ranging from 6 h to 7 d. Four control and 40 drug-treated animals were killed at 10 different times during drug infusion. Total RNA preparations from the livers of these animals were hybridized to 44 individual Affymetrix REA230A gene chips, generating data for 15,967 different probe sets for each chip. A series of three filters were applied sequentially. These filters were designed to eliminate probe sets that were not expressed in the tissue, were not regulated by the drug, or did not meet defined quality control standards. These filters eliminated 13,978 probe sets (87.5%) leaving a remainder of 1989 probe sets for further consideration. We previously described a similar dataset obtained from animals after administration of a single dose of MPL (50 mg/kg given iv). That study involved 16 time points over a 72-h period. A similar filtering schema applied to the single-bolus-dose data-set identified 1519 probe sets as being regulated by MPL. A comparison of datasets from the two different dosing regimens identified 358 genes that were regulated by MPL in response to both dosing regimens. Regulated genes were grouped into 13 categories, mainly on gene product function. The temporal profiles of these common genes were subjected to detailed scrutiny. Examination of temporal profiles demonstrates that current perspectives on the mechanism of glucocorticoid action cannot entirely explain the temporal profiles of these regulated genes.


Glucocorticoids are a class of steroid hormones that play a central role in regulating the production, storage, and distribution of substrates for systemic energy metabolism. Most tissues are targets for glucocorticoid action and contribute in some way to their wide-ranging physiological effects. Synthetic glucocorticoids (corticosteroids) are used therapeutically for a wide variety of conditions that require immune and/or inflammatory modulation. Because corticosteroids pharmacologically magnify the physiological actions of endogenous glucocorticoids, therapeutic use of this class of drugs is accompanied by a wide range of adverse effects that include hyperglycemia, dyslipidemia, muscle wasting, hypertension, nephropathy, fatty liver, and an increased risk of arteriosclerosis (15). The physiological and pharmacological effects of these drugs are complex and involve changes in the expression of many genes in multiple tissues.

Microarrays can provide a method of high-throughput data collection that is necessary for constructing comprehensive information on the transcriptional basis of such complex systemic polygenic phenomena. When microarrays are used in a rich in vivo time series, they yield temporal patterns of changes in gene expression that illustrate the cascade of molecular events that cause broad systemic responses. However, the magnitude of data produced in such studies provides challenges of data mining and analysis.

Previously, we described the mining and analysis of microarray time series illustrating the responses of liver, skeletal muscle, and kidney taken from the same set of animals to a single bolus dose of the corticosteroid methylprednisolone (MPL) (68). These time series included individual chips from multiple control animals as well as multiple animals at each of 16 times over a 72-h period after bolus dosing with MPL. Because these experiments were initiated using adrenalectomized animals, the drug in essence acts as a stimulus that perturbs the homeostatic balance of the system, and the experiment monitored the deviation of the system and its return to the original state. Mining such time series datasets presents uniquely different problems from those encountered when microarrays are used to distinguish one group from another (e.g. cancerous vs. noncancerous tissues) (911). For this type of application, one attempts to define a pattern or fingerprint that distinguishes such groups with very high probability, and need not include all differentially regulated genes. In those cases, it is identifying a distinguishing pattern of gene expression rather than the relationship between the genes that is the important focus. In mining a time series microarray dataset, the problem is sorting through the vast amount of data to identify probe sets with temporal patterns of change in expression that indicate that the gene is regulated over time. In this case, the mechanistic relationships between the genes whose expression is changing in response to the stimulus are of paramount importance. For example, the stimulus may change the expression of a particular transcription factor that in turn alters the expression of downstream genes. For this application, the goal of the initial data mining is to avoid discarding valuable data. This is of particular importance because in our hands, each gene that is identified as being potentially regulated becomes the subject of extensive literature searches to allow placement into a temporal context of all other regulated genes.

Although very useful, a single time series only provides a one-dimensional view of the dynamics of the system in response to the stimulus. A pharmacological time series is different from most time series studies (for example those assessing developmental changes) in that it can be repeated using a different dosing regimen. A second dosing regimen is valuable in two regards. First, it can serve to corroborate results of the first dosing regimen. When considering gene array results, this can be useful. Second, the results can be used to group genes into clusters with common mechanisms of regulation. If two or more genes have a common mechanism of regulation, then their response profiles should be the same regardless of the dosing regimen. In the present report, we describe the use of microarrays to broadly characterize the response of liver to a second dosing regimen that entailed chronic infusion of MPL. Here the drug essentially was used as an unbalancing stimulus, and the experiment evaluated the capacity of the system to rebalance in the continuous presence of the drug. This dataset was mined using a similar filtration approach as was applied to the acute dosing dataset, and results from both datasets were compared. Probe sets common to both analyses were identified, allowing the coincidental evaluation of the two profiles for each gene.

Materials and Methods

Animals

Adrenalectomized male Wistar rats with body weights of 339 ± 28 (SD) g were used in the study. All animals were housed in our University Laboratory Animal Facility maintained under constant temperature (22 C) and humidity with a controlled 12-h light, 12-h dark cycle. A time period of at least 2 wk was allowed before they were prepared for surgery. Rats had free access to rat chow and 0.9% NaCl drinking water. This research adheres to Principles of Laboratory Animal Care (National Institutes of Health publication 85-23, revised 1985) and was approved by the Institutional Animal Care and Use Committee of the State University of New York at Buffalo.

Forty rats were administered 0.3 mg/kg·h infusions of MPL sodium succinate (Solu-Medrol; The Upjohn Co., Kalamazoo, MI) reconstituted in supplied diluent. The infusions were administered via Alzet osmotic pumps (model 2001, flow rate 1 μl/h; Alza, Palo Alto, CA). The pump drug solutions were prepared for each rat based on its predose body weight. On the day of implantation, rats were anesthetized using 60 – 80 mg/kg ketamine and 8 –10 mg/kg xylazine im. Pumps were sc implanted between the shoulder blades on the back. Rats were killed at various times up to 7 d. The time points included were 6, 10, 13, 18, 24, 36, 48, 72, 96, and 168 h. A control group of four animals was implanted with a saline-filled pump and killed at various times throughout the 7-d study period. A more detailed description of the experiment can be found in previous reports (12, 13).

Microarrays

Liver samples from each animal were ground into a fine powder in a mortar cooled by liquid nitrogen and 100 mg was added to 1 ml prechilled Trizol reagent (Invitrogen, Carlsbad CA). Total RNA extractions were carried out according to manufacturer’s directions and were further purified by passage through RNeasy mini-columns (QIAGEN, Valencia, CA) according to manufacturer’s protocols for RNA clean-up. Final RNA preparations were resuspended in RNase-free water and stored at −80 C. The RNAs were quantified spectrophotometrically, and purity and integrity were assessed by agarose gel electrophoresis. All samples exhibited 260/280 absorbance ratios of approximately 2.0, and all showed intact ribosomal 28S and 18S RNA bands in an approximate ratio of 2:1 as visualized by ethidium bromide staining. Isolated RNA from each liver sample was used to prepare target according to manufacturer’s protocols. The biotinylated cRNAs were hybridized to 44 individual Affymetrix GeneChips Rat Genome 230A (Affymetrix, Inc., Santa Clara, CA), which contained 15,967 probe sets. These gene chips contain over 7000 more probe sets than the ones used (U34A) in our previous liver bolus-dose MPL study (68). The high reproducibility of in situ synthesis of oligonucleotide chips allows accurate comparison of signals generated by samples hybridized to separate arrays.

Data analysis

Affymetrix Microarray Suite 5.0 (Affymetrix) was used for initial data acquisition and analysis. The signal intensities were normalized for each chip using a distribution of all genes around the 50th percentile. The dataset was then loaded into a data mining program, GeneSpring 7.0 (Silicon Genetics, Redwood City, CA), for further analysis. The generated dataset has been submitted to the National Center for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/projects/geo/) database (GDS972). Before screening for probe sets with altered expression levels, the dataset was normalized again to the mean values of control samples so that all probe sets from control samples had a mean value of 1, and probe sets from treated animals had a value of either greater than, less than, or equal to 1, representing up-regulation, down-regulation, or no change. These normalized probe sets were then filtered with a series of predetermined screening criteria to identify probe sets with appreciable expression levels, expression changes, and acceptable data quality. This set of filters is approximately the same as used to analyze the acute bolus dosing datasets with minor modifications to accommodate the different number of samples in the two experiments (68).

The process of data mining was performed in the GeneSpring program, and the progress after each step was visualized using a cluster feature of the program. This cluster feature could rearrange the order of the probe sets and group them based on the similarity (Pearson’s correlation) of their expression dynamics. Then the probe sets that were not eliminated by the filter were displayed vertically as a gene tree, and their expression dynamics over time were displayed horizontally in colors with yellow in the graph representing an expression ratio around 1, or no change. The color progressing toward red indicates a normalized value greater than 1, or up-regulation, and the color toward blue indicates a value less than 1, or down-regulation from control levels. The brightness of the colors reflects the original signal intensities or expression levels before normalization. The more abundantly expressed mRNAs exhibit brighter colors. Figure 1 shows the gene tree of the entire dataset (15,967 probe sets). The x-axis represents the 11 time points including vehicle controls (nominally time 0). The y-axis represents the list of the probe sets in order of similarity. Figure 2 (top) shows a magnification of five probe sets on the tree with an apparent response of enhanced expression. It should be noted that two of these probe sets directly adjacent to each other, 1370200 and 1387878, are both for the enzyme glutamate dehydrogenase 1 (Glud1). This enzyme is involved in ammonia detoxification, which is necessary when amino acid carbon is used for gluconeogenesis (14). A significant deficit of the gene tree representation is that all time intervals are represented as equal, and therefore to some degree temporal patterns are misrepresented in the gene tree presentations. This is illustrated by comparing the top and bottom panels in Fig. 2. The bottom panel shows a linear plot of the data for the two probe sets for Glud1 presented in the top panel. As illustrated by this example, it is possible to visually identify genes under regulation using gene trees. However, this approach does not entail objective criteria for selection of probes for further consideration.

Fig. 1.

Fig. 1

Gene tree representation of all probe sets (15,967) on individual Affymetrix R230A gene chips hybridized to total RNA prepared from livers taken from animals treated chronically with MPL (0.3 mg/kg·h) for periods ranging from 6 –168 h. The values for each individual probe set at each time point were normalized to the mean value of that probe set for time zero controls. The x-axis represents the 11 time points, including time zero controls. The y-axis presents the list of probe sets grouped by similarity using Pearson’s correlation. Yellow indicates no change from controls, red indicates probe sets with enhanced expression relative to controls, and blue indicates suppressed expression relative to controls.

Fig. 2.

Fig. 2

The top panel provides a magnification of five probe sets selected from Fig. 1 that show apparent enhanced regulation by MPL. Two of these probe sets represent the same gene (Glud1). The linear plots for both Glud1 probe sets are presented in the lower panel.

