Skip to main content
International Journal of Clinical and Experimental Medicine logoLink to International Journal of Clinical and Experimental Medicine
. 2008 Jun 15;1(3):213–247.

Prevention of Trauma and Hemorrhagic Shock-Mediated Liver Apoptosis by Activation of Stat3α

Ana Moran 1, Ayse Akcan Arikan 2, Mary-Ann A Mastrangelo 1, Yong Wu 1, Bi Yu 1, Valeria Poli 3, David J Tweardy 1,4
PMCID: PMC2581418  NIHMSID: NIHMS73928  PMID: 18997875

Abstract

Trauma is a major cause of mortality in the United States. Death among those surviving the initial insult is caused by multiple organ failure (MOF) with the liver among the organs most frequently affected. We previously demonstrated in rodents that trauma complicated by hemorrhagic shock (trauma/HS) results in liver injury that can be prevented by IL-6 administration at the start of resuscitation; however, the contribution of the severity of HS to the extent of liver injury, whether or not resuscitation is required and the mechanism for the IL-6 protective effect have not been reported. In the experiments reported here, we demonstrated that the extent of liver apoptosis induced by trauma/HS depends on the duration of hypotension and requires resuscitation. We established that IL-6 administration at the start of resuscitation is capable of completely reversing liver apoptosis and is associated with increased Stat3 activation. Microarray analysis of the livers showed that the main effect of IL-6 was to normalize the trauma/HS-induced apoptosis transcriptome. Pharmacological inhibition of Stat3 activity within the liver blocked the ability of IL-6 to prevent liver apoptosis and to normalize the trauma/HS- induced liver apoptosis transcriptome. Genetic deletion of a Stat3β, a naturally occurring, dominant-negative isoform of the Stat3, attenuated trauma/HS-induced liver apoptosis, confirming a role for Stat3, especially Stat3α, in preventing trauma/HS-mediated liver apoptosis. Thus, trauma/HS-induced liver apoptosis depends on the duration of hypotension and requires resuscitation. IL-6 administration at the start of resuscitation reverses HS-induced liver apoptosis, through activation of Stat3α, which normalizes the trauma/HS-induced liver apoptosis transcriptome.

Keywords: Nucleosomes, TUNEL, expression Microarray, transcriptome

Introduction

Trauma is the leading cause of death for those under 45 years old in the United States [1]. While almost half of the deaths occur at the time of the injury, the leading cause of death among those surviving the initial insult is multiple organ failure (MOF) [2, 3]. The liver is one of the organs most frequently affected by trauma and hemorrhagic shock, and its central role in metabolism and homeostasis makes this organ a critical one for survival of the host after severe injury [4, 5].

We previously demonstrated in rats and mice that trauma complicated by hemorrhagic shock (trauma/HS) results in liver injury as evidence by hepatocyte apoptosis [6], liver necrosis [7] and elevated transaminases [8]; however, the contribution of the severity of hemorrhagic shock to the extent of liver injury and whether or not resuscitation is required for liver injury to occur have not been reported. We also previously demonstrated that administration of IL-6 at the start of resuscitation prevented liver apoptosis and necrosis [6, 7]. IL-6 activates two anti-apoptotic signaling pathways, one involving Akt and the other involving signal transducer and activator or transcription (STAT)3. Whether or not one or both pathways are involved in the anti-apoptotic effect of IL-6 has not been determined.

In the experiments reported here, we investigated the hypotheses: 1) that trauma/HS-induced liver apoptosis depends on the severity of hemorrhagic shock and requires resuscitation; and 2) that the protective effect of IL-6 administration is mediated by Stat3. We demonstrated that the extent of liver apoptosis induced by our model of trauma/HS depends on the duration of hypotension and requires resuscitation. We established that IL-6 administration at the start of resuscitation following the longest duration of hypotension is capable of completely reversing liver apoptosis and is associated with increased Stat3 activation. Microarray analysis of the livers showed that the main effect of IL-6 was to normalize the trauma/HS-induced apoptosis transcriptome. Pharmacological inhibition of Stat3 activity within the liver blocked the ability of IL-6 to prevent liver apoptosis and to normalize the trauma/HS-induced liver apoptosis transcriptome. Genetic deletion of a Stat3β, a naturally occurring, dominant-negative isoform of the Stat3, attenuated trauma/HS-induced liver apoptosis, confirming a role for Stat3, especially Stat3α, in preventing trauma/HS-mediated liver apoptosis. Thus, trauma/HS-induced liver apoptosis depends on the duration of hypotension and requires resuscitation. IL-6 administration at the start of resuscitation reverses HS-induced liver apoptosis, through activation of Stat3α, which normalizes the trauma/HS-induced liver apoptosis transcriptome.

Materials and Methods

Rat and mouse protocols for trauma plus hemorrhagic shock

These studies were approved by the Baylor College of Medicine Institutional Review Board for animal experimentation and conform to National Institutes of Health guidelines for the care and use of laboratory animals. Adult male Sprague-Dawley rats were obtained from Harlan (Indianapolis, IN). Stat3β homozygous-deficient (Stat3βΔ/Δ) mice were generated as described [9] and re-derived at Jackson labs. Pups from heterozygous matings were tailed and genotyped by PCR, as described, with minor modifications [9].

Eight-week old male Sprague-Dawley rats (200–250 gm) were used for all experiments in this study. Rats were subjected to the sham or hemorrhagic shock (HS) protocols, as described [10, 11] with modifications. Blood was withdrawn into a heparinized syringe episodically to maintain the target MAP at 35 mmHg until blood pressure compensation failed. Blood was then returned as needed to maintain the target MAP. The amount of shed blood returned (SBR) defined 5 different levels of shock severity reflected in the duration of hypotension: 0% SBR (SBR0) represented the lowest level of shock severity (duration of hypotension, 78 ± 2.5 minutes), 10% SBR (SBR10; duration of hypotension, 149 ± 41.4 minutes), 20% SBR (SBR20; duration of hypotension, 165 ± 32.7 minutes), 35% SBR (SBR35; duration of hypotension, 211 ± 7.6 minutes), and 50% SBR (SBR50; duration of hypotension, 273 ± 24.9 minutes). At the end of the hypotensive period, rats were resuscitated as described [10, 11] and humanely sacrificed 60 minutes after the start of resuscitation. Where indicated, rats received 10 μg/kg of recombinant human IL-6 in 0.1 ml PBS at the initiation of the resuscitation or PBS alone. Sham rats were anesthetized and cannulated for 250 minutes but were not subjected to hemorrhage or resuscitation. One group of rats (UHS) was subjected to the most severe hemorrhagic shock protocol (50% SBR), but not resuscitated and kept at the target MAP (35 mmHg) for an additional 60 minutes (duration of hypotension = 336 ± 10.3 minutes) before sacrifice.

Stat3βΔ/Δ mice and wild-type littermate mice were subjected to a trauma/HS protocol [8, 12], which was similar to the rat protocol except that the target MAP in the mouse was 30mm Hg and the duration of hypotension was 180 min in all mice. Sham mice were anesthetized and immobilized in a pair-wise fashion with HS mice and sacrificed at the same time as their HS companion.

Rat and mouse livers were harvested immediately after sacrifice. The right liver lobe was fixed with paraformaldehyde solution (2%) for histologic analysis and the left lobe was snap frozen in liquid nitrogen for protein and RNA extraction.

In vivo pharmacological inhibition of Stat3

To achieve pharmacological inhibition of Stat3 activity within the lungs of rats, rats were randomized to receive by tail vein injection the G-rich, quartet-forming oligodeoxynucleotide (GQ-ODN) T40214 or nonspecific (NS)-ODN (2.5 mg ODN/kg) complexed in polyethyleneimine, as described [13], 24 hours prior to subjecting them to the SBR50 protocol with IL-6 treatment. The half-life of T40214 in tissues is ≥ 48 hr [14].

Nucleosome ELISA

Levels of histone-associated DNA fragments (nucleosomes) were determined in liver homogenates with an ELISA method (Cell Death Detection ELISAplus; Roche Diagnostics, Manheim, Germany). Frozen livers were cut by cryotome into 5 micron sections and resuspended in cell lysis buffer using the reagents from the ELISA kit. The lysates were sonicated in ice 3 times, 10 seconds each, centrifuged and supernatants harvested. Total protein concentration of each supernatant was determined by Bradford assay (Bio-Rad Protein Assay, Bio-Rad Laboratories, Inc., Hercules, CA). Equal amounts of protein (200ug) were loaded into microtiter wells in duplicate. A positive control (lyophilized, stabilized nucleosome concentrate of known concentration, provided in the kit) and a negative control (water) were also loaded in duplicate. Serial dilutions of the positive control were loaded in duplicate and used to plot a standard curve. The nucleosome concentration for each sample was obtained by plotting each sample duplicate's OD against the standard curve. The final sample nucleosome concentration was the average of the duplicates [15]. The rest of the assay was performed according to manufacterer's instructions.

Terminal deoxynucleotidyl transferase (TdT) mediated nick end labeling (TUNEL) staining

TUNEL staining to enzymatically detect the free 3'-OH termini was performed using the ApopTag Plus Peroxidase in situ Apoptosis Detection Kit from Chemicon International. Slides were rehydrated from Xylene to PBS through a series of decreasing concentrations of ethanol and digested in proteinase K (20 ug/ml) for 3 minutes at 23°C. Endogenous peroxidases were quenched for 30 minutes in 3% hydrogen peroxide in PBS. TdT enzyme was diluted in TUNEL solution buffer then used as suggested by the manufacturer. Slides were counterstained with hematoxyllin. TUNEL positive cells were assessed microscopically by counting the total nuclei and the number of TUNEL-positive nuclei in twenty random 1000x fields by an experienced histologist, blinded to the treatment each rat received. Data is presented as the number of TUNEL positive cells per high power field (hpf).

Immunoblotting

Levels of STAT3 activation within the livers of rats were assessed by immunoblotting using whole-tissue extracts of liver sections with mouse monoclonal antibody to Tyr705 phosphorylated (p)STAT3 (Cell Signaling Technology, Inc., Danvers, MA; 1:1000 dilution). Briefly, frozen livers were cut by cryotome into 5 micron sections and resuspended in cell lysis buffer (Cell Death Detection ELISAplus Kit, Roche Diagnostics, Manheim, Germany). The supernatant was sonicated in ice 3 times, 10 seconds each. Samples were then centrifuged and the supernatant evaluated by Bradford assay for total protein quantification. Protein samples (60ug total protein) were separated by SDS- PAGE and transferred to a PVDF membrane. The membrane was incubated overnight with mouse monoclonal antibody and subsequently incubated with goat antimouse antibody with horseradish peroxidase (HRP) conjugate (Zymed, San Francisco, CA) for 1 hour. ECL agent (Amersham Biosciences, UK) was used for detection. The membrane was then stripped (using RestoreTM Western Blot Stripping Buffer, PIERCE, Rockford, IL) and immuoblotting performed to detect total STAT3 protein in the whole tissue extracts of livers using mouse IgG1 monoclonal antibody to STAT3 (BD Biosciences, Rockville, MD). Densitometry was performed using ImageQuant TL v2005 software (Amersham Biosciences, Buckinghamshire, England). Results are expressed as the ratio of pSTAT3 signal (after background signal subtraction) to total STAT3 signal (after background signal subtraction) for each sample.

RNA isolation and microarray hybridization and analysis procedures

Total RNA was isolated from 4–5 micron cryotome sections of liver using TRIzol® Reagent (Invitrogen, Carlsbad, California) single step RNA isolation protocol followed by purification with RNeasy® Mini Kit (QIAGEN, Hilden, Germany) as instructed by the RAE 230A following Affymetrix protocols used within the Baylor College of Medicine Microarray Core Facility. Gene expression profiling was performed with the Affymetrix Rat Array.

Microarray Analysis

We used Affymetrics GCOS, dChip and Array Analyzer (Insightful Corporation) software packages for quality assessment and statistical analysis and annotation. Expression estimation and group comparisons were done with Array Analyzer. Low-level analyses included background correction, quartile normalization and expression estimation using GCRMA [16]. One-way analysis of variance (ANOVA) with contrasts [17] was used for group comparisons on all genes and on the list of apoptosis related genes only. P-values were adjusted for multiple comparisons using the Benjamini-Hockberg method [18]. The adjusted p-values represent false discovery rates (FDR) and are estimates of the proportion of “significant” genes that are false or spurious “discoveries”. We used a FDR=10% as cut-off.

Statistical Analysis

Data are presented as mean ± standard error of the mean (SEM). Multiple group comparisons of means were done by one-way analysis of variance (ANOVA). Post hoc analysis was done by Student-Newman-Keuls test for 2-group comparisons of means. Correlation between duration of hypotension and nucleosome levels was done for each individual study animal by Pearson correlation coefficient. Goodness of fit was evaluated by R-square. All statistical analyses were done on SigmaStat 3.5 (SYSTAT Software Inc., Chicago, IL).

Results

HS-induced liver apoptosis depends on the severity of shock

To confirm our previous findings that trauma/HS induces liver apoptosis, to determine if apoptosis is an early event following trauma/HS as well as to evaluate the contribution of the severity of shock in our rat model of trauma/HS, we measured histone-associated DNA fragments (nucleosomes) in the livers of rats subjected to increasing severity of shock 1 hr after the initiation of resuscitation. Nucleosome levels increased exponentially with increasing duration of shock (Pearson correlation coefficient 0.879, p<0.001) with the level of nucleosomes in the SBR50 group (1817.3 ± 105.9 units/ml) achieving a level 13.1 times higher than sham (139 ± 67 units/ml; p<0.001, ANOVA; Figure 1). Thus, trauma/HS-induced liver apoptosis occurs within 1 hr of resuscitation and depends on the severity of shock.

Figure 1.

Figure 1

Effect of shock severity on liver apoptosis. Rats were subjected to sham protocol (S) or to trauma/HS protocol with increasing duration of shock as indicated followed by resuscitation. The livers were harvested 60 minutes after the start of resuscitation. Nucleosome levels were measured in protein extracts of frozen sections of each liver and the results plotted after correction for total protein, as a function of the duration of the hypotensive period for each animal. Curve fitting was performed and the best-fitting curve shown; nucleosome levels increased exponentially with duration of hypotension (Pearson correlation coefficient=0.879, p<0.001).

Trauma/HS-induced liver apoptosis requires resuscitation

To determine the specific contribution of resuscitation to liver apoptosis, we assessed nucleosome levels as well as number of TUNEL-positive cells in the livers of rats subjected to HS without resuscitation (UHS group) and compared these results with those obtained in the sham and the resuscitated SBR50 groups. The level of nucleosomes in the UHS group (193 ± 36 units/mg total protein) was statistically indistinguishable from that of the sham group (139 ± 67 units/mg total protein; Figure 2A). Similar results were obtained when liver apoptosis was assessed by TUNEL staining. The number of TUNEL- positive nuclei/hpf in the UHS group (1.4 ± 0.5; Figure 2B, C) similar to that of the sham group (0.9 ± 0.4). In addition, histological evaluation of the cells containing TUNEL-positive nuclei revealed that more than 80% were hepatocytes, a key parenchyma cell. Thus, apoptosis within the liver following trauma/HS requires resuscitation. The fact that no liver apoptosis occurs without resuscitation suggests that complete prevention of liver apoptosis may occur with an appropriate intervention introduced at the start of resuscitation.

Figure 2.