To screen for the probe sets objectively, the entire dataset was filtered with criteria similar to the ones applied to the dataset from a bolus-dose MPL experiment (68). This approach does not select for probe sets but rather eliminates those probe sets that do not meet certain criteria, leaving the remainder for further consideration. The first filter was designed to eliminate probe sets for genes that are not expressed in the liver. This filter employed a function in the Affymetrix Microarray Suite 5.0. During initial data analysis, a call of present (P), absent (A), or marginal (M) for each probe set on each chip was determined based on the intensity comparison of the matched and mismatched probe sequence pairs. The first filter eliminated all probe set that did not have a call of P on at least four of the 44 chips. This filter eliminated 6668 probe sets, leaving a remainder of 9299 probe sets for further consideration. These genes are more likely to be expressed in rat liver than those that were eliminated.

The second level of filtering that we applied was designed to eliminate probe sets that could not meet the basic criterion of a regulated probe. Specifically, this filtering approach was designed to eliminate probe sets whose average did not deviate from baseline by a certain value for a reasonable number of time points and employed two filters that were designed to eliminate probe sets that were neither down- nor up-regulated. The first of these filters eliminated probe sets that could not meet a minimal criterion for down-regulation. Starting with the 4P filtered list, we eliminated all probe sets that did not have average values less than 0.65 in at least two conditions (time points). Those probe sets that were not eliminated by this filter were retained as potentially down-regulated probe sets. The next filter was designed to eliminate probe sets that could not meet a minimal criterion for up-regulation. Starting with the 4P filtered list, we eliminated all probe sets that did not have average values above 1.5 in at least two conditions (time points). Those probe sets that were not eliminated by this filter were retained as potentially up-regulated probe sets. However, there were a small number of probe sets that were not eliminated by either filter. Using a Venn diagram, we removed these from both lists and created a list of probe sets with potential complex regulation. Probe sets not eliminated by this filtering approach included 1581 potential down-regulated probe sets, 1212 potential up-regulated probe sets, and a group of 85 probe sets that met both criteria.

The last filter we applied addressed the quality of the data. For this quality control filter, we eliminated probe sets that did not meet two conditions. The first condition focused on the control chips. As indicated above, our initial operation was to divide the value of each individual probe set on each chip by the mean of the values for that probe set on the four control chips. Therefore, the quality of the control data for each particular probe set is of unique importance in defining regulation by the drug. This filter eliminated probe sets whose control values exhibited coefficients of variation of greater than 50%. The second condition focused on the remaining 10 time points. This filter also eliminated probe sets with coefficients of variation for more than five of the remaining 10 time points exceeded 50%. After the application of this filter, 1989 probe sets remained for consideration. Of the 1989, 1049 were in the up-regulated list, 922 were in the down-regulated list, and 18 were in the list that met both criteria. Figure 3 shows a gene tree of all 1989 remaining probe sets. The three lists are published as supplemental data on The Endocrine Society’s Journals Online web site at http://endo.endojournals.org.

Fig. 3.

Fig. 3

Gene tree representation of probe sets remaining (1989) after filtering.

Results

Figure 4 (left) presents concentrations of MPL in plasma of animals receiving chronic glucocorticoid administration through Alzet pumps. The infusion dose (0.3 mg/kg·h) was chosen for dose equivalency at 168 h (the final time point in this study) with the single 50 mg/kg dose employed in our acute studies. By 6 h, MPL levels reach a stable steady state that is maintained throughout the 7-d infusion period. Preliminary experiments (data not shown) demonstrated that steady-state drug levels were attained by 6 h after pump implantation. Therefore, 6 h was chosen as the initial time point for these studies. In contrast, single bolus dose administration (right) results in drug levels that dissipate in a biexponential fashion and reach below the level of detection by 7 h after drug administration. Analyses of MPL kinetics for both acute and chronic dosing have been described previously (12, 15). In addition, pharmacokinetic/pharmacodynamic relationships for the expression of both tyrosine aminotransferase (TAT) and phosphoenolpyruvate carboxykinase (PEPCK) activities and mRNA levels assessed by Northern hybridization have been previously published for both the acute and chronic dose regimens (15, 16).

Fig. 4.

Fig. 4

MPL concentrations in rat plasma after chronic (left) and acute (right) administration of drug. MPL concentrations were determined by normal-phase HPLC analysis of plasma samples obtained from individual animals.

Data mining of gene arrays from this chronic MPL treatment series identified 1989 MPL-regulated probe sets on the R230A gene chips used in this experiment. We also previously obtained liver samples from a population of animals after administration of a single dose of MPL. Liver samples from those animals were collected in a time series that involved 16 time points over a 72-h period. RNAs from those livers were applied to the older Affymetrix RU34A chip. A similar filtering schema as applied to that dataset identified 1519 probe sets as being regulated by MPL. Using Affymetrix homology tables and Blast searches, we identified 464 probe sets of the 1519 on the U34A chip that corresponded to 417 of the 1989 identified probe sets on the U230A chip. Because both chips in some cases contain multiple probe sets for the same gene, and because there is a higher degree of probe set redundancy on the older U34 chip, the number of corresponding probe sets common to the two chips are not equal. Likewise, the number of genes actually represented in this common set is less than the number of probe sets. We identified 358 genes that were regulated by MPL in response to both dosing regimens. Most likely this list does not contain all genes regulated by MPL in liver. A perusal of both data-sets indicates that there were many probe sets that failed the quality control filter on one of the two chips and were thus eliminated. Nonetheless, these 358 genes have a very high degree of certainty of being regulated by MPL in the liver. In addition, the two profiles taken together provide an important foundation for understanding the mechanisms underlying the drug’s regulation of genes in the liver.

Response profiles

Of the 358 genes, profiles for 109 showed enhanced regulation after both dosing regimens. A reasonable hypothesis is that if different genes are responding by the same mechanism of regulation, then their profiles should be the same in response to these two different dosing regimens. However, examination of individual profiles demonstrates that this is not always the case. As an example, Fig. 5 shows response profile exemplars of two different genes with enhanced expression to the two dosing regimens but that differ from each other in response to MPL. The first gene (Fig. 5, left) is ornithine decarboxylase 1 (Odc1), the first enzyme in polyamine biosynthesis (17). This gene exhibits tolerance, a phenomenon we have earlier described for the enzyme TAT in liver (12, 18). Specifically, this gene almost recovers to its baseline after a period of time in response to chronic MPL infusion despite continuous presence of the drug (12). In various earlier reports, we presented data demonstrating that the glucocorticoid receptor (GR) is strongly down-regulated in response to MPL (12, 13, 15, 18, 19). Because GR mediates the effect of the drug, the large reduction in this effector molecule should greatly reduce the driving force for changes in gene expression, thus reducing the effect of the drug. The second gene (Fig. 5, right) showing enhanced expression is tryptophan 2,3-dioxygenase (Tdo2), the first enzyme in the kynurenine pathway (20). For this gene, the enhanced expression in response to the single bolus dose is more sustained than Odc1. However, the chronic time profile shows a second and higher period of enhanced expression that continues throughout the entire 168-h infusion period. These data illustrate that enhanced expression to corticosteroids probably involves multiple mechanisms and that our initial classification of enhanced regulation is too simplistic.

Fig. 5.

Fig. 5

Response profiles of two genes showing enhanced expression after acute and chronic MPL dosing. Ornithine decarboxylase (Odc1) expression is shown in the two left panels and exhibits a similar pattern of enhanced expression with acute and chronic dosing. Tryptophan 2,3-dioxygenase (Tdo2) is presented in the two right panels and exhibit dissimilar patterns with acute and chronic dosing.

Of the 358 genes, the profiles of 104 showed down-regulation after both dosing regimens. As with enhanced regulation, this classification is inadequate to capture the multiple patterns extant within this group. As an example, Fig. 6 shows the expression profiles of two genes that exhibit reduced expression in response to both dosing regimens. The first gene (Fig. 6, left) is progestin and adipoQ receptor family member VII (paqr7). One endogenous ligand for this receptor is adiponectin, a protein hormone produced and secreted by adipocytes and which regulates the metabolism of lipids and glucose (21). The response of this gene to acute dosing is a transient reduced expression. The first part of the response profile to chronic infusion also suggests a transient down-regulation with return to baseline. However, a second influence seems to then cause a slow decline in expression throughout the remainder of the infusion period. The second exemplar of a gene responding to both dosing regimens with decreased expression (Fig. 6, right) is C-type lectin, superfamily member 13, also known as Kupffer cell receptor (Kclr), which is involved in the cellular immune response (22). The response of this gene to acute dosing is a longer lasting transient period of decreased expression than is seen with paqr7. However, the response to chronic dosing is a decreased expression that is sustained throughout the entire infusion period.

Fig. 6.

Fig. 6

Response profiles of two genes showing reduced expression after acute and chronic MPL dosing. Progestin and adipoQ receptor family member VII (paqr 7) expression is shown in the two left panels and exhibits a similar pattern of reduced expression in the two dosing regimens. Kupffer cell receptor (Kclr), which exhibits dissimilar patterns of regulation with acute and chronic dosing, is presented in the two right panels.

The remaining 145 of the 358 genes showed profiles suggesting complex regulation involving both enhanced expression and down-regulated expression. In the initial analysis of the acute response dataset, we identified two clusters of genes with what we referred to as biphasic regulation in which there was an initial down-regulation followed by enhanced expression. Figure 7 shows the acute and chronic profiles of two genes that show initial down-regulation in the acute profile followed by a period of enhanced regulation. The first gene (Fig. 7, left) is G0/G1 switch gene 2 (GOS2), which is involved in cell cycle regulation (23). The acute profile shows down-regulation followed by enhanced expression. In the response to chronic infusion, the initial down-regulation phase is only suggested by one point and is followed by enhanced expression that continues throughout the entire 168 h. However, the pattern suggests that a third factor may be influencing expression beyond 48 h. In preliminary experiments, we determined that the release from the pump required about 6 h to reach steady-state blood MPL concentrations. This governed our choice of 6 h as the first sampling point. The second exemplar of down-regulation followed by enhanced regulation (Fig. 7, right) is arginase 1 (Arg1), which is involved in ammonia detoxification (24). Acutely the profile shows a sharp down-regulation followed by a period of enhanced regulation and a return to baseline by 48 h. However, in the response to chronic infusion, the initial down-regulation was missed due to our sampling schema. The profile does suggest that a second influence may be intervening after 24 h to maintain the enhanced expression throughout the remainder of the infusion period.

Fig. 7.

Fig. 7

Response profiles of two genes showing complex regulation of expression after acute and chronic MPL dosing. G9/G1 switch gene 2 (GOS2) and Arginase (Arg1) both exhibit initial down-regulation followed by a period of enhanced regulation.

Figure 8 provides examples of two genes in which the profile suggests a period of enhanced expression followed by down-regulation. The first exemplar (Fig. 8, left) is heterogeneous nuclear ribonucleoproteins methyltransferase-like 2 (Hrmt1L2), which is involved in the regulation of transcription (25). This enzyme shows a period of enhanced regulation followed by down-regulation in both profiles. The second exemplar in this group is nuclear receptor superfamily 0, group B, member 2 (NR0b2), also known as small heterodimer partner (Fig. 8, right). Proteins in this superfamily bind and are activated by small hydrophobic hormones such as retinoic acid, thyroid hormone, and steroids. This superfamily also includes orphan nuclear receptors (26). The acute profile for this gene shows a rapid and short-lived transient increase followed by a relatively long-lasting period of down-regulation. The chronic profile misses the initial up-regulation but captures what appears to be a two-phase down-regulation.

Fig. 8.

Fig. 8

Response profiles of two genes showing complex regulation of expression after acute and chronic MPL dosing. In this case, both heterogenous nuclear ribonucleoprotein methyltransferase (Hrm1L2) and nuclear receptor superfamily 0, group B member 2 (NROb2) show initial up-regulation followed by a later down-regulation.

Gene groupings and expression profiles

We also searched all 358 genes primarily using the NCBI “search across databases” feature. Based on this information and domain knowledge, we grouped the 358 genes primarily on function with two additional groups based on subcellular localization (mitochondrial and plasma membrane). These groups are presented in Tables 113 and list identifying criteria as well as temporal responses after both acute and chronic treatments. Although not perfect, these groupings together with the expression profiles provide insight into the global impact of corticosteroids on the liver. The most highly populated group is termed transcription-translation, which contains 54 genes (Table 1). The majority of genes in this group are transcription factors, indicating that a major influence of corticosteroids derives from their ability to alter the effect of other influences of transcription. Because alterations in the amount of messages have an impact only once they are reflected as changes in protein, the consequence of these changes should be delayed in time. Such a time delay may therefore explain the significant number of genes that express complex regulation. Also included in this group are a number of genes such as Nap1l1, Ddx3x, Nopp140, IMP3, EIF2C1, and EIF3 whose enhanced expression indicate increased translational activity, which is consistent with the generally anabolic effect of corticosteroids on liver.

TABLE 1.

MPL-regulated probe sets related to transcription-translation

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1387947 U56241 U56241 Mafb Transcription factor Maf1 Up Up
1387870 AB025017 X63369 TIS11 Transcription factor (immediate early gene) Up Up
1386910 AF311054 D44495 Apex Apurnic Endonuclease (DNA repair) Up Up
1371987 BI274697 AA891891 DNA repair Polymerase (DNA directed) sigma (DNA repair) Up Up
1367601, 1367602 NM_053698 AA900476 (2), AI014091 Cited2 Cbp/p300-interacting transactivator Up Up
1387779 NM_031668 AI237258, AI229637 Mybbp1a MYB binding protein (P160) 1a Up Up
1368308 NM_012603 Y00396 (2) Myc Myelocytomatosis proto-oncogene Up Up
1367831 NM_030989 X13058 Tp53 Tumor protein p53, p53 tumor sUppressor Up Up
1389508, 1374404 BI288619 AA944014 v-jun Sarcoma virus 17 oncogene Up Up
1389521 AI408553 AA799539 IVNS1ABP Influenza virus NS1A binding protein Up Up
1392633 BE105480 AA946439 H4 Histone H4 Up Up
1371872, 1370826, 1373473 BM386384 AF062594 (2), AA859920, AA866472 Nap1l1 Nucleosome assembly protein 1-like 1 Up Up
1372242, 1375901 AI169598 U21719 Ddx3x DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, helicase (RNA) Up Up
1368031, 1368032, 1368033 NM_022869 AA998882, M94287 Nopp140, Nolc1 Nucleolar phosphoprotein 140, interacts with RNA pol Up Up
1388321 BG378108 AA799369 IMP3 U3 small nucleolar ribonucleoprotein (ribosome biogenesis) Up Up
1368867 BF281131 H31692 EIF2C1, GERP95 Translation initiation factor 2C Up Up
1388576 BF281368 AA875205 (2) EIF3 Translation initiation factor 3 Up Up
1372090 AI231566 D14448 Max Myc family DNA-binding protein, leucine zipper Up Up
1388779, 1387962 U41164, BG381516 U41164 rKr1, Zfp180 Cys2/His2 zinc finger protein (Kruppel family) Up Up
1373012, 1373011 BE109520 (2) AA894086 Zfp622 Zinc finger protein 622 Up Up
1371864, 1370209 BE101336, AW524563 D12769 Bteb1 Klf9, Basic transcription element binding protein 1 Up Up
1387714, 1369737, 1369738 AB031423, NM_017334 (2) S66024 CREM Transcriptional repressor, cAMP-responsive element Up Up
1371781 BI285863 AI639141, X91810 Stat3 Signal transducer and activator of transcription 3 Up Up
1371714 BG378760 AA893885 Foxk2 Forkhead box K2 Up Up
1371489 AI011748 AF022081 Rnf4 Small nuclear RING finger protein Up Up
1370510 AB012600 AF015953 ARNTL, BMAL1b BMAL1b, aryl hydrocarbon receptor nuclear translocator-like Up Up
1370309 AJ238854 AB016536 alf-c1 Heterogeneous nuclear ribonucleoprotein type A/B Up/down Up
1370474, 1370474 J03933 J03819 Thrb, (c-erbA-β) Rat thyroid receptor hormone β Down/up Up
1387365 NM_031627 U11685 Nr1h3 Liver X receptor, α Down/up Up
1372876 AA799700 AA799700 SPS2 Selenophosphate synthetase 2 Down/up Up
1369679 AB060652 D78018, × 13167 NFI-A Transcription factor nuclear factor1-A1 Down/up Up
1369834 NM_012742 X55955 Hnf3a, Foxa1 Hepatocyte nuclear factor 3-α Forkhead box A1 Down Up
1375428 BE099979 AA858607 Creg1 Repressor of E1A-stimulated genes (cell growth and differentiation) Down Up
1376196 BG375059 AA800290 D receptor interacting protein Down Down
1389601, 1373644 BI293610 AA859994 Nfib Nuclear factor I/B Down Down
1387769 AF000942 AI171268, AF000942 Id3 DNA binding protein inhibitor 3 BHLH Down Down
1368712 NM_019620 U67082 Kzf1 Zinc finger protein 386 (Kruppel-like) Down Down
1369959 NM_017172 AI136891, AI112516 Zfp36l1 Zinc finger protein 36, C3H type-like 1, Kruppel Type 18 Down Down
1368711 NM_012743 L09647 Hnf3b, Foxa2 Forkhead box A2 Down Down
1373837 BI296633 AI227715 Rbl2 Retinoblastoma-like 2 (p130) Down Down
1369063 NM_012903 AA875495, D32209, Anp32 Acidic (leucine-rich) nuclear phosphoprotein 32 family, member A Down Down
1367826 NM_031789 AI177161 NF-E2-related factor 2 Nuclear factor erythroid 2-like 2 Down Down
1388650 BM385445 AA899854 Top2a Topoisomerase (DNA) 2α Down Down
1372889 AI407489 M64862, AA800797 DNA-binding protein; matrin F/G Down Down
1367759 NM_012578 AI232374 H1f0 Histone H1–0 Down Down
1370381 U61729 U61729 Prol2 Proline rich 2 Up/down Down
1368376 NM_057133 D86580 Nr0b2 Small heterodimer partner; SHP; SHP1 Up/down Down
1374752 AI408734 AI639149 Mdfic MyoD family inhibitor domain containing Up/down Down
1398756,1398757, 1398758 NM_012992 J03969, J04943 Npm1 Nucleophosmin 1, nuclear protein B23 Up Down
1398832 NM_012749 M55015, M55017 Ncl Nucleolin Up Down
1370711 AF000900 U63839, AF000899, AF000901 p58/p45 Nucleoporin p58 Up Down
1387028 M86708 L23148 Id1 DNA-binding protein inhibitor 1 BHLH Up Down
1372757, 1368835, 1387354 BM386875 AA892553 Stat1 Activator of transcription 1 Up Down
1386897 NM_024363 U60882 Hrmt1l2 Heterogeneous nuclear ribonucleoproteins methyltransferase-like 2 Up/down Up/down
1368303 NM_031678 AB016532 Per2 Period homolog 2 Oscillating Oscillating

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and categorized as relating to either transcription or translation. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

TABLE 13.

ESTs regulated by MPL

230A probe set 230A accession U34A probe set/accession Symbol Response
Acute Chronic
1394127 AA943800 AA892541 EST Up Up
1382516 AI170660 AA859725 EST Up Up
1376965 BM389656 AA893603 EST Up Up
1367543 BG377443 AA894234 EST Up Up
1383233 BI289032 AA891749 EST Down/up Up
1393061 AI030103 AA891739 EST Down/up Up
1385889 AA893212 AA893212 EST Down Up
1393751 AA859029 AA892778 EST Down Down
1372261 AI409067 AA891737 EST Down Down
1373970 AI716248 AA892986 EST Down Down
1389561 BE110624 AA891950 EST Down Down
1376792 AW251313 AA892027 EST Down Down
1374478 AA819329 AA892861 EST Down Down
1374767 AI105450 AA799396(2) EST Down Down
1376098 BF282304 AA875126 EST Up/down Up/down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration but at present unidentified. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification. EST, Expressed sequence tag.

The second most populated group is termed signaling with 53 members (Table 2). Eighteen members of this group show up-regulation in both the acute and chronic profiles, and 15 show down-regulation in both acute and chronic. What is unusual about this group is the large number of genes, 21, that show complex regulation. As might be expected, this group is dominated by kinases and phosphatases. We have also included in this group several membrane receptors that could have been included in the membrane group but were included in this group because of their involvement in signaling. Of particular interest is paqr7 whose endogenous ligand is adiponectin, a hormone produced by adipose tissue that is involved in the regulation of systemic energy metabolism (21). The down-regulation of this receptor after both acute and chronic dosing may provide additional insight into the metabolic dysregulation attendant on corticosteroid treatment.

TABLE 2.