Figure 2

Effect of resuscitation, IL-6 treatment and GQ-ODN pre-treatment on HSinduced liver apoptosis. Rats were subjected to sham protocol (Sham, n=3), unresuscitated hemorrhagic shock (UHS, n=3), HS treated with placebo at the beginning of resuscitation (SBR50, n=4), HS treated with IL-6 at the beginning of resuscitation (SBR50/IL-6, n=4), HS preceded by treatment with GQ oligodeoxynucleotide (GQ-ODN) 24 hours prior to resuscitation with IL-6 (SBR50/IL-6/G, n=3), or HS preceded by treatment with nonspecific-ODN (NS-ODN) 24 hours prior to resuscitation with IL-6 (SBR50/I-6/N, n=3). The livers were harvested 60 minutes after the start of resuscitation. Nucleosome levels were measured in protein extracts of frozen sections of each liver (Panel A). Data presented are mean + SEM of nucleosome level corrected for total protein for each group. Bars marked with an asterisk (*) differ significantly within the pair (p<0.05). In panel B, sections of paraformaldehyde-fixed liver were stained using the TUNEL assay. Representative photomicrographs of 1000x fields of liver specimens from each experimental group are shown. Apoptotic nuclei are indicated by arrows. In panel C, TUNEL-positive nuclei were counted; data shown are the mean ± SEM number of TUNELpositive nuclei per 1000x fields (20 fields counted). Bars marked with an asterisk (*) differ significantly within the pair (p<0.05).

IL-6 administration at the beginning of resuscitation prevents trauma/HS-induced liver apoptosis through activation of Stat3

In our mouse model of HS, we have previously demonstrated that IL-6 administration at the beginning of resuscitation prevented the development of HS-induced liver apoptosis detected 24 hrs after HS [6]. To confirm these findings and to gain an improved molecular and cellular understanding of the anti-apoptotic effects of IL-6, we measured apoptotic cell death in rats subjected to trauma/HS with the most severe HS protocol (50% SBR) and randomly assigned to receive either PBS (SBR50) or IL-6 (10 μg/kg, SBR50/IL-6) at the beginning of resuscitation. Nucleosome levels in the SBR50/IL-6 group (264 ± 36 units/ml) were decreased 3.3 times compared to those of the SBR50 group (874 ± 127 units/ml, p<0.001) and were similar to sham levels (139 ± 67 units/ml; Figure 2A). TUNEL staining confirmed these results. The number of TUNEL-positive nuclei/hpf in the SBR50/IL-6 group (1.9 ± 0.5) was decreased 14.2 times compared to the SBR50 group (27 ± 3.6, p<0.001), to levels statistically similar to those of the sham group (0.9 ± 0.4; Figures 2B and 2C). Thus, IL-6 administration at the beginning of resuscitation prevents trauma/HS-induced liver apoptosis occurring 1 hr after trauma/HS in rats as well as 24 hrs after trauma/HS in mice [6].

IL-6 binding to IL-Rα and gp130 results in gp130 dimerization and phosphorylation of gp130-associated protein-tyrosine kinases Jak1, Jak2, and Tyk2, which is followed by activation of two major signaling pathways within cells—Stat3 and SHP-2/Grb-2/ERK [19]. The SHP-2/Grb2/ERK pathway bifurcates resulting in activation of p38MAPK and PI-3K. Stat3 and P-I3K/Akt activation, but not p38MAPK, link to anti-apoptotic effects within cells. Stat3 mediates its anti-apoptotic effect in cancer cells through its ability to up-regulate anti-apoptotic genes such as Bcl-xL, Bcl-2 and Mcl-1 [20]. Akt is a highly promiscuous kinase with a large number of binding partners and targets [21] that posttranslationally modify transcription factor systems such as Forkhead [22, 23], IκB/NF-κB and cyclic AMP response element binding protein (CREB) [24], which together result in increased transcription of survival genes and decreased transcription of apoptotic genes [25]. To assess if the anti-apoptotic effects of IL-6 in the liver is mediated by Stat3 activation, we first determined if Stat3 is activated in the livers of rats resuscitated with IL-6. Extracts of cryotome sections of the liver harvested 1 hour after IL-6 treatment were examined by immunoblotting with mouse monoclonal antibody to Tyr705 phosphorylated (p)Stat3 (Figure 3A). Densitometric analysis of the signal intensity of the pStat3 bands normalized for total Stat3 indicated that Stat3 activity is increased 1.7 fold in the livers of IL- 6-treated rats compared to placebo-treated rats (p=0.002, ANOVA; Figure 3B).

Figure 3.

Figure 3

Effect of IL-6 treatment and GQ-ODN pre-treatment on Stat3 activity within the livers. Rats were subjected to the sham protocol or HS protocol and treated with placebo at the beginning of resuscitation (SBR50), HS treated with IL-6 at the beginning of resuscitation (SBR50/IL-6), HS preceded by treatment with GQ-oligodeoxynucleotide (GQ-ODN) 24 hours prior to resuscitation with IL-6 (SBR50/IL-6/G), or HS preceded by treatment with nonspecific-ODN (NS-ODN) 24 hours prior to resuscitation with IL-6 (SBR50/IL-6/N). The livers were harvested 60 minutes after the start of resuscitation. In panel A, protein extracts of whole liver were separated by SDS-PAGE and immunoblotted for phosphorylated (p)Stat3 and total Stat3 (NC = negative control, HepG2 cells incubated with PBS for 30 minutes prior to protein extraction; PC = positive control, HepG2 cells incubated with IL-6, 30 ng/ml for 30 minutes prior to protein extraction). In panel B, the pStat3 and total Sta3 bands were quantitated by densitometry and data presented as mean ± SEM of pStat3 signal corrected for total Stat3 signal for each group. Bands representing Stat3α, Stat3β and Stat3δ are indicated on the right [54, 55]. Bars marked with an asterisk (*) differ significantly within the pair (p<0.0001).

To further evaluate the role of Stat3 downstream of IL-6 in mediating its anti-apoptotic effects in the liver, we examined whether or not these effects of IL-6 could be reversed by pretreatment of rats with a G-rich oligodeoxynucleotide, G-quartet (GQ)-ODN, T40214, a novel Stat3 inhibitor, that forms a rigid G-quartet structure within cells and inhibits the growth of tumors in which Stat3 is constitutively activated [13, 14, 26, 27]. Rats were treated in a blinded fashion with GQ-ODN (SBR50/IL-6/G group) or nonspecific (NS) ODN (SBR50/IL-6/N group) 24 hours prior to being subjected to HS and resuscitation with IL-6. Pre-treatment with GQ-ODN reduced Stat3 activity within the livers of HS/I/G rats 1.9-fold compared to HS/I/N rats (Figures 3A, B). Importantly, the inhibition of Stat3 activation within the livers of the SBR50/IL-6/G rats was accompanied by a return of nucleosomes (1556 ± 241 units/ml) to levels similar to those of the placebo treated (SBR50) group (1874 ± 127 units/ml, p>0.05) and 11.2 fold higher that those of the IL-6 treated (SBR50/IL-6) group (264 ± 36 units/ml, p<0.001; Figure 2A). Similarly, the number of TUNEL-positive nuclei/hpf in livers of rats from the SBR50/IL-6/G group (12.3 ± 1.1) was 6 fold higher than that of the SBR50/IL-6 group (1.9 ± 0.5, p<0.0001); Figures 2B and C). Nucleosome levels and number of TUNEL- positive nuclei/hpf in livers of rats pre-treated with NS-ODN were indistinguishable from those of the SBR50/IL-6 group (Figures 2A and B). Thus, pharmacological inhibition of Stat3 using GQ-ODN in rats subjected to severe HS resuscitated with IL-6 markedly attenuated IL-6- mediated Stat3 activation and prevention of liver apoptosis.

Two isoforms of Stat3 are expressed in all cells—α (p92) and β (p83)—both derived from a single gene by alternative mRNA splicing with Stat3α predominating [28]. Stat3α functions as an oncogene [29] in part through inhibiting apoptosis, while Statβ antagonizes the oncogenic function of Stat3α [30]. While mice deficient in both isoforms of Stat3 are embryonic lethal at day 6.5 to 7 [31] and mice deficient in Stat3α die within 24 hr of birth, mice deficient in Stat3β have normal survival and fertility [9]. To further support the hypothesis that Stat3, in particular Stat3α, contributes to resistance to apoptosis within the liver in the setting of HS, we subjected Stat3β homozygous-deficient (Stat3βΔ/Δ) mice and their littermate control wild type mice to a severe HS protocol (target MAP 30 mm Hg for 5 hr) and examined their livers for nucleosome levels 1 hr after the start of resuscitation. As expected, nucleosome levels in wild type HS mice (1027.3 ± 273.3 mU/mg total protein) were increased compared to wild type sham mice (210.3 ± 29.8; p < 0.01; Figure 4B). In contrast, however, nucleosome levels in the livers of Stat3βΔ/Δ HS (463.9 ± 3.9) mice were reduced 2.2 times compared to wild type HS mice and were similar to wild type sham mice (Figure 4B). These findings indicate that Stat3, in particular Stat3α, protects the liver from apoptosis in the setting of trauma/HS.

Figure 4.

Figure 4

Effect of Stat3β ablation on trauma/HS- induced liver apoptosis. Stat3β homozygous-deficient (Stat3βΔ/Δ) mice and their littermate control wild type mice were subjected to the murine trauma/HS protocol or sham protocol and their livers harvested 1 hr after the start of resuscitation. Nucleosome levels were measured in protein extracts of frozen sections of the liver and the results corrected for total protein. Data presented are the means ± SEM of each group (n ≥ 3). Significant differences are indicated (Student's t-test).

Microarray analysis of the liver transcriptome focusing on differential expression of apoptosis-related genes

In addition to increasing the transcription of anti-apoptotic genes (Bcl-xL, Bcl-2, and Mcl-1) [29, 3239], Stat3 has been shown to decrease transcription of pro-apoptotic genes (Bad, Bnip3l, Casp3). To evaluate the role of Stat3 downstream of IL-6 at the transcriptome level, and to identify genes altered within the livers of animals subjected to trauma/HS especially those involved in apoptosis in a global and unbiased manner, we performed Affymetrix oligonucleotide microarray analysis with RAE 230A chips. Fifteen chips were hybridized using mRNA isolated from 4 livers each from sham, SBR50, and SBR50/IL-6 groups, and 3 livers from SBR50/IL-6/G groups. All fifteen chips were included in the normalization and expression estimation steps of the analysis and were included in the statistical analysis and differential expression comparison. The 15,866 probesets on the RAE 230A chip represent 9,818 annotated genes or expressed sequence tags, including 860 apoptosis-related genes. The list of 860 apoptosis-related genes present on the RAE 230A (Table 1) was created by combining gene lists obtained by querying annotation databases provided in GeneSpring and dChip, which were derived from the Gene Ontology (GO) Consortium.

Table 1.