MPL-Regulated probe sets related to cell signaling

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1371081 U78517 U78517 Cgef2, Rapgef4 Rap guanine nucleotide exchange factor (GEF) 4 Up Up
1367960 NM_019186 X77235 Arl4 ADP-ribosylation factor-like 4 Up Up
1398790 NM_017039 AI012595 Ppp2ca Protein phosphatase 2a, catalytic sub-unit, α isoform Up Up
1368147, 1368146 U02553, BE110108 U02553 Ptpn16, Dusp1 MAPK phosphatase-1, protein tyrosine phosphatase non-receptor-type 16 Up Up
1387078 NM_031002 U26397 Inpp4a Inositol polyphosphate-4-phosphatase, type 1 Up Up
1398847, 1370180 AA891213, BG376935 U95001 Nudt4 Diphosphoinositol polyphosphate phosphohydrolase 2 Up Up
1377136, 1367697 NM_031020, AW254190 AA924542, U73142 (2), AI171630, U91847, AI137862 Mapk14, p38 MAPK 14 Up Up
1367802 NM_019232 L01624 Sgk Serum/glucocorticoid-regulated kinase, serine/threonine protein kinase SGK Up Up
1367725 NM_022602 AF086624 Pim3 Serine/threonine-protein kinase pim-3 Up Up
1380262 AA893436 AA893436 sgk Serum/glucocorticoid regulated kinase Up Up
1368947 NM_024127 L32591 (2), AI070295, AI070295 Gadd45a Growth arrest and DNA-damage-inducible 45α Up Up
1369065 NM_017290 J04024 Atp2a2, Serca2 ATPase, Ca2+ transporting Up Up
1387154 NM_012614 M15880 Npy Neuropeptide Y (Npy) Up Up
1372558 AI177404 AI177404 NMDA receptor-regulated gene 1 Up Up
1389974 BF555171 L15618 CK2 Casein kinase II α subunit Up Up
1387907 J05510 J05510 InsP3R1
I145TR
Inositol 1,4,5-triphosphate receptor Up Up
1373306 BM386212 M81639 Snn Stannin Up Up
1372355 BE109242 U09793 c-Ki-ras Kirsten rat sarcoma viral oncogene homolog 2 Up Up
1373082 AA893743 AA893743 Protein kinase inhibitor, α Up Up/down
1372770 BF281357 X13933 CaMI Calmodulin 1 Up Down/up
1390240 BM389611 AA800456 CKLiK Calcium-calmodulin-dependent kinase I-like kinase (CKLiK) Down Up
1377417 BE099931 AA800678 Ticam1 Toll-like receptor adaptor molecule 1 Down Up
1370522 L04796 M96674 Gcgr Glucagon receptor Down Up
1387981 AF079864 AF079864 Olfr78 G-protein coupled receptor RA1c, olfactory receptor 59 Down Up
1368675 NM_032084 AI232194 CHN2 Chimerin (chimaerin) 2 Down Up
1368289 AA944965 M12450 Vdbp, DBP02 Group-specific component (vitamin D-binding protein) Down Up
1390798 BF288130 M10072 L-CA Leukocyte common antigen Down Down
1387024 NM_053883 X94185 mkp-3 MAPK phosphatase Down Down
1368871 NM_053887 U48596 Map3k1 MAPK kinase kinase 1 Down Down
1368646 NM_017322 L27112, AI231354 Mapk9 MAPK 9 Down Down
1387024 NM_053883 X94185, U42627 Dusp6 Dual specificity phosphatase 6 Down Down
1368646 NM_017322 AI231354 SAPK Stress-activated protein kinase α II Down Down
1372844, 1398273 AW531877, NM_053599 AA892417, D38056 Efna1, B61 Eph-related receptor tyrosine kinase ligand 1 Down Down
1389779 AA800626 AA800626 Sh2d4a SH2 domain containing 4A Down Down
1375879 BE111762 AF061443 Gpr48 G protein-coupled receptor 48 Down Down
1367644 L01115 L01115 Adcy6 Adenylyl cyclase type VI Down Down
1369644 NM_134408 AF063102 CIRL-2 Calcium-independent α-latrotoxin receptor homolog 2, latrophilin 2 Down Down
1368924 NM_017094 Z83757 (2) Ghr GH receptor Down Down
1377966 BI275560 AA894316 paqr7 Progestin and adipoQ receptor family member VII Down Down
1367745 NM_031143 D78588 Dgkz Diacylglycerol kinase ζ Down Down
1371969 BI291848 AI180288 Cald1 Caldesmon 1 Down Down
1368202 NM_024159 U95178 Dab2 Disabled homolog 2 Up/down Down
1367881 NM_013016 D85183 SHPS-1 Protein tyrosine phosphatase, non-receptor-type substrate 1 Up/down Down
1370949, 1370948, 1373432 BE111604, M59859 AA859896, AA925762, AA899253 Macs Myristoylated alanine-rich C-kinase substrate Up Down
1368856 NM_031514 U13396, AJ000557, U13396 Jak2 Janus kinase 2 Up Down
1368693 NM_024145 X57018 Fgr Gardner-Rasheed feline sarcoma viral oncogene homolog (kinase) Up Down
1367844 M12672 AA875225 G-α-i2 GTP-binding protein Up Down
1368975, 6, 1390325 NM_013127, D30795, BI289418 D29646 Cd38 ADP-ribosyl cyclase; CD38 Up Down
1368821, 1368822, 1372331 BG665037, BI290885, NM_024369 AA891233 Fstl1 Follistatin-related protein Up Down
1370288 AF372216 X02412, M60666 TP-3 Tropomyosin α-isoform Up Down
1388140 AW253722 M83678 Rab13 RAB13, member RAS oncogene family Down/up Down
1370414 M94043 M94043 Rab38 Rab38, member of RAS oncogene family Down/up Down
1368821 NM_024369 AA859885, AA891233 Frp, Fstl1 Follistatin-like 1, activin-binding protein Down/up Down
1368536 NM_057104 D28560 NPH-type III Phosphodiesterase I/nucleotide pyrophosphatase 2 Down Down/up/down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and categorized as relating to cell signaling. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

The third most populated group is termed small molecule metabolism and contains 38 genes (Table 3). The large numbers of genes in this group reflect both the major role of the liver in small molecule metabolism and the impact of corticosteroids on this function. This group is also unusual in the large number of genes showing complex regulation.

TABLE 3.

MPL-regulated probe sets related to small molecule metabolism

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1387109 NM_031576 AI137856, M10068 Por P450 (cytochrome) oxidoreductase Up Up
1371021, 1375412 AI101331 D49434 Arsb Arylsulfatase B, lysosomal Glycosaminoglycan degradation Up Up
1370370 AF034218 AF034218 (2) Hyal2 Hyaluronidase 2, lysosomal glycosaminoglycan degradation Up Up
1387963 M24396 J03959, × 13098 Uricase, urate oxidase 2 Up Up
1387740, 1379361 NM_053487 AA892128, AJ224120 Pex11a Peroxisome biogenesis factor 11A Down/up Up
1368180 NM_017013 AI235747 Gsta2 Glutathione-S-transferase, α-type 2 Down/up Up
1369986 NM_033349 AI012802 Hagh Hydroxyacyl glutathione hydrolase, both cytosol and M Down/up Up
1367798 NM_017201 M15185 Ahcy S-adenosylhomocysteine hydrolase, cytosol Down/up Up
1370698 M13506 M13506 UDPGTR-2 Liver UDP-glucuronosyltransferase, phenobarbital-inducible, microsomal Down/ up Up
1398307 D38381 D38381 Cyp3a18 P450 6β-2 Down/up Up
1387314 NM_022513 D89375 Sult1b1 Sulfotransferase family 1B, member 1 cytosol Down/up Up
1387825 NM_031533 J02589 Androsterone UDP-glucuronosyltransferase Down/up Up
1368226 NM_133525 U82591 Rcl, C6orf108 Nucleoside 2-deoxyribosyltransferase domain (c-Myc-responsive) Down/up Up
1387659 AF245172 AA859837 (2) Gda Guanine deaminase Up Down/up
1387973 U39206 U39206 CYP4F4 CYP4F4 Down Up
1387243 K02422 K03241 Cyp1a2 Cyp1a2 Down Up
1370496 AB008424 AB008424 Cyp2d3 Cyp2d3 Down Up
1367843 NM_134407 AA892821 (2) Akr7a2, Aiar Aldo-keto reductase family 7, member A2 (aflatoxin aldehyde reductase), perox Down Up
1388300 AA892234 AA892234 Gst3 Glutathione S-transferase 3, microsomal Down Up
1368717 NM_024132 U72497 Faah Fatty acid amide hydrolase (degrades bioactive fatty acid amides) Down Up
1387958 L32601 L32601 20 α-Hydroxysteroid dehydrogenase Down Up
1387672 NM_017084 X06150, AA893219 Gnmt Glycine methyltransferase Down/up/down Up
1371076 AI454613 K01721 Cyp2b15 Cyp2b15 Down/up Down/up
1368497 NM_012833 D86086 Abcc2 ATP-binding cassette, sub-family C (CFTR/MRP), member 2 Down/up Down
1370397 M33936 AA924591 Cyp4A3 Cytochrome P450 4A3 Up Down
1370080 NM_012580 J02722 HEOXG Heox Hmox Heme oxygenas Up Up/down
1387916 U39208 U39208 U39208 CYP4F6 Down Down
1369424 NM_012693 J04187 Cyp2a2 Cyp2a2 Down Down
1367988 U04733 U04733 cyp 2C23 Cytochrome P450 arachidonic acid epoxygenase Down Down
1387669 NM_012844 M26125 Ephx1 Epoxide hydrolase 1, microsomal Down Down
1387214 NM_031565 X81395 Ces1 Carboxylesterase 1 Down Down
1387022 NM_022407 AF001898 Aldh1a1 Aldehyde dehydrogenase family 1, member A1 cytosolic Down Down
1369296 NM_031732 L22339 (2) Sult1a2 Sulfotransferase family, cytosolic, 1C, member 1, N-hydroxy-2-acetylaminofluorene; sulfotransferase Down Down
1387221 NM_024356 AI639457 Gch GTP cyclohydrolase 1 Down Down
1378753 AI638971 AI638971 Tpmt Thiopurine methyltransferase Down Down
1368409 NM_012796 AI138143, D10026 Gstt2 Glutathione S-transferase, θ 2 Down Down
1370688, 1372523, 1370030 J05181, AA892770, NM_017305 J05181 (3), S65555 Glclc Glutamate-cysteine ligase, catalytic subunit Down Down
1368826 NM_012531 M93257, M60753 Comt Catechol-O-methyltransferase Up/down/up Down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and categorized as relating to small molecule metabolism. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

The next most populated group is classified as plasma membrane localized with 33 members (Table 4). Because the surface membrane of a cell mediates its interaction with the external environment, this group by necessity is difficult to distinguish from the signaling group. Pharmacologically, corticosteroids are used for their antiinflammatory and immunomodulatory effects. The liver plays a major role in immune protection both directly and indirectly. The direct effects are the production and secretion of a variety of proteins involved in immune responses such as complement proteins. Indirectly, the liver harbors the Kupffer cell population that provides a defense barrier between the hepatic portal system and general circulation. The immune related group contains 31 genes (Table 5). These 31 genes illustrate the broad impact of corticosteroids on both immune and inflammatory processes. In addition, these results illustrate the type of coordinated effects corticosteroids have on the immune system. For example, complement protein C1q is down-regulated in both the acute and chronic profiles, whereas complement protein C1q binding protein, which binds and inhibits C1q activation, is up-regulated in both (27, 28).

TABLE 4.

MPL-regulated probe sets localized to plasma membranes

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1371113 M58040 M58040 Tfrc, CD71 Transferrin receptor Up Up
1387027 U72741 U72741 Lgals9 β-Galactoside binding lectin 9 Up Up
1369716 NM_012976 L21711 Lgals5 β-Galactoside binding protein 5 Up Up
1386937, 1367814 M14137, AI232036 AI112173 Atp1b1 ATPase Na+/K+ transporting β1 Up Up
1367585, 1371108 M74494, M28647 M74494 Nkaa1b Na,K-ATPase α-1 subunit Up Up
1370262_at AI706785 AF100421 Lyric Metadherin, metastasis Up Up
1367568_a_at NM_012862 AI012030 Mgp Matrix Gla protein Up Up
1367579_a_at BI285434 AA892333 Tuba1 Tubulin, α6 Up Up
1371542_at BI284599 J00797 Tubulin, α4 Up Up
1367669 AI233190 U05784 MPL3 Light chain 3 subunit of microtubule-associated proteins 1A and 1B Up Up
1367721 NM_012649 S61868 SYND4 Syndecan 4, ryudocan Up Up
1369976_at NM_053319 AI009806 MGC94628 Dynein, cytoplasmic, light chain 1 Up Up
1376098_a_at BF282304 AA875126 (2) Myo1g Myosin IG Up Up
1369720_at NM_053986 X68199 Myo1b Myosin Ib Up Up
1374171, 1387287 D83598, AI170507 AF019628 Sur2 Sulfonylurea receptor 2 Up Up
1373842 BM390718 AA858620 N-WASP Wiskott-Aldrich syndrome gene-like Up/down Up
1370526_at AF020045 AF020046 Itgae Adhesion receptor, integrin α E1 Down/up Up
1369953 BI285141 U49062 (2) Cd24 CD24 antigen Up Down
1373932 BE098739 AA894029 Cybb Endothelial type gp91-phox Up Down
1390659 BI302830 M61875 HAMM CD44A METAA Cell surface glycoprotein (hyaluronate binding protein) Up Down
1368419 AF202115 AI639438 glypican 1 GPI-anchored ceruloplasmin Up Down
1387856_at BI274457 AA944422 Cnn3 Calponin 3 Up/down Down
1387206_at NM_031740 AF048687 B4galt6 UDP-Gal:β GlcNAc-β 1,4-galactosyltransferase, polypeptide 6 Up/down Down
1372087 BG666916 AJ012603 TACE TNF-α converting enzyme Up/down Down
1367849_at NM_013026 X60651, S61865 Synd1 Syndecan 1 Down Down
1368642, 1387259 AF097593, NM_031333 AF097593 (2) Cdh2 N-cadherin Down Down
1388441_at BG379987 AA892773 Thymosin, β 4 Down Down
1386938_at NM_031012 M25073 Anpep, CD13 Alanyl (membrane) aminopeptidase Down Down
1372780_at BM391310 AA892353 Transmembrane protein 53 Down Down
1368115_at NM_031700 M74067 Cldn3 Claudin 3 Down Down
1387061_at NM_031047 1387061_at Jup Junction plakoglobin, γ-catenin Down Down
1367812_at NM_019167 AB001347 Spnb3 β-Spectrin 3 Down Down
1383606_at BI302544 AI639417 Membrane targeting (tandem) C2 domain containing 1 Down Down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and identified as localized to plasma membranes. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

TABLE 5.

Immune-related MPL-regulated probe sets

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1398784 NM_019259 AI178135 C1qbp C1q binding, inhibits C1 activation Up Up
1390104 BI296551 AI010580 Irgq Immunity-related GTPase family, Q Up Up
1388331 BG057543 AA685903 Tumor rejection antigen gp96 Up Up
1375686 AI706907 AA892680, AA892298 Ppil3 Peptidylprolyl isomerase (cyclophilin)-like 3 Up Up
1367657 NM_017258 L26268 (2) Btg1 B-cell translocation gene 1, antiproliferative Up Up
1368668 NM_053866 U17901 Plaa Phospholipase A2, activating protein Up Up
1370750, 1369255 NM_013123 M95578 (2), U14010 (2) Il1r1 IL-1 receptor, type I Up Up
1371926, 1373140, 1370957 BM383427 M92340 Il6st IL-6 signal transducer Up Up
1386987 NM_017020 M58587 Il6r IL-6 receptor Up Up
1372926 AI009159 U27201 TIMP-3 Tissue inhibitor of metalloproteinase 3 Up Up
1370855 BG666933 AI231292 Cst3 Cystatin C (inhibitor of cysteine proteinases) cathepsins Down Up
1387127 NM_031351 AA859645 Atrn, DPPT-L Attractin Down Up
1398256 NM_031512 M98820 Il1b IL-1β Down Down
1374334 AI412189 AI234828 Ig heavy chain V-III region VH26 precursor Down Down
1371100 AA859049 D00362, M20629 Es2 Esterase 2 Down Down
1368755 NM_053753 M55532 Kclr C-type (calcium dependent) lectin, superfamily member 13 Down Down
1368741 NM_057146 U52948 C9 Complement component C9 Down Down
1373025 AI411618 AA891576 Complement protein C1q β chain Down Down
1370027, 1388229 M22359, NM_023103 M22360, M22359 Mug1 Murinoglobulin 1 homolog, plasma proteinase inhibitor α-1-inhibitor III Down Down
1371015 X52711 X52711 Mx1 Myxovirus (influenza virus) resistance 1 Up/down Down
1370056 NM_020103 M30691 Ly6c Ly6-C antigen Up/down Down
1368332 NM_133624 M80367 Gbp2 Guanylate nucleotide binding protein 2 Up/down Down
1367614 NM_012904 S57478, AI171962 Anxa1 Annexin A1, lipocortin I Up/down Down
1368073 NM_012591 M34253 (2) Irf1 Interferon regulatory factor 1 Up/down Down
1368592, 1371170 AJ245643, NM_017019 D00403 Il1a IL-1α (Il1a) Up/down Down
1367679 NM_013069 X13044 INVG34 Histocompatibility: class II antigens, γ-chain of Up/down Down
1383564 BF411036 AA799861(2) Irf7 Interferon regulatory factor 7 Up Down
1398246 NM_053843 M32062 (2) Fcgr3 Fc receptor, IgG, low affinity III Up Down
1387687 NM_133542 AJ223184 Igsf6 Ig superfamily, member 6 Up Down
1367574 NM_031140 X62952 Vim Vimentin Up Down
1376151 AI407953 AA891944 Interferon-γ-induced GTPase Down/up Down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and categorized as immune related. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

The next group are 29 genes related to protein and amino acid metabolism (Table 6). The liver both produces and degrades serum proteins (29). In addition, the liver is only one of two tissues that can synthesize glucose (30). Amino acid carbon from the musculature is a major substrate for gluconeogenesis, which requires that the resulting ammonia be detoxified. We have included proteosome genes, aminotransferases, and chaperonins as well as a variety of genes involved in the metabolism and synthesis of several amino acids.

TABLE 6.

MPL-regulated probe sets related to protein or amino acid metabolism

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1386923, 1375642 M62388 M62388 (2), AA799612 Ubiquitin-conjugating protein Up Up
1371814 BI282191 AA859722 Ube2g2 Ubiquitin-conjugating enzyme E2G 2 Up Up
1382059 BI289529 AI639506 Fbxo30 F-box protein 30 (Fbxo30), protein-ubiquitin ligases Up Up
1392633 AI045724 AI639312 Fbxo32 F-box only protein 32 (Fbxo32), protein-ubiquitin ligases Up Up
1370964 BF283456 X12459 Ass Argininosuccinate synthetase (ammonia detoxification) Up Up
1368720 NM_022403 AA945143 Tdo2 Tryptophan 2,3-dioxygenase (kynurenine pathway) Up Up
1368247 NM_031971 AI170613 (2) Hspa1a, Hsp10 mitochondrial chaperonin Up Up
1398960 AI172328 AA875047 TCP20 chaperonin subunit 6a (ζ) Up Up
1388698 AI236601 AI236601 HSP105 Heat shock protein 105 Up Up
1389021 BF284746 AA799531 ASNS Asparagine synthetase domain containing 1 Up Up
1368188 NM_017233 AA866302 Hpd 4-Hydroxyphenylpyruvic acid dioxygenase Down/up Up
1367695 NM_022390 J03481 Qdpr Quinoid dihydropteridine reductase (PKU) Down/up Up
1368794 NM_020076 D28339, D44494 Haao 3-Hydroxyanthranilate 3,4-dioxygenase (synthesis of quinolinic acid) Down/up Up
1368266 NM_017134 J02720 Arg1 Arginase 1 (ammonia detoxification) Down/up Up
1370200, 1387878 AI179613, AI233216 BI284411 Glud1 Glutamate dehydrogenase 1(ammonia detoxification) Down Up
1368085 NM_133595 U85512 Gchfr GTP cyclohydrolase I feedback regulator (phenylalanine, tyrosine, and tryptophan hydroxylases) Down Up
1368092 NM_017181 M77694 Fah Fumarylacetoacetate hydrolase (tyrosinemia type I) Down Up
1367627 NM_031031 U07971 Gatm Glycine amidinotransferase (L-arginine:glycine amidinotransferase) Down Down
1387307 NM_017159 M58308, AB002393 Hal Histidine ammonia lyase Down Down
1373686 AA893495 AA893495 Serpina6 Serine (or cysteine) proteinase inhibitor, clade A (α-1 antiproteinase, antitrypsin), member 6 Down Down
1387193, 1368447, 1368446 NM_012674 M35299 Spink1 Serine protease inhibitor, Kazal type 1 Down Down
1368280 NM_017097 D90404 (2) Ctsc Cathepsin C Down Down
1370386 AB002406 AB002406 Ruvbl1 Ribosomal protein s25 Up/down Down
1374255 BI281789 AA875602 Phenylalanine-tRNA synthetase-like, α-subunit Up/down Down
1373592 AI407094 AA875037 Serine proteinase inhibitor mBM2A, serine (or cysteine) peptidase inhibitor, clade B, member 9 Up/down Down
1373263, 1376737 AW523737 H31976 SUMO Sentrin specific protease 5 Up Down
1367710 NM_017257 D45250 Psme2 Proteasome (prosome, macropain) 28 subunit, β Up Down
1375421 AI600019 AA894089 Neurodap1 Neurodegeneration associated protein 1 Up Down
1372665 AI230228 AI230228 Phosphoserine aminotransferase Up Down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and categorized as relating to protein or amino acid metabolism. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

The next group of genes, containing 23 members, is termed nuclear encoded mitochondrial genes (Table 7). In this group, all but four genes have chronic profiles indicating enhanced expression. Interestingly, a significant number of these genes show an initial down-regulation followed by up-regulation, giving the acute profiles a biphasic characteristic. Of particular interest in this group is the up-regulation of pyruvate dehydrogenase kinase 1 (Pdk) and the down-regulation of pyruvate dehydrogenase phosphatase isoenzyme 2 (Pdp). The pyruvate dehydrogenase complex is a matrix multienzyme complex that provides the primary link between glycolysis and the tricarboxylic acid (TCA) cycle. This complex is inactivated when it is phosphorylated by Pdk and activated when it is dephosphorylated by Pdp (31). The coordinated opposite regulation of these two enzymes in effect breaks the link between glycolysis and the TCA cycle, which would contribute strongly to the insulin resistance caused by corticosteroids.

TABLE 7.