Apoptosis-related genes examined in the microarray experiments

# Accession Gene Name Gene Symbol
1 BF417479 24-dehydrocholestero reductase Dhcr24
2 NM_022225 5-hydroxtryptamine (serotomin) receptor IB Etsl
3 NM_030870 8-oxoguanine DNA-glycosylase 1 Oggl
4 NM_020306 a distegnin and metalloproteinase domain 17 (tumor necrosis factor, alpha, converting enzyme) Adaml7
5 NM_131911 acidic nuclear phosphoprotein 32 family, member B Anp32b
6 NM_012912 activating transcription factor 3 Atf3
7 BM391471 activating transcription factor 5 Atf5
8 NM_019361 activity regulated cytoskeletal-associated protein Atf3
9 AI600029 activity-dependent neuroprotective protein Adnp
10 NM_017155 adenosine A1 receptor Adoral
11 AF228684 adenoaine A2a receptor Adora2a
12 NM_012896 adenosine A3 receptor Adora3
13 NM_031006 adenosine deaminase, RNA-specific Adar
14 AW523747 adenosine molecule with Ig like domain 2 Amigo2
15 U07126 adrenergic receptor, alpha 1a Adralc
16 AY057895 adreoergic receptor, beta 2 Adrb2
17 NM_012715 adrenomedullin Adm
18 NM_134326 albumin Alb
19 NM_022407 aldehyde dehydrogenase family 1, member A1 Aldhlal
20 NM_012498 aldo-keto reductase family 1, member B4 (aldose reductase) Akrlb4
21 NM_017196 allograft inflammatory factor 1 Aifl
22 NM_012493 alpha-feroprotein Atp
23 NM_012892 amiloride-sensitive cation channel 1, neuronal (degenerin) Accnl
24 BM986220 amyloid beta (A4) precursor protein Afp
25 NM_053957 amyloid beta (A4) precursor protein-binding, family B, member 3 Apbb3
26 U90829 amyloid beta precursor protein binding protein 1 Appbpl
27 NM_012502 androgen receptor Ar
28 AF275151 androgen receptor-related apoptosis-associated protein CBL27 Cbl27
29 BI275292 angiopoietin 2 Angpt2
30 AA818262 angiopoietin-like 4 Angptl4
31 AF201331 angiotensin I converting enzyme (peptictyl-dipeptidase A) 1 Ace
32 BF552873 angiotensin II receptor, type 2 Agtr2
33 NM_031009 angiotensin receptor 1b Agtr2
34 NM_134432 angiotensinogen (serpin pepidase inhibitor, cladle A. member 8) Agt
35 AJ42B573 ankyrin 3, epithelial Ank3
36 L81174 ankyrin repeat domain 1 (cardiac muscle) Ankrdl
37 NM_012904 annexin A1 Anxal
38 NM_024155 annexin A4 Anxa4
39 NM_013132 annexin A5 Anxa5
40 BI275921 anterior pharynx defective la homolog (C. elegans) Aphla
41 NM_133400 apobec-1 complementation factor Acf
42 J02582 apolipoprotein E Apoe
43 NM_053720 apoptosis antagonizing transcription factor Aatf
44 AI233249 apoptosis inhibitor 5 (predicted) Api5_predicted
45 AW144082 Apoptosis, caspase activadon inhibitor (predicted) Amid_predicted
46 AA894233 apoptosis-inducing factor (AIF)-like mitochondrion-associated inducer of death (predicted) Amid_predicted
47 BE116857 apoptotic chromatin condensation inducer 1 Acin1
48 AF218388 apoptotic peptidase activating factor 1 Apaf1
49 L07268 aquaporin 1 Aqpl
50 NM_019158 anuaporin 8 Aqp8
51 NM_031010 arachidonate 15-lipoxygenase Aloxl5
52 BF285345 arrestin, beta 2 Arrb2
53 NM_013149 aryl hydrocarbon receptor Ahr
54 NM_012780 aryl hydrocarbon receptor nuclear translocator Amt
55 NM_021590 aryl hydrocarbon receptor-interacting protein-like 1 Aipll
56 BI274345 ataxin 10 Atxn10
57 NM_058213 ATPase, Ca++ transporting, cardiac muscle, fast twitch 1 Atp2al
58 J04024 ATPase, Ca++ transporting, cardiac muscle, slow twitch 2 Atxn10
59 AY082609 ATP-binding cassette, sub-family B (MDR/TAP), member 1/ATP-binding cassette, sub-family B (MDR/TAP), member 1A Abcb1 /// Abcb1a
60 NM_017228 atrophin 1 Atul
62 AI406520 AXL receptor tyrosine kinase Axl
63 NM_021752 baculoviral IAP repeat-containing 2 Bire2
64 NM_023987 baculoviral IAP repeat-containing 3 Bire3
65 AF304333 baculoviral IAP repeat-containing 4 Bire4
66 NM_022274 baculoviral IAP repeat-containing 5 Bire5
67 NM_031328 B-cell CLL/lymphoma 10 Bel10
68 NM_016993 B-cell leukemia/lynphoma 2 Bc12
69 NM_133416 B-cell leukemia-lynphoma 2 related protein A1 Bcl2a1
70 AI172204 B-cell receptor-associated protein 29 Bcap29
71 A1409930 B-cell receptor-associated protein 31 Bcap31
72 NM_017258 B-cell translocation gene 1, anti-proliferative Btg1
73 BI288701 B-cell translocation gene 2, anti-proliferative Btg2
74 NM_139258 Bcl2 modifying factor Bmf
75 NM_053420 BCL2/adeovirus E1B 19 kDa-interacting protein 3 Bnip3
76 NM_080888 BCL2/adenovirus E1B 19 kDa-interacting protein 3-like Bnip3l
77 NM_080897 BCL2/adenovirus E1B 19 kDa-interacting protein 1 Bnip1
78 AI178277 BCL2/adenovirus E1B 19kDa-interacting protein 1, NIP2 (predicted) Buip2_predicted
79 NM_053812 BCL2-antagonist/killer 1 Bakl
80 BI280304 Bcl2-associated athanogene 1 (predicted) Bag1_predicted
81 AI231792 Bcl2-associasd athanogene 3 Bag3
82 BI282898 BC12-associated athanogene 5 Bag5
83 AF279911 bcl2-associated death promoter Bad
84 AI717547 BCL2-associated transcription factor 1 Bclafl
85 AF235993 Bcl2-associated X protein Bax
86 U72350 Bcl2-like 1 Bcl211
87 NM_053733 Bcl2-like 10 Bcl2l10
88 NM_022612 BCL2-like 11 (apoptosis facilitator) Bcl2111
89 AI227978 BCL2-like 12 (proline rich) (predicted) Bcl2112_predicted
90 AA892271 BCL2-like 13 (apoptosis facilitator) (predicted) Bcl2113_predicted
91 NM_021850 Bcl2-like 2 Bcl212
92 AF051093 Bcl-2-related ovarian killer protein Bok
93 NM_053739 beclin 1 (coiled-coil, myosin-like BCL2-interacting protein) Becul
94 AI008680 benzodiazepine receptor, peripheral Bzrp
95 NM_057130 BH3 interacting (with BCL2 family) domain, apoptosis agonist Bid3
96 AF136282 BH3 interacing domain death agonist Bid
97 AI177631 bifunctional apoptosis regulator Bfar
98 NM_012827 bone morphogenetic protein 4 Bmp4
99 BE118651 bone morphogenic protein receptor, type II (serine/threonine kinase) Bmpr2
100 AA851481 brain and reproductive organ-expressed protein Bre
101 X67108 brain derived neurotrophic factor Bdnf
102 AI169085 brain zinc finger protein Zfpl79
103 NM_017253 branched chain aminotransferase 1, cytosolic Bcat1
104 NM_022622 BRCA1 asociaced RING domain 1 Bardl
105 BF404972 Breast cancer 1 Brcal
106 NM_012931 breast cancer anti-estrogen resistance 1 Bcarl
107 NM_134413 BTB (POZ) domain containing 14B Btbd14b
108 NM_031334 cadherin 1 Cdhl
109 NM_019161 cadherin 22 Cdh22
110 AF061947 calcineurin binding protein 1 Cabin1
111 BM958511 calcium binding protein p22 Chp
112 AB070350 calcium binding protein p22 /// similar to calcium binding protein P22 (predicted) /// similar to calcium binding protein P22 (predicted) Chp /// RGD1565588_predicted /// RGD1564956_predicted
113 BF404381 Calcium/calmodulin-dependent protein kinase II, alpha Chp /// RGD1565588_predicted /// RGD1564956_predicted
114 NM_016996 calcium-sensing receptor Casr
115 NM_019152 calpain 1 Capn1
116 NM_053295 calpastatin Cast
117 NM_022399 calreticulin Calr
118 NM_032462 Calsenilin, presenilin binding protein, EF hand transcription factor Csen
119 NM_031017 cAMP responsive element binding protein 1 Crebl
120 NM_017334 cAMP responsive element modulator Crem
121 NM_012784 cannabinoid receptor 1 (brain) Cnrl
122 AW252112 carbonic anhydrase 11 Carll
123 BF281311 casein kinase 2, beta subunit Csnk2b
124 NM_057138 CASP8 and FADD-like apoptosis regulator Cflar
125 D85899 caspase 1 Casp 1
126 NM_130422 caspase 12 Casp12
127 AF136231 caspase 2 Casp2
128 BM387008 caspase 3, apoptosis related cysteine protease Casp 3
129 NM_053736 caspase 4, apoptosis-related cysteine peptidase Casp4
130 NM_031775 caspase 6 Casp6
131 BF283754 caspase 7 Casp7
132 1369262_at caspase 8 Casp8
133 BF282281 caspase 8 associated protein 2 (predicted) Casp8ap2_predicted
134 AF262319 caspase 9 Casp9
135 NM_022303 caspase recruitment domain family, member 9 Card9
136 AI136555 castration induced prostatic apoptosis-related protein 1 Cipar1
137 NM_022597 cathepsin B Ctsb
138 NM_134334 cathepsin D Ctsd
139 AI548979 cationic trypsinogen LOC286911
140 NM_024125 CCAAT/enhancer binding protein (C/EBP), beta Cebpb
141 1368813_at CCAAT/enhancer binding protein (C/EBP), delta Cebpd
142 NM_021744 CD14 antigen Cd14
143 NM_017079 CD1d1 antigen Cd1d1
144 NM_012830 CD2 antigen Cd2
145 NM_013121 CD28 antigen Cd28
146 1389997_at CD3 antigen, epsilon polypeptide (predicted) Cd3e_predicted
147 AI044631 CD3 antigen, gamma polypeptide Cd3g_predicted
148 D30795 CD38 antigen Cd38
149 AF065147 CD44 antigen Cd44
150 NM_019295 CD5 antigen Cd5
151 NM_012523 CD53 antigen Cd53
152 NM_013069 CD74 antigen (invariant polypeptide of major histocompatibility complex, class II antigen-associated) Cd74
153 NM_031755 CEA-related cell adhesion molecule 1 Ceacam 1
154 U23056 CEA-related cell adhesion molecule 1 /// CEA-related cell adhesion molecule 10 Ceacaml /// Ceacam10
155 BF284899 cell death-inducing DNA fragmentation factor, alpha submit-like effector A (predicted) Cidea_predicted
156 L24388 cell division cycle 2 homolog (S.pombe)-like 1 Cdc2
157 AI059933 Cell division cycle 25 homolog A (S. cerevisiae) Cdc2
158 NM_023026 centaurin, gamma 1 Cengla
159 NM_031530 chemokine (C-C motif) ligand 2 Cd2
160 NM_031116 chemokine (C-C motif) ligand 5 Ccl5
161 BE095824 chemokine (C-C motif) ligand 6 Cc16
162 AA945737 chemokine (C-X-C motif) receptor 4 Cxcr4
163 AI012221 chloride intracellular channel 1 clicl
164 NM_012829 cholecystokinin Cdk
165 NM_012832 cholinergic receptor, nicotinic, alpha polypeptide 7 Chma7
166 AI171615 chromosome segregation 1-like (S. cerevisiae) (predicted) Csel1_predicted
167 NM_013092 chymase 1, mast cell Cmal
168 AA957183 Citron Cit
169 BG673439 claudin 11 Cldn1l
170 AF314657 clusterin Clu
171 NM_012950 coagulation factor II (thrombin) receptor F2r
172 NM_013057 coagulation factor III F3
173 BM389673 cofilin 1, non-muscle Cfll
174 AF092207 coiled-coil domain containing 5 Ccdc5
175 U00620 colony stimulating factor 2 (granulocyte-macrophage) Csf2
176 NM_130825 comparative gene identification transcript 94 Cgi94
177 NM_032060 complement component 3a receptor 1 C3ar1
178 NM_053619 complement component 5, receptor 1 C3arl
179 AA819870 complement component 8, beta polypeptide (mapped) C8b
180 NM_057146 complement component 9 C9
181 AW916366 COP9 (constitutive photomorphogenic) homolog, subunit 3 (Arabidopsis thaliana) Cops3
182 NM_031019 corticotropin releasing hormone Crh
183 AW433973 craniofacial development protein 1 Cfdp1
184 U47922 crystallin, alpha A Cryan
185 NM_012935 crystallin, alpha B Cryab
186 AF090695 CUG triplet repeat, RNA binding protein 2 Cugbp2
187 BI284428 cullin 1 (predicred) Cnl1_predicted
188 BI295890 cullin 2 (predicted) Cul2_predicted
189 BI285751 cullin 3 (predicted) Cul3_predicted
190 NM_022683 cullin 5 Cul5
191 X64589 cyclin B1 Ccnb1
192 AW913890 cyclin E Ccne
193 NM_080885 cyclin-dependent kinase 5 Cdk5
194 NM_053891 cyclin-dependent kinase 5, regulatory submit 1 (p35) Cdk5rl
195 H31766 cyclin-dependent kinase 9 (CDC2-related kinase) Cdk9
196 AI010427 cyclin-dependent kinase inhibitor 1A Cdkn1a
197 AI013919 cyclin-dependent kinase inhibitor 1C (P57) Cdkn1c
198 AF474976 cyclin-dependent kinase inhibitor 2A Cdkn2a
199 AI409867 cystatin B Cstb
200 BG666933 cystatin C Cst3
201 NM_031327 cysteine rich protein 61 Cyr61
202 NM_023965 cytochrome b-245, beta polypeptide Cybb
203 NM_012839 cytochrome c, somatic Cycs
204 NM_012840 cytochrome c, testis Cyet
205 NM_012840 cytochrome c, testis /// phosphodiesterase 11A Cyctpd11
206 X00469 cytochrome P450, family 1, subfamily a, polypeptide 1 Cpla1
207 NM_031543 cytochrome P450, family 2, subfamily e, polypeptide 1 Cyp2el
208 BF285068 cytokine induced apoptosis inhibitor 1 Ciapin1
209 BI298817 Cytotoxic granule-associated RNA binding protein 1 Tial
210 AI169146 D4, zinc and double PHD fingers family 2 (predicted) Dpf2_predicted
211 AI408110 DEAD (Asp-Glu-Ala-Asp) box polypeptide 19 Ddx19
212 BM389310 DEAD (Asp-Glu-Ak-Asp) box polypeptide 41 (predicted) Ddx41_predicted
213 BI285645 death associated protein 3 Dap3
214 AA818353 death associated protein kinase 1 (predicted) Dapk1_predicted
215 NM_031800 death effector domain-containing Dedd
216 NM_022526 death-associated protein Dap
217 NM_022546 death-associated protein kinase 3 Dapk3
218 AI013627 defender against cell death 1 Dadl
219 NM_080482 deleted in bladder cancer chromosome region candidate 1 (human) Dbccr
220 NM_012841 deleted in colorectal carcinoma Dcc
221 NM_013097 deoxyribonuclease I Dnase1
222 AF178975 deoxyribonuclease II Dnase 2
223 NM_053907 deoxyribonuclease I-like 3 LOC681124
224 NM_022531 desmin Des
225 BE110572 diablo homolog (Drosophila) Diablo
226 AI236726 DNA fragmentation factor, alpha subunit Dffa
227 NM_053362 DNA fragmentation factor, beta subunit Dffb
228 NM_024134 DNA-damage inducible transcript 3 Ddit3
229 NM_080906 DNA-damage-inducible transcript 4 Ddit4
230 BI282224 DnaJ (Hsp40) homolog, subfamily A, member 3 LOC294513
231 BM384926 DnaJ (Hsp40) homolog, subfamily B, member 1 (predicted) Dnajbl_predicted
232 NM_012699 DnaJ (Hsp40) homolog, subfamily B, member 9 Dnajb9
233 BI285682 DnaJ (Hsp40) homolog, subfamily C, member 7 Dnajc7