Nuclear-encoded mitochondrial genes regulated by MPL

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1398870, 1370785 D63411 U21871, D63411 TOM20 Outer mitochondrial membrane receptor rTOM20 Up Up
1387773 NM_012839 K00750 (2) Cycs Cytochrome c, apoptosis Up Up
1398763, 1368051 NM_032066 U81186 (2) Hsd17b12 Translocase of inner mitochondrial membrane 23 Up Up
1371795, 1370005 NM_030586 Y12517, AI232256 omb5 Cytochrome b5, outer mitochondrial membrane isoform Up Up
1367982 NM_024484 J03190 (2) Alas1 Aminolevulinic acid synthase 1, heme biosynthetic Up Up
1375504 BM390747 AA892950 Polymerase (DNA), γ2, mitochondrial DNA Up Up
1370918 BI275939 L19927 Atp5c1 ATP synthase, H+ transporting, mitochondrial F1 complex Down/up Up
1386917 NM_012744 U32314 (2) Pc Pyruvate carboxylase (pyruvate to oxaloacetate) Down/up Up
1377758 BF415386 AA893658 Short-chain dehydrogenase/reductase 9 Down/up Up
1389021 BF284746 AA891785 Idh2 Isocitrate dehydrogenase 2 Down/up Up
1368514 NM_013198 M23601 Maob Monoamine oxidase B, mitochondria Down/up Up
1369799 U29701 D87839 (2) Abat 4-Aminobutyrate aminotransferase, GABA amino-transferase mitochondria Down/up Up
1370151 NM_017072 M11710, Cps1 Carbamoyl-phosphate synthetase 1, mitochondrial (ammonia detoxification) Down/up Up
1369671 K03040 M11266 Otc Ornithine transcarbamylase mitochondria (ammonia detoxification) Down/up Up
1370592 AB019693 AA893035 HP33, Keg1 Kidney expressed gene 1 Down Up
1369023 NM_031052 M96633 Mipep Mitochondrial intermediate peptidase Down Up
1368566 AA964381 AB000098 Mipp65 NADH dehydrogenase (ubiquinone) flavoprotein 3-like Down Up
1370232 AI102838 J05031 Ivd Isovaleryl coenzyme A dehydrogenase Down Up
1368079 NM_053826 L22294 Pdk1 Pyruvate dehydrogenase kinase 1 Down Up
1370509 AF062741 AF062741(2) Pdp2 Pyruvate dehydrogenase phosphatase isoenzyme 2 Down Down
1368387, 1374765 BI288055, NM_053995 AA817846 BDH 3-Hydroxybutyrate dehydrogenase mitochondrial Down Down
1371824 AA891949 AA891949 Ak4 Adenylate kinase 4 Down Down
1369588, 1370350 AF368860, NM_012915 D13122 IF1PA ATPase inhibitor (mitochondrial IF1 protein) Down Down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and identified as nuclear-encoded mitochondrial genes. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

The liver also is centrally involved in systemic lipid distribution and metabolism. We identified 21 genes associated with lipid metabolism (Table 8). The regulation of these genes may provide insight into the dyslipidemia caused by corticosteroids. For example, the up-regulation of apolipoprotein C-IV suggests the increased production of very-low-density lipoproteins. Very-low-density lipoproteins transport triacylglycerols from liver to extrahepatic tissues (32). Corticosteroids also influence the expression of 16 transporters (Table 9). Because many of these transporters are located on the sinusoidal membranes of hepatocytes, they may influence a variety of functions such as small molecule metabolism. For example, Slc22a7 is involved in the hepatic uptake of organic anions such as salicylate (33). The down-regulation of this gene could alter the metabolism of such drugs.

TABLE 8.

MPL-regulated probe sets related to lipid metabolism

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1398272 NM_022860 D17809 Galgt1 (N-Acetylneuraminyl)-galactosylglucosylceramide N-acetylgalactosaminyltransferase Up Up
1369656, 1389299 M36071 AA925887 Pcyt1a Phosphate cytidylyltransferase 1, choline, α (phosphatidylcholine synthesis) Up Up
1368890 NM_053410 AA799779 (2) Gnpat Glyceronephosphate O-acyltransferase (bio-synthesis of ether phospholipids, peroxisomes) Up Up
1384417 AA998783 AA945171 Apoc4 Apolipoprotein C-IV Down Up
1369465 NM_012584 M67465 Hsd3b Steroid δ-isomerase, 3β (biosynthesis hormonal steroids) Down Up
1368239 NM_053541 AB009463 rLRp105 Low-density lipoprotein receptor-related protein 3 Down/up Up
1382680 AA874941 AA874941, AA893280 ADRP Adipose differentiation-related protein, Down/up Up
1367662 NM_031682 AA945583 Hsd17b10 Hydroxyacyl-coenzyme A dehydrogenase type II (β-oxidation of fatty acids) Down/up Up
1368232 NM_031063 AA924198, M29472 Mvk Mevalonate kinase, biosynthesis of cholesterol Down/up Down/up
1387183 J02844 U26033, J02844 CROT Carnitine octanoyltransferase Down Down
1370909, 1388153 D90109, BI277523 D90109, AI044900, AA893242 (2) ACS COAA Long-chain acyl-CoA synthetase Down Down
1392604 AA997187 AA893032 Nsdhl NAD(P)-dependent steroid dehydrogenase-like Down Down
1389906, 1367839 AW530769 H33426, M95591 Fdft1 Farnesyl diphosphate farnesyl transferase 1 Down Down
1367668 NM_031841 AA875269 SCD Stearoyl-coenzyme A desaturase 2, δ-9-desaturase Down Down
1387058 NM_017225 AF040261 Pctp Phosphatidylcholine transfer protein Down Down
1388348, 1387630 NM_134382 AA892832 ELO1 ELOVL family member 5, elongation of long chain fatty acids Down Down
1368075 NM_012732 AA874784, S81497 (2) Lipa Lysosomal acid lipase 1 Down Down
1370024 NM_030832 U02096 Fabp7 Fatty acid binding protein 7 Down Down
1370583, 1370465, 1370364, 1369161 NM_012690 L15079 Abcb4 ATP-binding cassette, sub-family B (MDR/TAP), member 4 Down Down
1398310 D17309 S80431, D17309 Akr1d1 Aldo-keto reductase family 1, member D1 Down/up Down
1386965 NM_012598 AI237731, L03294 Lpl Lipoprotein lipase Up Down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and categorized as relating to lipid metabolism. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

TABLE 9.

MPL-regulated probe sets related to transport

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1379739 BF408431 AA892414 SLC4A7 Solute carrier family 4, sodium bicarbonate co- transporter, member 7 Up Up
1369460 NM_022619 U53927 Slc7a2 Solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 Up Up
1376267, 1390036 AA859652 AA859652 Slc16a6, MCT6 Monocarboxylate Transporter 6 Up/down Up
1368600 NM_022287 L23413 sat-1, Slc26a1 Solute carrier family 26 (sulfate transporter), member 1 Down Up
1398295 NM_031684 AF015304 Slc29a1 Equilibrative nucleoside transporter 1; ent1 Down Up
1387228 NM_012879 NM_012879 GLUT2 Facilitated glucose transporter Down Up
1387896, 1370296 M62763 M62763, M58287 Scp2 Nonspecific lipid transfer protein Down Up
1368745 NM_017222 U07183 Slc10a2 Solute carrier family 10, member 2 (sodium/bile acid cotransporter family) Up Down
1375823 BF392130 AF004017 SLC4A4, NBC Electrogenic Na+ bicarbonate cotransporter Up/down Down
1392929 BF416678 AA800202 Slc4A11 Solute carrier family 4, sodium bicarbonate transporter-like, member 11 Down Down
1371525 BI277550 AA799691 KCC3 Solute carrier family 12 (potassium/chloride transporters), member 7 Down Down
1368047 NM_022866 AA892616 Slc13a3 Solute carrier family 13 (sodium-dependent dicarboxylate transporter), member 3 Down Down
1398267 NM_053537 L27651 (2) Slc22a7 Solute carrier family 22 (organic anion transporter), member 7 Down Down
1390416 H35736 AA892522 Slc25a30, KMCP1 Solute carrier family 25, member 30, mitochondrial carrier protein-1 Down Down
1390412, 1376972, 1387130 NM_133315 U76714 (2) Slc39a1 Solute carrier family 39 (iron-regulated transporter), member 1 Down Down
1368316 NM_019158 AB005547 Aqp8 Aquaporin 8 Down Down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and categorized as relating to transport. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

The next 10 genes are grouped because they are involved in cellular fates, proliferation, differentiation, and apoptosis (Table 10). We have included ornithine decarboxylase 1 (Odc1) and ornithine decarboxylase antizyme inhibitor (oa-zin) because polyamines are important to cell proliferation. Odc, the first enzyme in polyamine synthesis, is regulated by a destabilizing antizyme that both inhibits its activity and accelerates it degradation. Oazin binds and traps the anti-zyme, thus promoting Odc activity (34, 35). The fact that both are up-regulated illustrates that corticosteroid treatment promotes polyamine synthesis in a coordinated fashion.

TABLE 10.

MPL-regulated probe sets related to cell fates

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1374595, 1376999 BF389721 AA892570 Tnks2 Tankyrase, telomeric repeat-binding factor 1 Up Up
1388953 AA892598 AA800679, AA892598 (2) GNL3 Guanine nucleotide binding protein-like 3 (nucleolar), stem cell proliferation Up Up
1370163 BF281299 J04791 Odc1 Ornithine decarboxylase 1 Up Up
1370575 D50734 AI043631 Oazin Ornithine decarboxylase antizyme inhibitor Up Up
1370807 AF411216 AA859954 Vacuole membrane protein 1 (apoptosis) Up Up
1367764 NM_012923 X70871 Ccng1 Cyclin G1 Up Up
1388395 AI406939 AA893235 GOS2 G0/G1 switch gene 2 Down/up Up
1367847 NM_053611 NM_053611 Nupr1, p8 Nuclear protein 1 (cell proliferation) Down/up Up
1389179 BF284899 AA800243 DFF Cell death-inducing DNA fragmentation factor Down/up Up
1371643 AW143798 X75207 CCND1 CCND1 gene; cyclin D1 Down Down
1368311 NM_012861 M76704 MGMT O6-Methylguanine-DNA methyltransferase (DNA repair) Down Down
1371684 AA799330 AA799330 Pelota homolog (cell division) Up Down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and categorized as relating to cell fates (proliferation, differentiation, or apoptosis). Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

The two profiles also identified 10 genes involved in carbohydrate metabolism (Table 11). Some of these genes are involved in both glycolysis and gluconeogenesis. For example, aldolase B is a reversible enzyme that is necessary for gluconeogenesis, so its up-regulation with chronic corticosteroid treatment is reasonable. In contrast, malic enzyme 1 catalyzes the decarboxylation of malate to form pyruvate and provides a link between glycolysis and the TCA cycle (36). The down-regulation of malic enzyme is consistent with the inactivation of the pyruvate dehydrogenase complex discussed above.

TABLE 11.

MPL-regulated probe sets related to carbohydrate metabolism

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1386864, 1369473 NM_053290 AI169417, U20195 Pgam1 Phosphoglycerate mutase 1 (glycolytic) Up Up
1387052 NM_031039 D10354 Gpt Glutamic pyruvic transaminase 1, soluble Down/up Up
1369560, 1371363 NM_022215 AB002558 Gpd3 Glycerol-3-phosphate dehydrogenase 1 (soluble) Down Up
1370299 M10149 AA892395 Aldob Aldolase B Down Up
1369635 NM_017052 X74593, AI030175 Sord Sorbitol dehydrogenase Down Down
1373337 AI412065 AA892799, AA892799, AA892799 Glyoxylate reductase/hydroxypyruvate reductase Down Down
1370870, 1370067 NM_012600 AI171506, M26594, AI171506, AI008020 Me1 Malic enzyme 1 (cytosolic) Down Down
1369954 NM_031510 AA892314 IDH1 Isocitrate dehydrogenase 1, soluble Down Down
1370725, 1386944 NM_013098 L37333 G6pc Glucose-6-phosphatase, catalytic Down Down/up
1387312 NM_012565 X53588 Gck Glucokinase Down/up Down/up

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration and categorized as relating to carbohydrate metabolism. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

We found 19 genes whose expression was regulated but were difficult to put into the above categories and were characterized as other (Table 12). For the most part, these are genes involved in secretory functions of the liver. The accession numbers for every one of the identified probe sets on both chips were submitted to the NCBI Blast program. However, there still remained 16 probe sets that Blasted to the same sequence(s) but could not be identified (Table 13). We have maintained them because identification is an ongoing process.

TABLE 12.

Other MPL-regulated probe sets

230A probe set 230A accession U34A probe set/accession Symbol Identification Response
Acute Chronic
1398784 NM_019259 AI178135 C1qbp C1q binding, inhibits C1 activation Up Up
1367794 NM_012488 AA900582, M22670, M22670, AI113046 A2m α2-Macroglobulin Up Up
1367801 NM_053596 D29683, AA956930 Ece1 Endothelin converting enzyme 1 Up Up
1369065 NM_017290 J04024 Atp2a2, Serca2 ATPase, Ca2+ transporting Up Up
1371345 AI008699 AI073164 Scamp1 Secretory carrier membrane protein Up Up
1369982 NM_031008 AA800186 Adaptor protein complex AP-2 Up Up
1386983 NM_013168 X06827 PBGD Hydroxymethylbilane synthase PBGD Down/up Up
1368224 NM_031531 D00753 Spin2c Serine protease inhibitor Down Up
1367647 NM_022519 X16273 Serpina1 Serine protease inhibitor α1 Down Up
1387819 NM_012552 L00117 Ela1 Elastase 1, serine protease Down Up
1371237 AF411318 AI102562 Mt1a Metallothionein 1 Down Up
1387323 NM_012725 M30282 Klk3 Kallikrein B, plasma 1 Down Down
1374524 BM384384 AA893080 Selenocysteine lyase Down Down
1368322 NM_012880 Z24721 ECSODPT Superoxide dismutase 3 (secreted) Down Down
1367720 NM_012899 AA800745 Alad Aminolevulinate, δ- dehydratase Down Down
1367980 NM_019124 D85844 RABPT5 Rabaptin 5 Down Down
1387146 X57764 S65355, AA818970 Ednrb Endothelin receptor type B Up/down Down
1367800 NM_013151 M23697 Plat Plasminogen activator, tissue Up/down Down
1370080 NM_012580 AI179610, J02722 Hmox1 Heme oxygenase (decycling) 1 Up Up/down

Probe sets regulated by MPL in rat liver after both acute and chronic drug administration but uncategorized. Affymetrix 230A arrays use a probe set number distinct from GenBank accession numbers, whereas U34A chips use a common probe set/GenBank accession number for identification.

Discussion

This report describes the mining of a microarray dataset obtained from the analysis of livers from a population of adrenalectomized animals treated with a chronic infusion of MPL for up to 1 wk. Liver RNA from four control animals and four animals killed at each of 10 time points over a 168-h period were applied to individual Affymetrix R230A chips. The dataset was mined using a filtering approach similar to the one applied to datasets developed from the liver, skeletal muscle, and kidneys of animals treated with a single bolus dose of MPL where animals were killed at 16 time points after dosing and compared with untreated controls (68). This filtration approach does not select for probes but rather eliminates probe sets that do not meet explicit requirements. Those probe sets not eliminated are retained for analysis. The filtration yielded a remainder of 1989 probe sets for further consideration. These probe sets were compared with 1518 probe sets that remained after filtering of the dataset obtained after bolus dosing with MPL (6). This comparison yielded 464 probe sets in the bolus dosing dataset that corresponded to 417 probe sets in the chronic infusion dataset. The results identified 358 different genes regulated by both dosing regimens. Because the filtering process is quite stringent, these 358 genes most likely do not include all genes regulated by MPL in the tissue. However, they do provide a basis for evaluating the global effects of corticosteroids on the liver.

The objective of obtaining two time series profiles for each gene is to identify genes with common mechanisms of regulation. The hypothesis is that if two or more genes have a common mechanism of regulation then they should have the same temporal profile in response to all dosing regimens. There are available a variety of clustering methods designed to group genes based on their profiles. However, at present, there is no analytical method available to cluster using two profiles, bi-clustering. Our filtering approach crudely identifies genes that meet minimal criteria for up- and down-regulation based on deviation from baseline. In both the acute (bolus dose) and chronic (infusion) datasets, some profiles met both criteria, suggesting complex regulation. We visually inspected all profiles (464 acute and 417 chronic) and categorized each based on up or down deviation from baseline. In many cases, because of probe set redundancy, more than one profile was available for acute, chronic, or both. These instances are noted on the tables. With this information we proceeded to evaluate acute and chronic profiles together. Based solely on this crude classification, we identified four basic patterns. For more than 60% of the genes, the profile was either up or down in both the acute and chronic profiles. In the remaining cases, the profiles were complex involving both up- and down-regulation.

The value of expression profiling using a rich in vivo time course such as this, particularly when two different dosing regimens are examined, is that patterns of transcriptional changes become apparent. Examination of expression patterns, as illustrated by examples presented in Figs. 58, indicate that our crude classification into four basic categories is too simplistic. Even when the more than 60% of the genes where the profiles from the two dosing regimens were either both up or down were examined, the mechanism of regulation is generally not straightforward. Single time point studies allow one to determine only magnitude of change at a single instant, which may not be indicative of either the actual extent or in some cases even the direction of change. This is particularly evident in cases of genes that express a biphasic pattern, where at different points in time the gene may show enhanced as opposed to reduced expression. Furthermore, temporal patterns may provide insight as to the mechanisms involved in regulation of transcriptional activity of a particular gene or genes.

The generally accepted mechanism for most glucocorticoid effects involves binding of free steroid to a cytoplasmically localized receptor, translocation of ligand-bound receptor to the nucleus, binding of a ligand receptor dimer to a specific DNA site [glucocorticoid response element (GRE)], and modulation of the amount of selective mRNAs (37). Although some effects on mRNA stability have been noted, a common mechanism involves increasing or decreasing the rate of transcription of particular genes. Previously, studying the enzyme TAT in liver and using both a repeated dosing and a chronic dosing paradigm, we described the phenomena of steroid tolerance (12, 13, 18). In those reports, we demonstrated that MPL treatment caused a rather long-lived down-regulation of the GR (mRNA and protein) and that when a second dose was administered before full recovery of receptor, the enhanced expression of TAT (mRNA and protein) was reduced proportional to the reduced concentration of receptor (18). In subsequent studies, we found that chronic infusion of MPL caused a sustained down-regulation of the receptor (mRNA and protein) and that the expression of TAT (mRNA and protein) returned back toward the baseline in the continuous presence of the drug (12, 13). If the receptor mediates the effect of the drug, then this is a rational result. Figure 5 (left) shows that the acute and chronic profiles of Odc1 come close to approximating those for TAT. Similarly, Fig. 6 (left) to some degree approximates this result for down-regulation. However, the major question posed by a perusal of the remaining profiles is: how can the drug continue to have sustained high-level effects when the receptor is greatly diminished to the point of almost being gone?

There are a number of possible explanations for more complex expression profiles that do not show tolerance. One rational possibility is that our concept of the structure and function of the GRE is entirely too simplistic. If multiple GREs with greatly different affinities for the drug receptor complex exist, then the type of result seen for Kclr in Fig. 6 (right) could be explained. A second and related possibility is that the GR is not a single entity. This possibility is supported by the recent report by Lu and Cidlowski (38), who showed the existence of multiple GR isoforms with different trafficking and transcriptional activities. It is possible that different isoforms may have different GRE binding affinities. Another possibility is that our concept that only the GR can mediate the effects of corticosteroids may be simplistic and that some other receptor can mediate the effects of these drugs. Alternatively, secondary or tertiary effects may involve glucocorticoid modulation of a secondary biosignal (which likely could be a different transcription factor) that in turn would modulate transcriptional activity. In addition, changes in the physiological states of the animal (such as altered glucose or lipid levels) with chronic treatment could also act as an additional biosignal that leads to secondary and tertiary changes in gene expression. This possibility is suggested by the chronic profile for Tdo seen in Fig. 5. Initially, it appears as if Tdo is going to respond with a pattern similar to Odc1 and return to baseline, but the expression is enhanced again to even a higher level that is maintained throughout the remainder of the infusion period. In any case, the results demonstrate that at present there is much we do not know about how corticosteroids influence gene expression.

We attempted to use domain knowledge to sort the 358 genes into groups, which are presented in Tables 113. In particular, Tables 1 and 2 demonstrate the broad impact these drugs have on transcription and translation as well as signaling. These results provide interesting examples of the type of physiological coordination that exists. Corticosteroids cause both insulin resistance and gluconeogenesis. The enhanced expression of Pdk along with the down-regulation of Pdp should maintain the pyruvate dehydrogenase complex in an inactive state, preventing pyruvate from entering the mitochondrial TCA cycle. The down-regulation of malic enzyme would also contribute to the cellular depletion of pyruvate. In contrast, the expression of aldolase B is enhanced, contributing to gluconeogenesis. Likewise, the down-regulation of complement protein C1q along with the enhanced expression of C1q binding protein demonstrates similar coordination, as does the enhanced expression of both Odc and Oazin.

Tables 113 present lists of genes regulated by both acute and chronic corticosteroid treatment. In these tables, we qualitatively list the response of each gene as down, up, down/up, or up/down to both acute and chronic treatments. We have not attempted to include a quantitative measure of magnitude of change for each gene in the two treatments. We feel that to choose a single time point to define a maximum or minimum would be potentially misleading, because it is the response pattern over time that is a true measure of magnitude of response. For example, one probe set response may show a sharp peak that rises and declines over a 4-h period, whereas a different gene may respond with a broader but shallower peak over an extended time range. In such a case, presenting magnitude of change at a single time is not a valid comparison of expression levels of the two. The situation becomes more complicated when one considers complex patterns of regulation (i.e. initial up-regulation followed by later down-regulation or vice versa).

All datasets described in this and related cited publications are available online in GEO. In addition, all data are available online at the Public Expression Profiling Resource site (http://pepr.cnmcresearch.org) developed and maintained by the Hoffman laboratory at Children’s National Medical Center (39). These data are available to all researchers in a user-friendly format, where individual temporal profiles are searchable, and all data can be obtained and used without requirement for any additional specialized software.

Acknowledgments

This dataset was developed at the Children’s National Medical Center under the auspices of a grant from the National Heart, Lung, and Blood Institute/National Institutes of Health Programs in Genomic Applications HL 66614 (Eric P. Hoffman, PI). This work was supported by GM 24211 and GM 67650 from the National Institute of General Medical Sciences, National Institutes of Health, Bethesda, MD.

Abbreviations

GR

Glucocorticoid receptor

GRE

glucocorticoid response element

MPL

methylprednisolone

TAT

tyrosine aminotransferase

TCA

tricarboxylic acid

Footnotes

Disclosure Statement: The authors have nothing to disclose.