234 BF406540 DnaJ (Hsp40) related, subfamily B, member 13 Dnajb13
235 L12407 dopamine beta hydroxylase Dbh
236 NM_012547 dopamine receptor 2 Drd2
237 M35077 dopamine receptor D1A Drdla
238 BE110108 dual specificicy phosphatase 1 Dusp1
239 AI172067 dual specificity phosphatase 22(predicted) Dusp22_predicted
240 U23438 dual specificity phosphatase 4 Dusp4
241 NM_133578 dual specificity phosphatase 5 Dusp5
242 NM_053883 dual specificity phosphatase 6 Dusp6
243 L24562 dynamin 2 Dnm2
244 NM_053319 dynein light chain LC8-type 1 Dynll1
245 NM_012551 early growth response 1 Egr1
246 AF115249 endothelial differentiation, sphingolipid G-protein-coupled receptor, 8 Edg8
247 NM_023090 endothelial PAS domain protein 1 Epasl
248 NM_053596 endothelin converting enzyme 1 Ecel
249 AB023896 endothelin converting enzyme-like 1 Ecell
250 X57764 endothelin receptor type B Ednrb
251 BI291645 engulfment and cell motility 3, ced-12 homolog (C. elegans) Elmo3
252 NM_012842 epidermal growth factor Egfr
253 M37394 epidermal growth factor receptor Egfr
254 AF187818 epidermal growth factor receptor /// peptidase D (mapped) Egfr /// Pepd_mapped
255 BF564277 epilepsy, progressive myoclonic epilepsy, type 2 gene alpha Epme
256 NM_017001 erythropoietin Epo
257 AA866269 Estrogen receptor 1 Esr1
258 AF042058 estrogen receptor 2 beta Esr2b
259 BF398331 estrogen receptor-binding fragment-associated gene 9 Ebag9
260 AI412114 etoposide induced 2.4 mRNA Ei24
261 NM_012660 eukaryotic translation elongation factor 1 alpha 2 Eefla2
262 AI600237 eukaryotic translation elongation factor 1 epsilon 1 (predicted) Eeflel_predicted
263 NM_053950 enkaryotic translation initiation factor 2B, subunit 4 delta Eif2b4
264 NM_053974 eukaryotic translation initiation factor 4E Eif4e
265 BI283681 eukaryotic translation initiation factor 5A Eif5a
266 BM388758 excision repair cross-complementing rodent repair deficiency, complementation group 3 Ercc3
267 D13374 expressed in non-metastatic cells 1 Nme1
268 AI385371 extra spindle poles like 1 (S. cerevisiae) (predicted) Espll_predicited
269 NM_080895 Fas apoptotic inhibitory molecule Faim
270 AF044201 Fas apoptotic inhibitory molecule 2 Faim2
271 NM_080891 Fas deith domain-associated protein Daxx
272 NM_012908 Fas ligand (TNF superfamily, member 6) Faslg
273 AI227743 Fas-activated serine/threonine kinase Fastk
274 NM_130406 Fas-associated factor 1 Fafl
275 NM_053843 Fe receptor, IgG, low affinity III /// Fe gamma receptor II beta Fcgr3///LOC498276
276 AA999104 Feminization 1 homolog b (C. elegans) (predicred) Fem1_predicted
277 NM_019305 fibroblast growth factor 2 Fgf2
278 NM_130817 fibroblast growth factor 3 Fgf3
279 AB079673 fibroblast growth factor 4 Fgf4
280 NM_133286 fibroblast growth factor 8 Fgf8
281 S54008 Fibroblast growth receptor 1 Fgfrl
282 NM_053429 fibroblast growth factor receptor 3 Fgfr3
283 AA893484 fibronectin 1 Fnl
284 AI103600 Filamin C, gamma (actin binding protein 280) (predicted) Flnc_predicted
285 AF040256 folate hydrolase Folhl
286 M36804 follicle stimulating hormone beta Fshb
287 NM_012561 follistatin Fst
288 BI295511 forkhead box O1A Foxola
289 AI231684 forkhead bos O3a(predicted) Foxo3a_predicted
290 NM_012953 fos-like antigen 1 Fosl1
291 NM_012954 fos-like antigen 2 /// FBJ osteosarcoma oncogene B Fos12 /// Fosb
292 NM_017181 fumarylacetoacetate hydrolase Fah
293 BE108192 Gl to S phase transition 1 Gsptl
294 NM_033237 galanin Gal
295 NM_019172 galanin receptor 2 Galr2
296 NM_0533840 gamma-glutamyltransferase 1 Ggt1
297 NM_019281 gap junction membrane channel protein alpha 9 Gja9
298 NM_053388 gap junction membrane channel protein beta 6 Gja6
299 NM_012849 gastrin Gast
300 AA945758 gb:AA945758/DB_XREF=gi:3105674/DB_XREF=EST201257/CLONE=RLUAS87/FEA=EST/CNT-13/TID=Rn.7908.1/TIER=Stack/STK=8/UG=Rn.7908/UG_TITLE=ESTs NS
301 AI230220 gb:AI230220/DB_XREF=gi:3814107/DB_XREF=EST226915/CLONE=REMCT79/FEA=EST/CNT=9/TID=Rn.24381.1/TIER=Stack/STK=7/UG=Rn.24381/UG_TITLE=ESTs, Moderately similar to MLE3 RAT MYOSIN LIGHT CHAIN 3, SKELETAL MUSCLE ISOFORM (R.norvegicus) NS
302 BF555051 gb:BF555051/DB_XREF=gi:11664781/DB_XREF=UI-R-E0-cg-f-04-0-Ur1/CLONE=UI-R-E0-cg-f-04-0-UI/FEA=EST/CNT=3/TID=Rn.65517.1/TIER=ConsEnd/STK=1/UG=Rn.65517/UG_TITLE=ESTs, Weakly similar to VITAMIN K-DEPENDENT PROTEIN S PRECURSOR (R.norvegicus) NS
303 BM384229 gb:BM384229/DB_XREF=gi:18184282/DB_XREF=UI-R-DZO-cks-c-03-0-UI.s1/CLONE=UI-R-DZO-cks-c-03-0-UI/FEA=EST/CNT=12/TID=rn.14615.1/TIER=Stack/STK=11/UG=Rn.14615/UG_TITLE=ESTs, Highly similar to TRA2 MOUSE TNF RECEPTOR ASSOCIATED FACTOR 2 (M.musculus) NS
304 J02582 gb:J02582/DB_XREF=gi:202957/FEA=DNA_2/CNT=1/TID=Rn.64667.1/TIER=ConsEnd/STK=0/UG=Rn.64667/UG_TITLE=Rat apolipoprotein E gene, complete cds/DEF=Rat apolipoprotein E gene, complete cds NS
305 BI285576 gelsolin Gsn
306 NM_021669 ghrelin precursor Ghrl
307 NM_019139 glial cell line derived neurotrophic factor Gdnf
308 BF281741 glioma tumor suppressor candidate region gene 2 Gltscr2
309 NM_012728 glucagon-like peptide 1 receptor Glp1r
310 BI283882 glucose phosphate isomerase Gpi
311 NM_017006 glucose-6-phosphate dehydrogenase X-linked G6pdx
312 U08259 glutamate receptor, ionotropic, NMDA2C Grin2c
313 NM_017010 glutamate receptor, ionotropic, N-methyl D-aspartate 1 Grin1
314 AF001423 glutamate receptor, ionotropic, N-methyl D-aspartate 2A Grin 2a
315 M91562 glutamate receptor, ionotropic, N-methyl D-aspartate 2B Grin2b
316 NM_017011 glutamate receptor, metabotropic 1 Gria2bml
317 M92075 glutamate receptor, metabotropic 2 Grm2
318 AW522430 glutamate receptor, metabotropic 3 Grm3
319 NM_022202 glutamate receptor, metabotropic 8 Gria2bm8
320 J05181 glutamate-cysteine ligase, catalytic subunit Gclc
321 BG380882 glutaminyl-tRNA synthetase /// similar to glutaminyl-tRNA synthetase (predicted) Qars /// RGD1562301_predicted
322 NM_022278 glutaredoxin 1 (thioltransferase) Glrxl
323 S41066 glutathione peroxidase 1 Gpx1
324 NM_017165 glutathione peroxidase 4 Gpx4
325 NM_017013 glutathione-S-transferase, alpha type 2 Gsta2
326 X02904 glutathione-S-transferase, pi 1 /// glutathione S-transferase, pi 2 Gstp1 /// Gstp 2
327 NM_017008 glyceraldehyde-3-phosphate dehydrogenase /// similar to glyceraldehyde-3-phosphate dehydrogenase (predicted) /// similar to glyceraldehyde-3-phosphate dehydrogenase (predicted) /// similar to glyceraldehyde-3-phosphate dehydrogenase (predicted) /// similar to glyceraldehyde-3-phosphate dehydrogenase (predicted) Gapdh /// RGD1564688_predictated /// RGD1564351_predicted /// RGD1561683_predicted /// RGD1565368_predicted
328 BF287444 glycogen synthase kinase 3 beta Gsk3b
329 AI103970 glyoxylase 1 Glo1
330 BM391371 goliath LOC652955
331 NM_031038 gonadotropin releasing hormone receptor Gnrhr
332 NM_012767 gonadotropin-releasing hormone 1 Gnrh1
333 M34097 granzyme B Gzmb
334 U57063 granzyme G Gzmg
335 NM_019282 gremlin 1 homolog, cysteine knot superfamily (Xenopus laevis) Grem1
336 NM_024127 growth arrest and DNA-damage-inducible 45 alpha Gadd45a
337 B1287978 growth arrest and DNA-damage-inducible 45 beta Gadd45b
338 AI599423 growth arrest and DNA-damage-inducible 45 gamma Gadd45g
339 NM_057100 growth arrest specific 6 Gas6
340 X62853 Growth factor receptor bound protein 2 Grb2
341 V01238 growth hormone 1 Ghl
342 AI170771 growth hormone receptor Ghr
343 U94321 growth hormone secretagogue receptor Gshr
344 NM_024356 GTP cyclohydrolase 1 Gch
345 BM389208 GTPase, IMAP family member 4 Gimap4
346 M12672 guanine nucleotide binding protein, alpha inhibiting 2 Gnai2
347 BE117491 gnanine nucleotide binding protein, alpha q polypeptide Gnaq
348 BM390519 GULP, engulfment adaptor PTB domain containing 1 Gulp1
349 BG379941 Harvey rat sarcome viral (v-Ha-ras) oncogene homolog Hras
350 NM_012966 heat shock 10 kDa protein 1 (chaperonin 10) Hspel
351 AI236601 heat shock 105kDa/110kDa protein 1 H2phl
352 NM_031970 heat shock 27kDa protein 1 Hspb1
353 NM_031971 heat shock 70kD protein 1A /// heat shock 70kD protein IB (mapped) Hspala /// Hspalb_mapped
354 BI278231 heat shock 70kD protein IB (mapped) Hspalb_mapped
355 M14050 heat shock 70kDa protein 5 (glucose-regulated protein) Hspa5
356 BI282281 heat shock 70kDa protein 9A (predicted) Hspa9a_predicted
357 AI237389 heat shock 90kDa protein 1, beta Hspcb
358 NM_022229 heat shock protein 1 (chaperonin) Hspd1
359 BG671521 heat shock protein 1, alpha Hspca
360 AF077354 heat shock protein 4 Hspa4
361 NM_012580 heme oxygenase (decyclinj) 1 Hmox1
362 NM_013185 hemopoietic cell kinase Hck
363 1387701_at hepatocyte growth factor Hgf
364 NM_012734 hexokinase 1 Hk1
365 NM_012735 hexokinase 2 Hk2
366 BG378885 high mobility group AT-hook 1 Hmga1
367 BE107162 high mobility group box 1 Hmgb1
368 AF275734 high mobility group box 1 /// similar to High mobility group protein 1 (HMG-1) (predicted) /// similar to Hmgb1 protein (predicted) /// similar to High mobility group protein 1 (HMG-1) (predicted) Hmgbl /// RGD1562312_predicted /// RGD1563786_predicted /// RGD1563012_predicted
369 AI180339 histone deacetylase 1 (predicted) Hdacl_predicted
370 NM_053448 histone deacetylase 3 Hdae3
371 BF403027 histone deacetylase 5 Hdac5
372 NM_053609 HLA-B-associated transcript 3 Bat3
373 M37568 homeo box C8 (mapped) Hoxc8_mapped
374 BM392321 homeodomain interacting protein kinase 2 (predicted) Hipk2_predicted
375 NM_031787 homeodomain interacting protein kinase 3 Hipk3
376 AB003726 homer homolog 1 (Drosophila) Homer1
377 NM_053523 homocysteine-inducible, endoplasmic reticulum stress-inducible, ubiquitin-like domain member 1 Herpud1
378 BE111733 hormone-regulated proliferation associated protein 20 Hrpap20
379 AW253339 huntingtin interacting protein 1 Hipl
380 U18650 Huntington disease gene homolog Hdh
381 NM_019371 hypothetical gene supported by NM_019371 LOC497816
382 BM390522 hypothetical gene supported by NM_130426 LOC497808
383 H31665 hypoxia induced gene 1 Hig1
384 NM_024359 hypoxia inducible factor 1, alpha subunit Hifla
385 BI282904 hypoxia up-regulated 1 Hyou1
386 AI176519 immediate early response 3 Ier3
387 AI411947 immunoglobulin heavy chain 1a (serum IgG2a) Igh-1a
388 NM_023973 indoleamine-pyrrole 2,3 dioxygenase Indo
389 NM_012590 inhibin alpha Inha
390 NM_017128 inhibin beta-A Inhba
391 NM_013060 inhibitor of DNA binding 2 Id2
392 AF000942 inhibitor of DNA binding 3 Id3
393 NM_053355 inhibitor of kappaB kionase beta Ikbkb
394 J05510 inositol 1,4,5-triphosphate receptor 1 Itpr1
395 NM_019311 inositol polyphosphate-5-phosphatase D Inppd5
396 NM_019129 insulin 1 Igf2bp1
397 NM_032074 insulin receptor substrate 3 Irs3
398 M15481 insulin-like growth factor 1 Igfl
399 NM_052807 insulin-like growth factor 1 receptor Igflr
400 NM_031511 insulin-like growth factor 2 Igf2
401 NM_012588 insulin-like growth factor binding protein 3 Igfbp3
402 BF282337 integral membrane protein 2B Itm2b
403 NM_017022 integrin beta 1 (fibronectin receptor beta) Itgbl
404 NM_133409 integrin linked kinase Ilk
405 NM_019127 interferon beta 1, fibroblast Ifnblf
406 AF010466 interfereon gamma Ifng
407 NM_012591 interferon regulatory factor 1 Irf1
408 NM_017019 interleukin 1 alpha Il1a
409 NM_031512 interleukin 1 beta Il1b
410 L02926 interleukin 10 Il10
411 AF347836 interleukin 11 receptor, alpha chain 1 Il11ra1
412 NM_053828 interleukin 13 I113
413 AF015718 interleukin 15 I115
414 AJ222813 interleukin 18 I118
415 NM_013163 interleukin 2 receptor, alpha chain I12ra
416 NM_013195 interleukin 2 receptor, beta chain I12rb
417 NM_031513 interleukin 3 I16
418 X16058 interleukin 4 I14
419 NM_012589 interleukin 6 I16
420 AF367210 interleukin 7 I17
421 BF405951 Interleukin-1 receptor-associated kinase 4 (predicted) Irak4_predicted
422 1388184_at isoprenylcysteine carboxyl methyltransferase Icmt
423 NM_031514 Janus kinase 2 Jak2
424 BE096021 Jun D proto-oncogene Jundl
425 BI288619 Jun oncogene Jun
426 NM_021836 Jun-B oncogene Junb
427 NM_012696 kininogen 1 /// K-kininogen /// similar to alpha-1 major acute phase protein prepeptide Kng1 /// LOC215087 /// MGC108747
428 NM_031135 Kruppel-like factor 10 Klf10
429 BM385790 Kruppel-like factor 2 (lung) (predicted) Klf2_predicted
430 NM_053394 Kruppel-like factor 5 Klf5
431 NM_053902 kynureninase (L-kynurenine hydrolase) Kynu
432 NM_012594 Lactalbiuain, alpha Lalba
433 NM_019904 lectin, galactose binding, soluble 1 Lgals1
434 NM_031832 lectin, galactose binding, soluble 3 Lgals3
435 NM_022582 lectin, galactose binding, soluble 7 Lgals7
436 NM_031048 leukemia inhibitory factor receptor Lifr
437 NM_031727 LIM motif-containing protein kinase 1 Limk1
438 NM_130741 lipocalin 2 Lcn2
439 BF289368 lipopolysaccharide binding protein Lbp
440 BI284739 LPS-induced TN factor Litaf
441 AA874924 lymphocyte antigen 86 (predicted) Lv86_predicted
442 AI137137 lymphocyte protein tyrosine kinase (mapped) Lck_mapped
443 AI012109 lymphocyte specific 1 Lsp1
444 NM_080769 lymphotoxin A Lta
445 NM_053538 Iysosomal-associated protein transmembrane 5 Laptm5
446 NM_031051 macrophage migration inhibitory factor Mif
447 NM_024352 Macrophage stimulating 1 (hepatocyte growth factor-like) Mst1
448 NM_019191 MAD homolog 2 (Drosophila) Smad2
449 AA997679 MAD homolog 3 (Drosophila) Smad3
450 NM_019275 MAD homolog 4 (Drosophila) Smad4
451 AW521447 MAD homolog 7 (Drosophila) Madh7
452 NM_053585 MAP-kinase acivating death domain Madd
453 U65656 matrix metallopeptidase 2 Mmp2
454 NM_031055 matrix metallopeptidase 9 Mmp9
455 BI289109 max binding protein (predicted) Mnt_predicted
456 AW143154 megakaryoblastic leukemia (translocation) 1 (predicted) MKl1_predicted
457 NM_053409 melanoma antigen, family D, 1 Magedl
458 AF411318 metallothionein 1a Mtla
459 NM_053307 methionine sulfoxide reductase A Msrb2
460 BI281702 microtubule-associated protein 1b Map1b
461 BE107978 microtubule-associated protein tau /// hypothetical gene supported by NM_017212 Mapt /// LOC497674
462 BG685132 mitochondrial carrier homolog 1 (C. elegans) Mtchl
463 AA943734 mitochondrial protein. 18 kDa MGC94604
464 BG378230 mitochondrial ribosomal protein S30 (predicted) Mrps30_predicted
465 NM_053842 mitogen activated protein kinase 1 Mapk1
466 NM_012806 mitogen activated protein kinase 10 Mapk10
467 AW254190 mitogen activated protein kinase 14 Mapk14
468 AF155236 mitogen activated protein kinase 3 Mapk3
469 NM_053777 mitogen activated protein kinase 8 interacting protein Mappk8ip
470 D13341 mitogen activated protein kinase 1 Map2k1
471 D14592 mitogen activated protein kinase kinase 2 Map2k2
472 NM_053887 mitogen activated protein kinase kinase kinase 1 Map3k1
473 NM_013055 mitogen activated protein kinase kinase kinase 12 Map3k12
474 AI146037 Mitogen activated protein kinase kinase kinase 7 (predicted) Map3k7_predicted
475 AI575972 Mitogen-activaced protein kinase 8 interacting protein 2 Mapk8ip2
476 NM_017322 mitogen-activated protein kinase 9 Mapk9
477 BI281589 mitogen-activated protein kinase kinase kinase 11 Map3k11
478 NM_053847 mitogen-activated protein kinase kinase kinase 8 Map3k8
479 D00688 monoamine oxidase A Maoa
480 NM_012982 msh homeo box homolog 2 (Drosophila) Msx2
481 NM_053337 Msx-interacting-zinc finger Mizl
482 BI274326 mucin 1, transmembrane Muc1
483 BM391100 mucin 4 Muc4
484 NM_031053 mutL homolog 1 (E. coli) /// hypothetical gene supported by NM_031053 Mlh1 /// LOC407834
485 NM_021837 myc-like oncogene, s-myc protein Myes
486 NM_012798 myelin and lymphocyte protein. T-cell differentiation protein Ma1
487 BF281184 myeloblastosis oncogene-like 2 (predicted) Mybl2_predicted
488 NM_012603 myelocytomatosis viral oncogene homolog (avian) Myc
489 AI172056 myeloid cell leukemia sequence 1 Mcl1
490 BI284349 myeloid differentiation primary response gene 116 Mydl16
491 AI238590 myeloid differentiation primary response gene 88 Myd88
492 NM_030860 myocyte enhancer factor 2D Mef2d
493 J02679 NAD(P)H dehydrogenase, quinone 1 Nqo1
494 NM_053683 NADPH oxidase 1 Noxal
495 AI178285 NCK-associated protein 1 Nckap1
496 NM_031069 NEL-like 1 (chicken) Nell1
497 NM_012610 nerve growth factor receptor (TNFR superfamily, member 16) Ngfr
498 NM_053401 nerve growth factor receptor (TNFRSF16) associated protein 1 Ngfrap1
499 BM388972 nerve growth factor, beta (mapped) Norb
500 U02323 neuregulin 1 Nrg1
501 NM_023868 neurotrophic Y receptor Y2 Npy2r
502 NM_021589 neurotrophic tryosine kinase, receptor, type 1 Ntrk1
503 NM_031073 neutrophin 3 Ntf3
504 AI598730 neurotrophin receptor a associated death domain Nradd
505 NM_053734 neutrophil cytosolic factor 1 Ncf1
506 BI285459 nicastrin Ncstn
507 L12562 nitric oxide synthase 2, inducible Nos2
508 AJ011116 nitric oxide synthase 3, endothelial cell Nos3
509 NM_053507 non-metastatic cell expressed protein 3 Nme3
510 BF389398 Notch gene homolog 1 (Drosophila) Notchl
511 AI011448 Notch gene homolog 2 (Drosophila) Notch2
512 NM_020087 Notch gene homolog 3 (Drosophila) Notch3
513 BG377358 nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 4 Nfatc4
514 AA858801 nuclear factor of kappa light chain gene enhancer in B-cells 1, p105 Nfkb1
515 AW672589 nuclear factor of kappa light chain gene enhancer in B-cells inhibitor, alpha Nfkbia
516 NM_030867 nuclear factor of kappa light chain gene enhancer in B-cells inhibitor, beta Nfkbib
517 NM_012991 nuclear pore associated protein Npap60
518 NM_021745 nuclear receptor subfamily 1, group H, member 4 Nrlh4
519 NM_05298O nuclear receptor subfamily 1, group I, member 2 Nrcli2
520 NM_017323 nuclear receptor subfamily 2, group C, member 2 Nrc2c2
521 AY066016 nuclear receptor subfamily 3, group C, member 1 Nr3c 1
522 NM_013131 nuclear receptor subfamily 3, group C, member 2 Nr3c2
523 NM_024388 nuclear receptor subfamily 4, group A, member 1 Nr4a1
524 NM_031628 nuclear receptor subfamily 4, group A, member 3 Nr4a3
525 NM_022799 nuclear ubiquitous casein kinase and cyclin-dependent kinase substrate Nucks
526 NM_053516 nucleolar protein 3 (apoptosis repressor with CARD domain) Nol3
527 NM_012992 nucleophosmin 1 Npml
528 J04943 nucleophosmin 1 /// similar to Nucleophosmin (NPM) (Nucleolar phosphoprotein B23) (Numatrin) (Nucleolar protein NO38) Npml /// LOC300303
529 BI286040 nucleoporin 62 Nup62
530 NM_133525 Nucleoside 2-deoxynbosyltransferase domain containing protein RGD620382 RGD620382
531 NM_012861 O-6-methylgnanine-DNA methyltransferase Mgmt
532 L20684 opioid receptor, mu 1 Oprl1
533 NM_133585 optic atrophy 1 homolog (human) Opal
534 NM_053288 orosomucoid 1 Orml
535 NM_130402 osteoclast inhibitory lectin Ocil
536 NM_133306 oxidized low density lipoprotein (lectin-like) receptor 1 Oldlr1
537 NM_019210 p21 (CDKN1A)-activated kinase 3 Pak3
538 NM_053289 pancreatitis-associated protein Pap
539 NM_017044 parathyroid hormone Pth
540 BI281756 Parkinson disease (autosomal recessive, early onset) 7 Park7
541 BG673589 paxillin Pxn
542 AI009656 PEF protein with a long N-terminal hydrophobic domain Peflin
543 BI291292 peptidylprolyl isomerase C Ppic
544 AA957342 peptidylprolyl isomerase D (cyclophilin D) Ppid
545 U68544 peptidylprolyl isoinerase F (cyclophilin F) Ppif
546 NM_017330 perforin 1 (pore forming protein) Prfl
547 NM_017169 peroxiredoxin 2 Prdx2
548 NM_013196 peroxisome proliferator activated receptor alpha Ppara
549 U75918 Peroxisome proliferator activated receptor delta Ppard
550 NM_013124 peroxisome proliferator activated receptor gamma Pparg
551 AI598971 PERP, TP53 apoptosis effector (predicted) Perp_predicted
552 NM_021657 PH domain and leucine rich repeat protein phosphatase Phlpp
553 NM_031606 phosphatase and tensin homolog Pten
554 NM_053823 phosphatidylinositol 3-kinase, C2 domain containing, gamma polypeptide Pik3ca
555 BI290699 phosphatidylinositol 3-kinase, catalytic, alpha polypeptide Pik3ca
556 D64048 phosphatidylinositol 3-kinase, regulatory subunit, polypeptide 1 Pik3rl
557 NM_022185 phosphatidylinositol 3-kinase, regulatory subunit, polypeptide 2 Pik3r2
558 AI232697 phosphatidyhenrie receptor Ptdsr
559 AI454840 Photphodiesterase 1A, calmodulin-dependent Pdela
560 AF327906 phosphodiesterase 1B, Ca2+calmodulin dependant Pdelb
561 NM_022958 phosphoinositide-3-kinase class 3 Pik3c3
562 NM_133551 phospholipase A2, group IVA (cytosolic, calcium-dependent) Pla2g4a
563 U51898 phopholipase A2, group VI Pla2g6
564 U69550 phospholipase D1 Pldl
565 BE112895 phosphoprotein enriched in astrocytes 15 Peal5
566 NM_053491 plasminogen Plg
567 NM_013151 plasminogen activator, tissue Plat
568 NM_013085 plasminogen activator, urokinase Plau
569 AF007789 plasminogen activator, urokinase receptor Plaur
570 BE100812 platelet derived growth factor, alpha Pdgfa
571 BM392366 platelet-activating factor acetylhydrolase, isoform 1b, alpha2 subunit Pafahlb2
572 AI009219 Pleckstrin homology domain containing, family A member 5 PlekhaS
573 NM_017180 pleckstrin homology-like domain, family A, member 1 Phlda1
574 NM_012760 pleiomorphic adenoma gene-like 1 Pkgl1
575 AB019366 poly (ADP-ribose) glycohydrolase Parp1
576 NM_013063 poly (ADP-ribose) polymerase family, member 1 Parp1
577 NM_017141 polymerase (DNA directed), beta Polb
578 AW531224 polymerase (RNA) II (DNA directed) polypeptide A (mapped) Polr2a_mapped
579 AW435212 potassium channel, subfamily K, member 3 Kcnk3
580 NM_053405 potassium channel, subfamily K, member 9 Kcnk9
581 NM_013186 potassium voltage gated channel, Shab-related subfamily, member 1 Kcnb1
582 BM385544 presenilin 1 Psenl
583 AB004454 presenilin 2 Psen2
584 AI232272 presenilin enhancer 2 homolog (C. elegans) Psenen
585 BI278802 prion protein Prnp
586 U05989 PRKC, apoptosis WT1, regulator Pawr
587 BI285575 procollagen, type 1, alpha 1 Colla1
588 BE108058 procollagen, type XVIII alpha 1 Coll8a1
589 AI599419 Progesterone receptor Pgr
590 AI704628 programmed cell death 2 Pdcd2
591 NM_022265 programmed cell death 4 Pdcd4
592 BF408447 programmed cell death 5 (predicted) Pdcd5_predicted
593 BI296393 programmed cell death 6 (predicted) Pdcd6_predicted
594 BE328942 programmed cell death 6 interacting protein Pdcd6ip
595 AF262320 programmed cell death 8 Pdcd8
596 AI013847 programmed cell death protein 7 (predicted) Pdcd7_predicted
597 BI282863 prohibition Phb
598 NM_012629 prolactin Prl
599 L48060 prolactin receptor Prlr
600 BI290159 proline-serine-threonine phosphatase-interacting protein 1 (predicted) Pstpip1_predicted
601 NM_138857 prominin 2 Prom2
602 NM_031644 prostaglandin D2 synthase 2 Ptgds2
603 U03389 prostaglandin-endoperoxide synthase 2 Ptgs2
604 A1600136 protease, serine, 25 Prss25
605 NM_031978 proteasome (prosome, macropain) 26S subunit, non-ATPase, 1 Psmdl
606 NM_130430 proteasome (prosome, macropain) 26S subunit nou-ATPase, 9 Psmd9
607 NM_012803 protein C Prc
608 AI639478 protein disulfide isomerase associated 2 (predicted) Pdia2_predicted
609 NM_017319 protein disulfide isomerase associated 3 Pdia3
610 BF415343 protein kinase C, alpha Prkca
611 X04440 protein kinase C, beta 1 Prtcb1
612 NM_133307 protein kinase C, delta Prkcd
613 AA799421 protein kinase C, epsilon Prkce
614 1370197_a_at protein kinase C, zeta Prkcz
615 NM_019142 protein kinase, AMP-activated, alpha 1 catalytic subunit Prkaa1
616 NM_023991 protein kinase, AMP-activated, alpha 2 catalylic subunit Prkaa2
617 NM_013012 protein kinase, cGMP-dependent type II Prkg2
618 BF400782 protein kinase, DNA activated, catalytic polypeptide (predicted) Prkdc_predicted
619 NM_019335 Protein kinase, interferon-inducible double stranded RNA dependent Prkr
620 NM_031527 protein phosphatase 1, catalytic subunit, alpha isoform Ppp1ca
621 NM_013065 protein phosphatase 1, catalytic subunit, beta isoform Ppp1cb
622 NM_022676 protein phosphatase 1, regulatory (inhibitor) subunit 1 A Ppp1rA
623 AI172276 protein phosphatase 1, regulatory (inhibitor) subunit 2 Ppp1r2
624 AB023634 protein phosphatase 1F (PP2C domain containing) Ppmlf
625 BF408792 protein phosphatase 2 (formerly 2A), catalytic subunit, alpha isoform Ppp2ca
626 NM_017040 protein phosphatase 2 (formerly 2A), catalytic subunit beta isoform Ppp2cb
627 AA800669 protein phosphatase 2 (formerly 2A), regulatory subunit A (PR 65), alpha isoform Ppp2r1a
628 1373959_at protein phosphatase 2 (formerly 2A), regulatory subunit A (PR 65), beta isoform Ppp2r1b
629 AI717081 protein phosphatase 2 (formerly 2A), regulatory subunit B (PR 52), alpha inform Ppp2r2a
630 BE113127 Protein phosphatase 3, catalytic subunit, alpha isoform Ppp3ca
631 NM_031729 protein phosphatase 5, catalytic subunit Ppp5c
632 U06230 protein S (alpha) Pros1
633 U69109 protein tyrosine kinase 2 beta Ptk2b
634 NM_012637 protein tyrosine phosphatase, non-receptor type 1 Ptpn1
635 AI172465 Protein tyrosine phosphatase, non-receptor type 11 Ptpn11
636 NM_053908 protein tyrosine phosphatase, non-receptor type 6 Ptpn6
637 M10072 protein tyrosine phosphatase, receptor type, C Ptprc
638 NM_022925 protein tyrosine phosphatase, receptor type, Q /// hypothetical gene supported by NM_022925; NM_198323 Ptprq
639 AI178772 prothymosin alpha Ptma
640 NM_017034 proviral integration site 1 Piml
641 BI294798 PTK2 protein tyrosine kinase 2 Ptk2
642 AF231010 purinergic receptor P2X, ligand-gated ion channel, 1 P2rx1
643 AF020757 purinergic receptor P2X, ligand-gated ion channel, 2 P2rx2
644 NM_019256 purinergic receptor P2X, ligand-gated ion channel, 7 P2ry2
645 NM_017255 purinergic receptor P2Y, G-protein coupled 2 P2ry2
646 BI282953 PYD and CARD domain containing Pycard
647 NM_013018 RAB3A, member RAS oncogene family Rab3a
648 U70777 rabaptin, RAB GTPase binding effector protein 1 Rabep1
649 AJ249986 Rap guanine nucleotide exchange factor (GEF) 1 Rapgef1
650 AF002251 Ras association (RalGDS/AF-6) domain family 5 Rassfs
651 AF081196 RAS guauyl releasing protein 1 Rasgrp2
652 AI408053 ras homolog gene family, member A Rhoa
653 NM_022542 ras homolog gene family, member B Rhob
654 NM_013135 RAS p21 protein activator 1 Rasa1
655 BF414025 Ras-induced senescence 1 Ris1
656 AA799542 Ras-related C3 botulinum toxin substrate 1 Rac1
657 AF036537 receptor-interacting serine-threonine kinase 3 Ripk3
658 NM_012641 regenerating islet-derived 1 Reg1
659 L20869 regenerating islet-derived 3 gamma Reg3g
660 NM_031546 regucalcin Rgn
661 AJ299017 ret proto-oncogene Ret
662 AF051335 reticulon 4 Rtn4
663 AI178012 retinoblastoma 1 Rb1
664 NM_031094 retinoblastoma-like 2 Rb12
665 NM_031528 retinoic acid receptor, alpha Rara
666 BF419646 retinoic acid receptor, beta Rarb
667 BI285959 retinoid X receptor alpha Rxra
668 AI408677 Rho GDP dissociation inhibitor (GDI) alpha Arhgdia
669 NM_031098 Rho-associated coiled-coil forming kinase 1 Rock1
670 NM_013022 Rho-associated coiled-coil forming kinase 2 Rock2
671 NM_022510 ribosomal protein L4 Rp14
672 BI282255 ribosomal protein S5 Rps5
673 M57428 ribosomal protein S6 kinase, polypeptide 1 Rps6kb1
674 AI179991 ring finger protein 34 Rnf34
675 AA858518 ring finger protein 7 (predicted) Rnf7_predicted
676 AI175966 Rons sarcoma oncogene Src
677 BF403180 Runt related transcription factor 2 Runx2
678 NM_012618 S100 calcium-binding protein A4 S100a4
679 NM_013191 S100 protein, beta polypeptide S100b
680 AA850867 sarcoglycan, gamma (dystrophin-associated glycoprotein) Sgcg
681 NM_031541 scavenger receptor class B, member 1 Scarb1
682 BI294932 SCF apoptosis response protein 1 LOC499941
683 NM_053587 schlafen 3 Slfn3
684 BF394953 SDA1 domain containing 1 Sdad1
685 NM_019364 sec1 family domain containing 1 Scfdl
686 AF220608 secreted frizzled-related protein 4 Sfrp4
687 AB001382 secreted phosphoprotein 1 Sppl
688 NM_017310 sema domain, immnuoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3A Sema3a
689 BI299759 sema domain, transmembrane domain (TM), and cytoplasmic domain, (semaphorin) 6A (predicted) Sema6a_predicted
690 NM_133291 seminal vehicle antigen-like 2 Sva12
691 M25590 seminal vesicle protein 4 Svp4
692 BG663993 sequestosome 1 Sqstm1
693 NM_012620 serine (or cysteine) peptidase inhibitor, clade E, member 1 Serpine1
694 NM_021696 serine (or cystsine) proteinase inhibitor, clade B, member 2 Se2pinb2
695 AA944455 serine incorporator 3 Serine3
696 NM_133392 serine/threonine kinase 17b (apoptosis-inducing) Stk17b
697 NM_019349 serine/threonine kinase 2 Stk2
698 NM_031735 serine/threonine kinase 3 (STE20 homolog, yeast) Stk3
699 AF388527 Seipine1 mRNA binding protein 1 Serbp1
700 NM_017170 serum amyloid P-component Apcs
701 NM_019232 serum/glucocorticoid regulated kinase Sgk
702 NM_080905 seven in absentia 1A Siah1a
703 NM_134457 seven in absentia 2 Siah2
704 NM_012650 sex hormone binding globulin Shbg
705 BF284481 SH3-domain GRB2-like B1 (endophilin) Sh3glbl
706 AF255888 SH3-domain kinase binding protein 1 Sh3kbp1
707 BF550890 sialophorin Spn
708 NM_032612 signal transducer and activator of transcription 1 Stat1
709 B1285863 signal transducer and activator of transcription 3 Stat3
710 NM_017064 signal transducer and activator of transcription 5A Stat5a
711 AI177626 signal transducer and activator of transcription 5B Stat5b
712 BE110607 similar to apoptosis related protein APR-32; p18 protein (predicted) RGD1311805_predicted
713 BF396386 similar to cell division cycle and apoptosis regulator 1 (predicted) RGD1560358_predicted
714 BI296385 similar to chemokine (C-X-C motif) ligand 16 Cxcl16
715 BI294745 similar to livin inhibitor of apoptosis isoform beta (predicted) RGD1562883_predicted
716 NM_021846 similar to MAP/microtubule affinity-regulating kinase 4 (MAP/microtubule affinity-regulating kinase like 1) (predicted) RGD1561096_predicted
717 AA848545 similar co programmed cell death 10 MGC72992
718 BF282636 similar to RIKEN cDNA 1700023M03 RGD1305457
719 AW254416 Similar to TGF-beta induced apoptosis protein 12 (predicted) RGD130816_predicted
720 AA946199 snail homolog 1 (Drosophila) Snail
721 X89383 SNF related kinase Snrk
722 NM_012647 sodium channel, voltage-gated, type 2, alpha 1 polypeptide Scn2a
723 NM_013178 sodium channel, voltage-gated, type IV, alpha polypeptide Snc4a
724 BI284218 solute carrier family 2(facilitated glucose transporter), member 1 Slc2a1
725 NM_017102 solute carrier family 2(facilitated glucose transporter), member 3 Slc2a3
726 NM_017223 solute carrier family 20, member 2 Slc20a2
727 BG666999 solute carrier family 25 (mitochondrial carrier; adenine nucleotide translocator), member 4 Slc25a4
728 NM_017206 solute carrier family 6 (neurotransmitter transporter, taurine), member 6 Slc6a6
729 NM_053442 solute carrier family 7 (cationic amino acid transporter, y+ system), member 8 /// synaptic Ras GTPase activating protein 1 homolog (rat) slc7a8 /// Syngap1
730 U04933 solute carrier family 8 (sodium/calcium exchanger), member 1 Slc7a8 /// Syngap1
731 NM_133522 somatostatin receptcor 3 /// hypothetical gene supported by NM_133522 SSTR
732 BI275248 Son cell proliferation protein Son
733 NM_017221 sonic hedgehog homolog (Drosophila) Shh
734 NM_012655 Sp1 transcription factor Sp1
735 AB049572 sphingosine kinase 1 Sphk1
736 BM386306 sphingosine kinase 2 Sphk2
737 AI159638 sphingosiue-1-phosphate phosphatase 1 Sgpp1
738 U53883 ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 1 St8sia2
739 AY083159 Stam binding protein Stambp
740 BM386683 stanniocalcin 1 Stc1
741 NM_022230 stanniocalcin 2 Stc2
742 NM_017166 stathmin 1 Stmn1
743 AF335281 STEAP family member 3 Steap3
744 AI235465 steroid sensitive gene 1 Ssg1
745 AI009817 succinate dehydrogenase complex, subunit C, integral membrane protein Sdhc
746 NM_134378 sulfatase 1 Sdhc
747 NM_017050 superoxide dismutase 1 Sod1
748 BG671549 superoxide dismutase 2, mitochondrial Sod2
749 NM_058208 suppressor of cytokine signaling 2 Socs2
750 NM_053555 suppressor of cytokine signaling 3 Socs3
751 BM390864 survival motor neuron domain containing 1 Smndc1
752 AI170385 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 2 Smarca2
753 BE329013 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 Smarca4
754 NM_053442 synaptic Ras GTPase activating protein 1 homolog (rat) Syngap1
755 NM_013026 syndecan 1 Sdc1
756 AA946430 synovial apoptosis inhibitor 1, synoviolin Syvn1
757 NM_019169 synuclein, alpha Snca
758 BG671061 tachykinin 1 Tac1
759 BM392226 TAF10 RNA polymerase II, TATA box binding protein (TBP)-associated factor (predicted) Taf10_predicted
760 NM_133615 TAF9-like RNA polymerase II, TATA box binding protein (TBP)-associated factor, 31kDa Taf91
761 AI228250 Tax 1 (human T-cell leukemia virus I) binding protein 1 Taxlbp1
762 AA957545 T-box 3 Tbx3
763 AB029495 TCF3 (E2A) fusion partner Tfpt
764 AF247818 telomerase reverse transcriptase Tert
765 NM_019381 testis enhanced gene transcript Tegt
766 NM_133396 testis-specific kinase 2 Tesk2
767 AA943723 THAP domain containing, apoptosis associated protein 3 (predicted) Thap3_piedicoed
768 NM_053800 thioredoxin 1 Txn1
769 AA800180 thioredoxin 2 Txn2
770 BM390196 thioredoxin domain containing 5 (predicted) Txndc5_predicted
771 BF555110 thioredoxin-like 1 Txnl1
772 BM384228 THO complex 1 Thoc1
773 AA998057 thymoma viral proto-oncogene 1 Akt1
774 AI105076 Thymoma viral proto-oncogene 2 Akt2
775 NM_031575 thymoma viral proco-oncogene 3 Akt3
776 AI145313 thymus cell antigen 1, theta Thy1
777 NM_012888 thyroid stimulating hormone receptor Tshr
778 AI101391 Tial1 cytotoxic granule-associated RNA binding protein-like 1 (mapped) Tial1
779 NM_053819 tissue inhibitor of metalloproteinase 1 Timp1
780 NM_012989 tissue inhibitor of metalloproteinase 2 Timp2
781 NM_012886 Tissue inhibitor of metalloproteinase 3 (Sorsby fundus dystrophy, pseudoinflammatory) Timp3
782 AI104533 titin Ttn
783 BI287742 TM2 domain containing 1 (predicted) Tm2dl_predicted
784 BM389034 TNF receptor-associated protein 1 Trap1
785 BM386846 TNFRSF1A-associated via death domain Tradd
786 AF057025 toll-like receptor 4 Tlr4
787 AI012419 Transcribed locus NS
788 AW533194 Transcribed locus NS
789 AI104523 Transcribed locus NS
790 AW251860 transcription factor 7, T-cell specific (predicted) Tcf7_predicted
791 NM_031326 transcription factor A, mitochondrial Tfam
792 BI284455 transcription factor Pur-beta pur-beta
793 NM_133317 transducer of ErbB-2.1 Tob1
794 M58040 transferrin receptor Tfrc
795 BI297236 Transformation related protein 53 inducible nuclear protein 1 Trp53inp2
796 AJ277449 transformation related protein 63 Trp63
797 NM_012671 transforming growth factor alpha Tgfa
798 AW254561 transforming growth factor beta regulated gene 4 Tbrg4
799 1370082_at transforming growth factor, beta 1 TgSb1
800 BF420705 transforming growth factor, beta 2 Tgfb2
801 NM_012775 transforming growth factor, beta receptor 1 Tgfbr1
802 BI275994 transglutaminase 2, C polypeptide Tgm2
803 AB015231 transient receptor potential cation channel, subfamily V, member 1 Trpv1
804 NM_023970 transient receptor potenial cation channel, subfamily V, member 4 /// transient receptor potential cation channel, subfamily V, member 1 Trpv4
805 AB020967 tribbles homolog 3 (Drosophila) Trib3
806 NM_080903 tripartite motif protein 63 Trim63
807 AI104913 tropomodulin 1 Tmod1
808 NM_031345 TSC22 domain family 3 Tsc22d3
809 BI285434 tubulin, alpha 1 /// tubulin, alpha 6 /// similar to Tubulin alpha-2-chain (Alpha-tubulin 2) (predicted) Tuba1 /// Tuba6 /// RGD1565476_predicted
810 BI285434 tubulin, gamma 1 Tubg1
811 AA819227 tumor necrosis factor (TNF superfamily, member 2) Tnf
812 BF283688 tumor necrosis factor ligand superfamily member 12 Tnfsf12
813 NM_012870 tumor necrosis factor receptor superfamily, member 11b (osteoprotegerin) Tfrsf11b
814 BI303379 tumor necrosis factor receptor superfamily, member 12a Tnfrsfl2a
815 AI169601 tumor necrosis factor receptor superfamily, member 14 (herpesvirus entry mediator) Tnfrsf14
816 NM_013091 tumor necrosis factor receptor superfamily, member 1a Tnfrsf1a
817 NM_013049 tumor necrosis factor receptor superfamily, member 4 Tnfrsf4
818 AW433947 tumor necrosis factor receptor superfamily, member 5 Tofrsf5
819 NM_139194 Tumor necrosis factor receptor superfamily, member 6 Tofrsf6
820 BM387084 tumor necrosis factor superfamily, member 5-induced protein 1 (predicted) Tnfsf5ip1_predicted
821 AY009504 tumor protein p53 Tp53
822 NM_053867 tumor protein, translationally-controlled 1 Tpt1
823 BG057543 tumor rejection antigen gp96 (predicted) Tral_predicted
824 AI234654 tumor susceptibility gene 101 Tsg101
825 NM_013052 tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, eta polypeptide Ywhah
826 NM_019378 tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, gamma polypeptide Ywhag
827 BF281342 tyrosine 3-monooxygenase/trytohpan 5-monooxygenase activation protein, theta polypeptide Ywhaq
828 AI228292 tyrosine hydroxylase Th
829 AI407490 tyrosyl-tRNA synthetase Yars
830 NM_053747 Ubiquilin 1 Ubqln1
831 B1276086 Ubiquitin specific protease 7 (herpes virus-associated) Usp7
832 BI296848 ubiquitiuation factor E4B, UFD2 homolog (S. cerevisiae) (predicted) Ube4b_predicted
833 AI102437 ubiquitin-like 1 (sentrin) activating enzyme E1B Uble1b
834 AF159706 unc-13 homolog B (C. elegans) Unc 13b
835 NM_022206 unc-5 homolog A (C. elegans) Uuc5a
836 NM_022207 unc-5 homolog B (C. elegans) Unc5b
837 BM388453 uncharacterized protein family UPF0227 member RGD1359682 RGD1359882
838 U30789 upregulated by 1,25-dihydroxyvitamin D-3 Txnip
839 NM_053864 valosin-containing protein Vcp
840 AF080594 vascular endothelial growth factor A Vegfa
841 NM_012759 vav 1 oncogene Vav1
842 NM_021687 v-erb-a erythroblastic leukemia viral oncogene homolog 4 (avian) Erbb4
843 NM_017003 v-erb-b2 erythro leukemia viral oncogene homolog 2, neuro/glioblastoma derived, oncogene homolog (avian) Erbb2
844 NM_012555 v-ets ervthroblastosis virus E26 oncogene homolog 1 (avian) Ets1
845 NM_031140 vimentin Vim
846 AF268467 voltage-dependent anion channel 1 Vdac1
847 NM_012639 v-raf-1 murine leukemia viral oncogene homolog 1 Raf1
848 BF283772 v-rel reticuloendotheliosis viral oucogene homolog A (avian) Rela
849 NM_022548 wild-type p53-induced gene 1 Wig1
850 NM_031534 Wilms tumor 1 Wt1
851 NM_031590 WNT1 inducible signaling pathway protein 2 Wisp2
852 BI300732 WW domain-containing oxidoreductase (predicted) RGD1565791_predicted
853 BI182111 Y box protein 1 related, pseudogene 3 /// similar to nuclease sensitive element binding protein 1 (predicted) /// Y box protein 1 YbKl-ps3 /// RGD1560265_predicted /// Ybx1
854 NM_031615 zinc finger protein 148 Zfp148
855 BE109605 zinc finger protein 162 Zfpl62
856 BE111799 Zinc finger protein 346 (predicted) Zfp346_predicted
857 BM392399 zinc finger protein 622 Zfp622
858 AA819804 Zinc finger protein 91 Zfp91
859 BI289543 zinc finger, MYND domain containing 11 Znrynd11
860 AI317880 zinc responsive protein ZD7 LOC474154
*

Signal detected above background for gene probeset in 20% or more of the chips.

“Yes” indicates significant differential gene expression within the Sham, SBR50, SBR50/IL-6, and SBR50/IL-6/G groups using False Discovery Rate (FDR) = 10%. “No” indicates not significant differential gene expression. “N/A” indicates genes not included in the analysis because of not being detected in at least 20% of the chips. “NS” indicates no gene symbol.

To identify genes differentially expressed among the experimental groups, the data were filtered to remove genes with nearly uniformly low expression (absent on ≥ 80% of chips). Of the 860 apoptosis-related genes represented on the chips, 731 genes met the requirement of this filtering process and were included in the analysis. One-way ANOVA (see Materials and Methods) was then performed which identified 350 apoptosis genes with differential expression among four experimental groups– sham, SBR50, SBR50/IL-6, and SBR50/IL-6/G– at a False Discovery Rate (FDR) = 10% (Table 2). Of the 350 apoptosis pathway genes whose expression was altered among the four groups, 311 were altered in the SBR50 vs. sham comparison (Figure 5A and Table 2). Among the genes whose differential expression was altered in the SBR50 vs. sham comparison, the transcripts of the majority of these genes (193 genes) were increased in SBR50 vs. sham by 4.2 ± 1.2 fold (range = 1.02 to 92.3 fold) while transcripts of 118 genes were decreased in SBR50 vs. sham by 1.5 ± 1.2 fold (range = 1.1 to 3.3 fold; Figure 5B). Importantly, 106 of the 193 genes (55%) that were increased in the SBR50 vs. sham group, were decreased significantly in the SBR50/IL- 6 vs. SBR50 group by 1.8 ± 0.9 fold (range = 1.3 to 14.3 fold) and 108 of the 118 genes (92%) that were decreased in SBR50 group, were increased significantly in the SBR50/IL-6 group by 1.4 ± 0.7 fold (range = 1.2 to 2.9 fold; Figure 5B). Thus, of the genes whose transcript levels were altered in SBR50 vs. sham group, 214 of 311 (69%) returned to sham level or were “normalized” in the SBR50/IL-6 group.

Table 2.

Apoptosis-related genes differentially expressed in the SBR50 vs. SHAM comparison

SBR50 vs. Sham SBR50/IL-6 vs. SBR50 SBR50/IL/6/Gvs.SBR50/IL-6

mRNA Probeset Number Gene Name* Gene Symbol Fold P/Sham FDR Fold IL-6/p FDR Fold G/IL-6 FDR
GROUP IA GENES INCREASED IN SBR50 vs. SHAM AND DECREASED IN SBR50/IL-6 vs. SBR50
NM_012620 serine (or cysteine) peptidase inbitior, clade E, member 1 Serpine1 29.09 0.000 0.25 0.004 1.73 0.147
B1284218 solute carrier family 2 (faciliated glucose transporter), member 1 Slc2a1 20.29 0.000 0.26 0.002 1.72 0.079
NM_012580 heme oxygenase (decycling) 1 Hmoxl 19.97 0.002 0.13 0.020 1.20 0.846
M58040 transferrin receptor Tfrc 10.82 0.000 0.39 0.044 1.07 0.899
D64048 phosphatidylinositol 3-kinase, regulatory subunit, polypeptide 1 Pik3rl 6.17 0.003 0.21 0.014 0.64 0.433
U02315 neuregulin 1 Nrg1 5.08 0.002 0.43 0.062 0.70 0.428
NM_017334 cAMP responsive element modulator 3081 0.001 0.59 0.079 0.57 0.078
AI172056 myeloid cell leukemia sequence 1 Mcl1 3.43 0.001 0.46 0.019 0.87 0.669
NM_080902 hypoxia induced gene 1 Hig1 3032 0.001 0.36 0.005 1.09 0.773
AF279286 Bc12-like 1 Bc1211 3023 0.001 0.40 0.007 1.36 0.300
BI290699 phosphatidylinositol 3-kinase, catalytic, alpha polypeptide Pik3ca 3.18 0.000 0.31 0.002 0.90 0.692
NM_019142 protein kinase, AMP-activated, alpha 1 catalytic subunit Prkaa1 3.08 0.000 0.42 0.004 1.12 0.657
L062638 platelet derived growth factor, alpha Pdgfa 3.00 0.002 0.43 0.015 1.05 0.885
U69550 phospholipase D1 PlaD1 2.72 0.000 0.54 0.009 0.98 0.945
AB001382 secreted phosphoprotein 1 Sppl 2.67 0.013 0.45 0.048 2.08 0.077
NM_013052 tyrosine 3-monooxygenase/tyrptophan 5-monooxygenase activation protein, eta polypeptide Ywhah 2.42 0.002 0.66 0.095 2.95 0.001
NM_053843 Fc receptor, IgG, low affinity III /// Fc gamma receptor II beta Fcgr3 2.37 0.002 0.52 0.019 1.67 0.058
NM_017039 protein phosphate 2 (formerly 2A), catalytic subunit, alpha isoform Ppp2ca 2.33 0.001 0.59 0.018 1.76 0.015
BE113920 signal transducer and activator of transcription 3 Stat3 2.33 0.086 0.38 0.068 1.57 0.395
NM_019376 tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, gamma polypeptide Ywhag 2.30 0.000 0.57 0.004 1.35 0.059
NM_053887 mitogen activated protein kinase kinase kinase 1 Map3k1 2.24 0.036 0.38 0.022 1.19 0.686
NM_031140 Vimentin Vim 2.19 0.001 0.51 0.004 1.66 0.018
NM_031514 Janus kinase 2 Jak2 2.18 0.017 0.049 0.037 0.96 0.917
NM_031832 lectin, galactose binding, soluble 3 Lgals3 2.03 0.066 0.46 0.061 2.58 0.033
NM_019232 serum/glucocorticoid regulated kinase Sgk 2.00 0.080 0.47 0.074 0.94 0.902
AI409867 cystatin B CstB 1.89 0.002 0.55 0.007 1.41 0.080
NM_017255 purinergic receptor P2Y, G-protein coupled 2 P2ryl 1.82 0.006 0.49 0.005 1.46 0.082
NM_017022 integrin beta 1 (fibronectin receptor beta) Itgbl 1.82 0.001 0.61 0.006 1.15 0.351
NM_012886 tissue inhibitor of metalloproteinase 3 (Sorsby fundus dystrophy, pseudoinflammatory) Timp3 1.80 0.022 0.64 0.093 0.95 0.859
AI600237 eukaryotic translation elongation factor 1 epsilon 1 (predicted) Eefle1 1.79 0.008 0.67 0.061 0.72 0.135
AF228684 adenosine A2a receptor Adora2a 1.76 0.006 0.60 0.016 1.93 0.005
BE108192 G1 to S phase transition 1 Gspt1 1.74 0.000 0.70 0.004 1.03 0.772
NM_053619 complement component 5, receptor 1 C5rl 1.73 0.003 0.51 0.004 1.84 0.004
B1285434 tubulin, alpha 1 /// tubulin, alpha 6 /// similar to Tubulin alpha-2 chain (Alpha-tubulin 2) (predicted) Tubal 1.73 0.002 0.62 0.010 1.65 0.008
BM385544 presenilin 1 Psen1 1.67 0.008 0.65 0.029 1.80 0.008
BF281342 tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, theta polypeptide Ywhaq 1.63 0.001 0.69 0.009 1.09 0.463
NM_021989 tissue inhibitor of metalloproteinase 2 Timp2 1.56 0.002 0.68 0.009 0.82 0.135
AI236590 myeloid differentiation primary response gene 88 Myd88 1.56 0.044 0.56 0.019 1.30 0.272
BM392321 Homeodomain interacting protein kinase 2 (predicted) Hipk2_predicted 1.53 0.026 0.69 0.068 0.88 0.534
BG379941 Harvey rat sarcoma viral (v-Ha-Ias) oncogene homolog HraS 1.52 0.003 0.73 0.025 1.28 0.079
BF417479 24-dehydrocholestrol reductase Dhcr24 1.51 0.015 0.69 0.038 0.63 0.018
NM_017141 polymerase (DNA directed), beta Polb 1.51 0.075 0.61 0.048 1.46 0.134
NM_031787 Homeodomain interacting protein kinase 3 Hipk3 1.50 0.093 0.52 0.019 0.59 0.060
AA893484 fibronectin 1 Fank1 1.49 0.028 0.64 0.022 0.74 0.116
AI172276 protein phosphatase 1, regulatory (inhibitor) subunit 2 Ppplr2 1.47 0.008 0.79 0.092 1.06 0.727
NM_053720 apoptosis antagonizing transcription factor Aatf 1.46 0.070 0.58 0.021 1.17 0.490
M37394 epidermal growth factor receptor Hgfr 1.44 0.092 0.52 0.012 0.52 0.014
BG666933 cystatin C CstC 1.40 0.002 0.77 0.014 0.71 0.004
BM389673 cofilin 1, non-muscle Cfl1 1.39 0.086 0.61 0.021 1.33 0.171
EE111972 transforming growth factor, beta receptor 1 Trfbr1 1.37 0.090 0.63 0.027 0.92 0.680
AF388527 Serpinel mRNA binding protein 1 Serbp1 1.31 0.016 0.83 0.096 1.11 0.365
NM_0131315 RAS p21 protein activator 1 Rasal 1.28 0.033 0.66 0.005 1.04 0.771
B1282863 Prohibition Phb 1.26 0.092 0.76 0.069 0.70 0.026
BI282281 heat shock 70kDa protein 9A (predicted) Hspa9a_predicted 1.22 0.012 0.73 0.004 0.75 0.004
AI103616 ras-related C3 botulimum toxin substrate 1 Rax1 1.21 0.029 0.74 0.007 1.31 0.012
NM_019371 EGL nine homolog 3 Egin3 17.30 0.000 0.07 0.000 0.81 0.493
AI602811 dual specificity phosphatase 6 Dusp6 15.48 0.000 0.44 0.012 1.27 0.418
BI303379 tumor necrosis factor receptor superfamily, member 12a Tnfrsf12a 9.82 0.002 0.29 0.064 2.08 0.277
NM_012591 interferon regulatory factor 1 Irf1 9.32 0.000 0.39 0.005 1.33 0.311
NM_133306 oxidized low density lipoprotein (lectin-like) receptor 1 Oldlr1 9.02 0.000 0.30 0.004 2.01 0.041
NM_024127 growth arrest and DNA-damage-inducible 45 alpha Gadd45a 6.25 0.001 0.50 0.098 1.95 0.130
NM_053420 BCL2/adenovirus E1B 19 kDa-interacting protein 3 Bnip3 6.07 0.000 0.19 0.002 0.88 0.734
AI232697 phosphatidylserine receptor Jmjd6 3.51 0.007 0.43 0.058 0.89 0.802
NM_021846 similar to MAP/microtubule affinity-regulating kinase 4 (MAP/microtubule affinity-regulating kinase like 1)(predicted) RGD1561096 3.46 0.001 0.43 0.018 0.86 0.662
B1287978 growth arrest and DNA-damage-inducible 45 beta Gadd45b 3.20 0.002 0.54 0.061 1.33 0.383
NM_021744 CD14 antigen Cd14 3.07 0.003 0.49 0.039 0.73 0.362
NM_012637 protein tyrosine phosphatase, non-receptor type 1 Ptpn1 3.02 0.007 0.35 0.015 0.47 0.069
B1275994 transglutaminase 2, C polypeptide Tgm2 2.98 0.000 0.45 0.004 2.99 0.000
NM_013026 syndecan 1 Sdc1 2.91 0.000 0.48 0.005 2.92 0.001
B1284739 LPS-induced TN factor Litaf 2.70 0.000 0.50 0.005 1.23 0.289
J04943 nucleophosmin 1 /// similar to Nucleophosmin (NPM) (Nucleolar phosphoprotein B23) (Numatrin)(Nucleolar protein NO348) Npm1 /// LOC300303 2.67 0.001 0.50 0.010 1.42 0.152
AB049572 sphingosine kinase 1 Sphk1 2.63 0.002 0.61 0.067 0.60 0.074
AI411788 protein phosphatase 2 (formerly 2A), regulatory subunit A (PR 65), beta isoform Ppp2f1b 2.63 0.000 0.39 0.002 1.55 0.049
B1289109 max binding protein (predicted) Mnt 2.44 0.004 0.51 0.027 1.40 0.258
X57764 endothelin receptor type B Ednrb 2.43 0.002 0.58 0.028 1.11 0.673
NM_134432 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Agt 2.11 0.002 0.43 0.004 1.31 0.233
AF136231 caspase 2 Casp2 2.04 0.009 0.38 0.004 0.86 0.592
BM387008 caspase 3, apoptosis related cysteine protease Casp3 2.00 0.001 0.54 0.004 1.46 0.037
DI3341 mitogen activated protein kinase kinase 1 Map2k1 1.96 0.007 0.62 0.055 1.81 0.026
NM_053974 eukaryotic translation initation factor 4E Eif4eb 1 1.92 0.003 0.64 0.028 1.12 0.584
AF066016 nuclear receptor subfamily 3, group C, member 1 Nr-3c1 1.85 0.006 0.59 0.022 1.10 0.680
BF283772 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Rela 1.67 0.012 0.71 0.091 1.27 0.256
NM_013091 tumor necrosis factor receptor superfamily, member 1a Trfrsf1a 1.62 0.036 0.54 0.017 2.08 0.009
NM_017040 protein phosphatase 2 (formerly 2A), catalytic subunit, beta isoform Ppp2cb 1.60 0.002 0.64 0.005 0.98 0.899
BM386683 Stanniocalcin 1 Stc1 1.59 0.016 0.65 0.033 0.80 0.264
AI012221 chloride intracellular channel 1 Clic1 1.59 0.002 0.77 0.149 1.82 0.001
B1282898 BCL2-associated athanogene 5 Bag5 1.57 0.002 0.61 0.004 0.78 0.077
B1296393 programmed cell death 6 (predicted) Pdcd6 1.49 0.002 0.70 0.007 1.23 0.083
AI169001 antophagy-related 12 (yeast) Atg12 1.47 0.004 0.65 0.006 0.90 0.387
B1285751 cullin 3 (predicted) Cu13_predicted 1.46 0.012 0.62 0.006 0.87 0.373
NM_133307 protein kinase C, delta Prkcd 1.42 0.025 0.73 0.058 1.23 0.207
B1289418 CD38 antigen Cd38 1.42 0.091 0.68 0.088 1.19 0.433
NM_012696 kininogen 1 /// K-kininogen /// similar to alpha-1 major acute phase protein prepeptide Kng1 1.42 0.019 0.68 0.019 1.84 0.002
AA859665 Nuclear pore associated protein Npap60 1.40 0.012 0.69 0.012 0.89 0.401
NM_080887 thioredoxin-like 1 Txnll 1.39 0.002 0.74 0.007 1.24 0.034
AA848545 similar to programmed cell death 10 Pdcd10 1.38 0.063 0.61 0.016 1.52 0.033
NM_012839 cytochrome c, somatic Cycs 1.38 0.001 0.77 0.007 1.03 0.772
B1282255 ribosomal protein S5 Rps5 1.34 0.001 0.85 0.025 0.78 0.005
NM_022510 ribosomal protein L4 Rp14 1.33 0.031 0.73 0.025 0.75 0.044
AF411318 metallothionein 1a MtIa 1.29 0.058 0.62 0.006 0.90 0.456
NM_013065 protein phosphatase 1, catalytic subunit, beta isoform Ppp2cb 1.26 0.051 0.75 0.028 0.98 0.865
B1283681 eukaryotic translation initation factor 5A Eif5a 1.25 0.031 0.77 0.023 1.29 0.031
NM_080888 BCL2/adenovirus E1B 19 kDa-interacting protein 3-like Bnip31 1.19 0.077 0.73 0.010 0.60 0.001
BF281741 glioma tumor suppressor candidate region gene 2 GItscr2 1.18 0.067 0.84 0.068 0.93 0.461
BM389310 DEAD (Asp-Glu-Ala-Asp) box polypeptide 41 (predicted) Ddx41_predicted 1.18 0.096 0.77 0.021 1.33 0.019
GROUP 1B GENES INCREASED IN SBR50 vs. SHAM AND UNCHANGED IN SBR50/IL-6 vs.SBR50
NM_012912 activating transcription factor 3 Atf3 92.25 0.000 1.36 0.435 0.45 0.054
NM_031970 heat shock 27kDa protein 1 Hspbl 11.79 0.006 4.12 0.100 0.44 0.360
NM_053565 suppressor of cytokine signaling 3 Socs3 10.14 0.000 0.51 0.118 1.80 0.177
NM_012603 myeloctomatosis viral oncogene homolog (avain) Myc 9.53 0.000 0.65 0.357 1.08 0.894
BM384926 DnaJ (Hsp40) homolog, subfamily B, member 1 (predicted) Dnajb1 9.09 0.005 2.79 0.155 0.26 0.081
NM_013154 CCAAT CCAAT/enhancer binding protein (C/EBP), delta Cebpd 6.32 0.000 0.60 0.151 1.38 0.387
AF080594 vascular endothelial growth factor A Vegfa 6.14 0.000 0.60 0.188 1.04 0.923
BG671521 heat shock protein 1, alpha Hspca 4.72 0.001 1.34 0.465 0.82 0.658
BI284349 myeloid differentiation primary response gene 116 Myd116 4.65 0.004 1.71 0.270 0.98 0.969
AF007789 plasminogen activator, urokinase receptor Plaur 4.28 0.001 0.62 0.151 1.17 0.662
NM_021836 Jun-B oncogene Junb 4.12 0.000 1.31 0.303 0.94 0.845
NM_022542 ras homolog gene family, member B Rhob 4.06 0.001 1.19 0.629 1.05 0.900
NM_012551 early growth response 1 Egrl 3.89 0.001 1.15 0.693 0.92 0.839
NM_058208 suppressor of cytokine signaling 2 Socs2 3.13 0.045 0.45 0.178 1.51 0.499
NM_012699 DnaJ (Hsp40) homolog, subfamily B, member 9 Dnajb9 2.53 0.019 0.55 0.132 0.55 0.147
NM_013085 plasminogen activator, urokinase Plau 2.23 0.085 0.81 0.690 0.87 0.791
NM_133416 B-cell leukemia/lymphoma 2 related protein A1 Bcl2a1 2.22 0.048 0.81 0.630 2.78 0.028
BM392366 platelet-activating factor acetylhtdrolase, isoform 1b, alpha2 subunit Pafah1b2 2.13 0.007 0.84 0.511 1.26 0.398
AI178012 retinoblastoma 1 Rb1 1.96 0.000 1.14 0.331 1.43 0.018
BI294137 Hexokinase 2 Hk2 1.89 0.006 0.71 0.119 1.00 0.985
AI008680 benzodiazepine receptor, peripheral Bzrp 1.86 0.003 0.85 0.383 2.82 0.000
NM_017034 proviral integration site 1 Pim1 1.85 0.012 0.88 0.586 1.09 0.736
NM_012904 annexi A1 Anxa 5 1.82 0.036 0.97 0.939 0.78
BE109605 zinc finger protein 162 Zfp162 1.74 0.016 0.75 0.199 0.95 0.837
NM_013151 plasminogen activator, tissue Plat 1.68 0.008 0.91 0.630 1.14 0.499
AI178772 prothymosin alpha Ptma 1.67 0.010 0.86 0.424 1.42 0.084
NM_012734 hexokinase 1 Hk1 1.67 0.012 0.73 0.116 0.86 0.459
NM_012523 CD53 antigen Cd53 1.66 0.013 0.73 0.117 1.14 0.517
AI237389 heat shock 90kDa protein 1, beta Hspcb 1.64 0.032 1.30 0.258 1.49 0.106
NM_022531 Desmin Des 1.60 0.032 0.82 0.380 1.53 0.076
BI296385 similar to chemokine (C-X-C motif) ligand 16 Cxcl16 1.59 0.017 0.82 0.312 1.65 0.024
AA945737 chemokine (C-X-C motif) receptor 4 Cxcr4 1.49 0.051 0.88 0.546 1.62 0.037
NM_012715 Adrenomedullin Adm 1.48 0.057 1.12 0.605 1.76 0.020
J04024 ATPase, Ca++ transporting, cardiac muscle, slow twitch 2 Atp2a2 1.47 0.035 0.99 0.988 1.03 0.900
NM_024359 hypoxia inducible factor 1, alpha subunit Hifla 1.47 0.008 0.86 0.299 1.19 0.235
M83297 protein phosphatase 2 (formerly 2A), regulatory subunit B (PR 52), alpha isoform Ppp2ca 1.37 0.080 0.81 0.275 1.06 0.778
BE116857 apoptotic chormatin condensation inducer 1 Acin1 1.30 0.089 0.92 0.605 1.14 0.421
AI411586 Sequestosome 1 Sqstm1 1.29 0.040 0.94 0.673 1.21 0.153
M25590 seminal vesicle protein 4 Svp4 1.28 0.072 0.92 0.601 0.93 0.625
U69550 phospholipase D1 Pldl 1.26 0.75 0.093 0.585 0.77 0.074
NM_019161 cadherin 22 Cdh22 1.06 0.048 0.97 0.215 0.96 0.207
NM_012641 regenerating islet-derived 1 Regl 1.02 0.06 1.00 0.825 0.96 0.018
BI288701 B-cell translocation gene 2, anti-proliferative Btg2 25.68 0.000 0.41 0.135 1.31 0.672
NM_024388 nuclear receptor subfamily 4, group A, member 1 Nr4a1 9042 0.002 2.22 0.220 0.98 0.977
NM_024134 DNA_damage inducible transcript 3 Ddit3 7.53 0.002 1.21 0.757 3.78 0.033
NM_1335578 dual specificity phosphatase 5 Dusp5 6.41 0.000 1.08 0.857 0.96 0.909
NM_013060 inhibitor of DNA binding 2 Id2 5.69 0.000 0.69 0.201 1.77 0.063
NM_031628 nuclear receptor subfamily 4, group A, member 3 Nr4a3 4.96 0.036 1.34 0.726 0.68 0.653
AI010427 cyclin-dependent kinase inhibitor 1A Cdkn1a 2.90 0.013 0.55 0.151 1.83 0.171
NM_053847 mitogen-activated protein kinase kinase kinase 8 Map3k8 2.89 0.008 0.59 0.170 1.52 0.289
NM_057138 CASP8 and FADD-like apoptosis regualtor Cfr 2.82 0.051 0.44 0.136 1.77 0.317
NM_053319 dynein light chain LC8-type 1 Dynll1 2.80 0.001 1.26 0.375 1.35 0.255
NM_053819 tissue inhibitor of metalloproteinase 1 Timp1 2.66 0.002 0.82 0.488 1.96 0.030
NM_194457 seven in absentia 2 Siah2 2.47 0.005 0.83 0.527 0.87 0.662
NM_012954 fos-like antigen 2 /// FBJ asteosarcoma oncogene B Fos12 2.42 0.008 1.02 0.974 1.49 0.223
NM_012966 heat shock 10 kDa protein 1 (chaperonin 10) Hspel 2.31 0.000 1.23 0.255 0.55 0.008
BG671549 superoxide dismutase 2, mitochondrial Sod2 2.27 0.077 0.57 0.238 3.76 0.018
U05989 PRKC, apoptosis. WTI, regulator Paur 2.06 0.003 1.35 0.165 0.99 0.976
NM_053957 amyloid beta (A4) precursor protein-binding, family B, member 3 Apbb3 2.03 0.014 0.69 0.184 1.24 0.463
NM_017180 pleckstrin homology-like domain, family A, member 1 Phidal 1.96 0.051 1.09 0.844 0.98 0.968
AB020367 tribbles homolog 3 (Drosophila) Trib3 1.91 0.036 0.60 0.118 1.87 0.069
NM_053394 Kruppel-like factor 5 Klf5 1.85 0.062 0.62 0.165 0.86 0.680
NM_012953 fos-like antigen 1 Fosl1 1.84 0.061 0.65 0.204 0.82 0.595
NM_017258 B-cell translocation gene 1, anti-proliferative Btg1 1.70 0.010 0.73 0.117 1.17 0.450
NM_022612 BCL2-like 11 (apoptosis faciliator) Bcl2111 1.55 0.020 0.75 0.129 0.83 0.353
AA892271 BCL2-like 13 (apoptosis faciliator) (predicted) Bcl2113 1.49 0.036 0.87 0.499 1.19 0.387
AA957342 peptidylprolyl isomerase D (cyclophilin D) Ppid 1.46 0.051 1.24 0.286 1.20 0.384
NM_053289 pancreatitis-associated protein Pap 1.43 0.100 0.70 0.120 1.11 0.672
NM_017006 glucose-6-phosphate dehydrogenase x-linked G6pdx 1.41 0.005 0.89 0.293 1.83 0.000
NM_012829 Cholecystokinin Cck 1.39 0.037 0.89 0.463 0.62 0.013
AF235993 Bc12-associated X protein Box 1.37 0.072 1017 0.399 2.76 0.000
NM_019282 gremlin 1 homolog cysteine knot superfamily (Xenopus laevis) Grem1 1.26 0.087 0.84 0.215 0.92 0.568
NM_131911 acidic nuclear phosphoprotein 32 family, member B Anp32b 1.23 0.025 0.86 0.117 1.18 0.095
B1280304 Bc12-associated athanogene 1 (predicted) Bag1 1.20 0.086 1.01 0.955 1.34 0.019
BF287444 glycogen synthase kinase 3 beta Gsk3b 1.18 0.067 0.92 0.349 1.04 0.669
NM_031038 Gonadotrophin releasing hormone receptor Gnrhr 1.15 0.050 0.89 0.113 0.97 0.736
AF081196 RAS guanyl releasing protein 1 Rasgrp1 1.10 0.018 0.94 0.140 0.94 0.154
NM_012650 sex hormone binding globulin Shbg 1.02 0.065 0.99 0.566 0.97 0.028
GROUP 2A GENES DECREASED IN SBR50 vs. SHAM AND INCREASED IN SBR50/IL-6 vs. SBR50
NM_052980 nuclear receptor subfamily 1, group I, member 2 Nrli2 0.09 0.000 1.38 0.027 1.39 0.031
NM_021745 nuclear receptor subfamily 1, group H, member 4 Nrlh4 0.40 0.002 2.99 0.003 1.35 0.251
NM_022407 aldehyde dehydrogenase family 1, member A1 Aldhlal 0.43 0.015 2.13 0.038 0.42 0.024
NM_012805 retinoid X receptor alpha Rxra 0.49 0.000 1.27 0.084 0.81 0.130
NM_012493 alpha-fetoprotein Afp 0.50 0.007 2.02 0.012 0.86 0.547
AI235465 steroid sensitive gene 1 Ssgl 0.50 0.012 2.07 0.015 0.53 0.030
NM_133400 apobec-1 complementation factor Acf 0.55 0.004 1.63 0.019 0.85 0.387
BM392373 CEA-related cell adhestion molecule 1 Ceacaml 0.58 0.003 1.58 0.013 0.69 0.038
BG377107 DnaJ (Hsp40) homolog, subfamily A, member 3 Dnaja3 0.60 0.000 1.44 0.005 0.93 0.482
NM_012931 breast cancer anti-estrogen resistance 1 Bcarl 0.60 0.018 1.65 0.027 1.25 0.310
NM_022614 inhibin beta C Inhbc 0.61 0.020 1.64 0.028 0.94 0.809
BF282337 integral membrane protein 2B Itm2b 0.63 0.000 1.27 0.007 1.13 0.117
BI276999 testis enhanced gene transcript Tegt 0.63 0.000 1.39 0.002 0.79 0.005
AY083159 Stam binding protein Stambp 0.63 0.009 1.56 0.017 1.43 0.049
BY275921 anterior pharynx defective la homolog (C. elegans) Aphla 0.64 0.002 1.38 0.021 2.05 0.000
AF347936 interlekin 11 receptor alpha chain 1 Ill1rals 0.65 0.009 1.60 0.011 1.27 0.154
AW672589 nuclear factor of kappa light chain gene enhancer in B-cells inhibitor, alpha Nfkbia 0.65 0.003 1.82 0.002 2.10 0.000
BI279735 Ubiquilin 1 Frapl 0.68 0.001 1.41 0.004 1.34 0.008
NM_017181 fumarylac etoacetate hydrolase Fah 0.69 0.002 1.29 0.002 0.75 0.019
NM_031546 Regucalcin Rgn 0.69 0.004 1.57 0.004 0.31 0.000
NM_022277 caspase 8 Casp8 0.70 0.013 1.32 0.058 1.92 0.001
AW523747 adhesion molecule with Ig like domain 2 Amigo2 0.72 0.045 1.54 0.019 1.20 0.131
BI274345 ataxin 10 Atxn10 0.73 0.008 1.22 0.079 0.90 0.387
BI295511 forkhead box O1A Foxola 0.73 0.020 1.39 0.022 0.88 0.353
AI009817 succinate dehydrogenase complex, subunit C, integral membrane protein Sdhc 0.73 0.034 1.37 0.043 1.02 0.925
AI011448 Notch gene homolog 2 (Drosophila) Notch2 0.74 0.088 1.59 0.019 1.18 0.384
BI285459 Nicastrin Ncstn 0.76 0.003 1.20 0.037 1.34 0.005
AB015946 tubulin, gamma 1 Tubgl 0.78 0.090 1.79 0.004 1.31 0.104
NM_022399 Calreticulin Calr 0.80 0.016 1.31 0.012 1.05 0.595
M15481 insulin-like growth factor 1 Igfl 0.83 0.069 1.36 0.014 0.51 0.000
NM_024352 Macrophage stimulating 1 (hepatocyte growth factor-like) Mstl 0.83 0.038 1.28 0.017 0.84 0.077
NM_053491 Plasminogen Parl 0.83 0.009 1.13 0.073 0.82 0.014
BI281756 Parkinson disease (auosomal recessive, early onset) 7 Park7 0.85 0.036 1.35 0.004 1.13 0.166
NM_133317 transducer of ErbB-2 1 Tobl 0.34 0.002 2.81 0.006 1.41 0.284
AF335281 STEAP family member 3 Steap3 0.41 0.002 1.62 0.066 0.69 0.170
AI412114 etoposide induced 2–4 Mma Ei24 0.46 0.000 2.14 0.002 0.98 0.917
NM_012870 retinoblastoma-like 2 Rb12 0.49 0.006 1.54 0.079 1.03 0.909
NM_012870 tumor necrosis factor receptor superfamily, member 11b (osteoprotegerin) Tnfrsf11b 0.50 0.004 1.99 0.007 1.23 0.360
NM_0530902 kynurenimase (L-kynurenime hydrolase) Kynu 0.52 0.003 1.73 0.014 0.47 0.003
AI169638 sphingosine-l-phosphate phosphatase 1 Sgppl 0.53 0.002 1.51 0.028 1.47 0.048
BM388758 excision repair cross-complementing rodent repair deficiency, complementation group 3 Ercc3 0.57 0.002 1.54 0.016 1.66 0.008
BF398331 estrogen receptor-binding fragment-associated gene 9 Ebag9 0.61 0.013 1.41 0.082 0.81 0.300
AB004454 presenilin 2 Psen 2 0.62 0.002 1.32 0.051 1.60 0.006
NM_019158 aquaporin 8 Aqp8 0.64 0.018 1.66 0.016 0.92 0.692
NM_057146 complement component 9 C9 0.64 0.002 1.29 0.042 1.36 0.022
AF275151 androgen receptor-related apoptosis-associated protein CBL27 Cb127 0.65 0.001 1.36 0.011 1.03 0.772
NM_017319 protein disulfide isomerase associated 3 Pdia3 0.66 0.001 1.67 0.002 1.22 0.062
NM_053907 deoxyribonuclease I-like 3 Dnase112 0.66 0.005 1.36 0.034 0.66 0.012
AF262320 programmed cell death 8 Pdcd8 0.69 0.000 1.19 0.009 0.92 0.164
BM391371 Goliath LOC652955 0.71 0.001 1.26 0.016 0.82 0.034
AA819870 complement component 8, beta polypeptide (mapped) C8b 0.79 0.018 1.28 0.024 0.80 0.038
AI409930 B-cell receptor-associated protein 31 Bcap31 0.81 0.007 1.28 0.006 1.08 0.333
GROUP 2B GENES DECREASED IN SBR50 vs. SHAM AND UNCHANGED IN SBR50/IL-6 vs. SBR50
NM_012842 epidemal growth factor Egfr 0.32 0.000 1.29 0.204 0.59 0.021
BM385790 Kruppel-like factor 2 (lung) (predicted) Klf2 0.40 0.006 1.10 0.789 1.33 0.384
NM_032612 signal transducer and activator of transcription 1 Stat1 0.45 0.