References

  • 1.Bialis MC, Routledge PA. Adverse effects of corticosteroids. Adverse Drug React Toxicol Rev. 1998;17:227–235. [PubMed] [Google Scholar]
  • 2.Frauman A. An overview of the adverse reactions to adrenal corticosteroids. Adverse Drug React Toxicol Rev. 1996;15:203–206. [PubMed] [Google Scholar]
  • 3.Locsey L, Asztalos L, Kincses Z, Gyorfi F, Berczi C. Dyslipidaemia and hyperlipidaemia following renal transplantation. Int Urol Nephrol. 1996;28:419–430. doi: 10.1007/BF02550506. [DOI] [PubMed] [Google Scholar]
  • 4.Reynolds RM, Walker BR. Human insulin resistance: the role of glucocorticoids. Diabetes Obes Metab. 2003;5:5–12. doi: 10.1046/j.1463-1326.2003.00221.x. [DOI] [PubMed] [Google Scholar]
  • 5.Wald JA, Farr RS. Abnormal liver-function tests associated with long-term systemic corticosteroid use in subjects with asthma. J Allergy Clin Immunol. 1991;88:277–278. doi: 10.1016/0091-6749(91)90340-t. [DOI] [PubMed] [Google Scholar]
  • 6.Almon RR, Dubois DC, Jin JY, Jusko WJ. Pharmacogenomic responses of rat liver to methylprednisolone: an approach to mining a rich microarray time series. AAPS J. 2005;7:E156–E194. doi: 10.1208/aapsj070117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Almon RR, DuBois DC, Piel WH, Jusko WJ. The genomic response of skeletal muscle to methylprednisolone using microarrays: tailoring data mining to the structure of the pharmacogenomic time series. Pharmacogenomics. 2004;5:525–552. doi: 10.1517/14622416.5.5.525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Almon RR, Lai W, DuBois DC, Jusko WJ. Corticosteroid-regulated genes in rat kidney: mining time series array data. Am J Physiol Endocrinol Metab. 2005;289:E870–E882. doi: 10.1152/ajpendo.00196.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Friedman N, Kaminski N. Statistical methods for analyzing gene expression data for cancer research. Ernst Schering Res Found Workshop. 2002;38:109–131. doi: 10.1007/978-3-662-04747-7_6. [DOI] [PubMed] [Google Scholar]
  • 10.Kaminski N. Microarray analysis of idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol. 2003;29:S32–S36. [PubMed] [Google Scholar]
  • 11.Lock C, Hermans G, Pedotti R, Brendolan A, Schadt E, Garren H, Langer-Gould A, Strober S, Cannella B, Allard J, Klonowski P, Austin A, Lad N, Kaminski N, Galli SJ, Oksenberg JR, Raine CS, Heller R, Steinman L. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med. 2002;8:500–508. doi: 10.1038/nm0502-500. [DOI] [PubMed] [Google Scholar]
  • 12.Ramakrishnan R, DuBois DC, Almon RR, Pyszczynski NA, Jusko WJ. Fifth-generation model for corticosteroid pharmacodynamics: application to steady-state receptor down-regulation and enzyme induction patterns during seven-day continuous infusion of methylprednisolone in rats. J Pharmacokinet Pharmacodynam. 2002;29:1–24. doi: 10.1023/a:1015765201129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ramakrishnan R, DuBois DC, Almon RR, Pyszczynski NA, Jusko WJ. Pharmacodynamics and pharmacogenomics of methylprednisolone during 7-day infusions in rats. J Pharmacol Exp Ther. 2002;300:245–256. doi: 10.1124/jpet.300.1.245. [DOI] [PubMed] [Google Scholar]
  • 14.Mavrothalassitis G, Tzimagiorgis G, Mitsialis A, Zannis V, Plaitakis A, Papamatheakis J, Moschonas N. Isolation and characterization of cDNA clones encoding human liver glutamate dehydrogenase: evidence for a small gene family. Proc Natl Acad Sci USA. 1988;85:3494–3498. doi: 10.1073/pnas.85.10.3494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sun YN, DuBois DC, Almon RR, Jusko WJ. Fourth-generation model for corticosteroid pharmacodynamics: a model for methylprednisolone effects on receptor/gene-mediated glucocorticoid receptor down-regulation and tyrosine aminotransferase induction in rat liver. J Pharmacokinet Biopharmaceut. 1998;26:289–317. doi: 10.1023/a:1023233409550. [DOI] [PubMed] [Google Scholar]
  • 16.Jin JY, DuBois DC, Almon RR, Jusko WJ. Receptor/gene-mediated pharmacodynamic effects of methylprednisolone on phosphoenolpyruvate carboxykinase regulation in rat liver. J Pharmacol Exp Ther. 2004;309:328–339. doi: 10.1124/jpet.103.061515. [DOI] [PubMed] [Google Scholar]
  • 17.Law GL, Itoh H, Law DJ, Mize GJ, Merchant JL, Morris DR. Transcription factor ZBP-89 regulates the activity of the ornithine decarboxylase promoter. J Biol Chem. 1998;273:19955–19964. doi: 10.1074/jbc.273.32.19955. [DOI] [PubMed] [Google Scholar]
  • 18.Sun YN, DuBois DC, Almon RR, Pyszczynski NA, Jusko WJ. Dose-dependence and repeated-dose studies for receptor/gene-mediated pharmacodynamics of methylprednisolone on glucocorticoid receptor down-regulation and tyrosine aminotransferase induction in rat liver. J Pharmacokinet Biopharmaceut. 1998;26:619–648. doi: 10.1023/a:1020746822634. [DOI] [PubMed] [Google Scholar]
  • 19.McKay LI, DuBois DC, Sun YN, Almon RR, Jusko WJ. Corticosteroid effects in skeletal muscle: gene induction/receptor autoregulation. Muscle Nerve. 1997;20:1318–13120. doi: 10.1002/(sici)1097-4598(199710)20:10<1318::aid-mus17>3.0.co;2-z. [DOI] [PubMed] [Google Scholar]
  • 20.Comings DE, Muhleman D, Dietz G, Sherman M, Forest GL. Sequence of human tryptophan 2,3-dioxygenase (TDO2): presence of a glucocorticoid response-like element composed of a GTT repeat and an intronic CCCCT repeat. Genomics. 1995;29:390–396. doi: 10.1006/geno.1995.9990. [DOI] [PubMed] [Google Scholar]
  • 21.Hu E, Liang P, Speigelman BM. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J Biol Chem. 1996;271:10697–10703. doi: 10.1074/jbc.271.18.10697. [DOI] [PubMed] [Google Scholar]
  • 22.Toth CA, Thomas P, Broitman SA, Zamcheck N. A new Kupffer cell receptor mediating plasma clearance of carcinoembryonic antigen by the rat. Biochem J. 1982;204:377–381. doi: 10.1042/bj2040377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zandbergen F, Mandard S, Escher P, Tan NS, Patsouris D, Jatkoe T, Rojas-Caro S, Madore S, Wahli W, Tafuri S, Muller M, Kersten S. The G0/G1 switch gene 2 is a novel PPAR target gene. Biochem J. 2005;392:313–324. doi: 10.1042/BJ20050636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Takiguchi M, Haraguchi Y, Mori M. Human liver-type arginase gene: structure of the gene and analysis of the promoter region. Nucleic Acids Res. 1988;16:8789–8802. doi: 10.1093/nar/16.18.8789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wang H, Huang ZQ, Xia L, Feng Q, Erdjument-Bromage H, Strahl BD, Briggs SD, Allis CD, Wong J, Tempst P, Zhang Y. Methylation of histone H4 at arginine 3 facilitating transcriptional activation by nuclear hormone receptor. Science. 2001;293:853–857. doi: 10.1126/science.1060781. [DOI] [PubMed] [Google Scholar]
  • 26.Seol W, Choi HS, Moore DD. An orphan nuclear hormone receptor that lacks a DNA binding domain and heterodimerizes with other receptors. Science. 1996;272:1336–1339. doi: 10.1126/science.272.5266.1336. [DOI] [PubMed] [Google Scholar]
  • 27.Sellar GC, Cockburn D, Reid KB. Localization of the gene cluster encoding the A, B, and C chains of human C1q to 1p34.1–1p36.3. Immunogenetics. 1992;35:214–216. doi: 10.1007/BF00185116. [DOI] [PubMed] [Google Scholar]
  • 28.Ghebrehiwet B, Lim BL, Peerschke EI, Willis AC, Reid KB. Isolation, cDNA cloning, and overexpression of a 33-kD cell surface glycoprotein that binds to the globular “heads” of C1q. J Exp Med. 1994;179:1809–1821. doi: 10.1084/jem.179.6.1809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fowlkes DM, Mullis NT, Comeau CM, Crabtree GR. Potential basis for regulation of the coordinately expressed fibrinogen genes: homology in the 5′ flanking regions. Proc Natl Acad Sci USA. 1984;81:2313–2316. doi: 10.1073/pnas.81.8.2313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hanson R, Reshef L. Regulation of phosphoenolpyruvate carboxykinase (GTP) gene expression. Annu Rev Biochem. 1997;66:581–611. doi: 10.1146/annurev.biochem.66.1.581. [DOI] [PubMed] [Google Scholar]
  • 31.Sugden MC, Holness MJ. Therapeutic potential of the mammalian pyruvate dehydrogenase kinases in the prevention of hyperglycaemia. Curr Drug Targets Immune Endocr Metabol Disord. 2002;2:151–165. [PubMed] [Google Scholar]
  • 32.Kamboh MI, Aston CE, Hamman RF. DNA sequence variation in human apolipoprotein C4 gene and its effect on plasma lipid profile. Atherosclerosis. 2000;152:193–201. doi: 10.1016/s0021-9150(99)00459-1. [DOI] [PubMed] [Google Scholar]
  • 33.Sekine T, Cha SH, Tsuda M, Apiwattanakul N, Nakajima N, Kanai Y, Endou H. Identification of multispecific organic anion transporter 2 expressed predominantly in the liver. FEBS Lett. 1998;429:179–182. doi: 10.1016/s0014-5793(98)00585-7. [DOI] [PubMed] [Google Scholar]
  • 34.Koguchi K, Kobayashi S, Hayashi T, Matsufuji S, Murakami Y, Hayashi S. Cloning and sequencing of a human cDNA encoding ornithine decarboxylase antizyme inhibitor. Biochim Biophys Acta. 1997;1353:209–216. doi: 10.1016/s0167-4781(97)00106-1. [DOI] [PubMed] [Google Scholar]
  • 35.Koguchi K, Murakami Y, Hayashi S. Involvement of antizyme-like regulatory protein in polyamine-caused repression of ornithine decarboxylase in insect-derived Trichoplusia ni 5 cells. Biochim Biophys Acta. 1997;1357:291–296. doi: 10.1016/s0167-4889(97)00033-5. [DOI] [PubMed] [Google Scholar]
  • 36.Granner D, Pilkis S. The genes of hepatic glucose metabolism. J Biol Chem. 1990;265:10173–10176. [PubMed] [Google Scholar]
  • 37.Sun YN, McKay LI, DuBois DC, Jusko WJ, Almon RR. Pharmacokinetic/pharmacodynamic models for corticosteroid receptor down-regulation and glutamine synthetase induction in rat skeletal muscle by a receptor/gene-mediated mechanism. J Pharmacol Exp Ther. 1999;288:720–728. [PubMed] [Google Scholar]
  • 38.Lu NZ, Cidlowski JA. Translational regulatory mechanisms generate N-terminal glucocorticoid receptor isoforms with unique transcriptional target genes. Mol Cell. 2005;18:331–342. doi: 10.1016/j.molcel.2005.03.025. [DOI] [PubMed] [Google Scholar]
  • 39.Chen J, Zhao P, Massaro D, Clerch LB, Almon RR, DuBois DC, Jusko WJ, Hoffman EP. The PEPR GeneChip data warehouse, and implementation of a dynamic time series query tool (SGQT) with graphical interface. Nucleic Acids Res. 2004;32:D578–D581. doi: 10.1093/nar/gkh003. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES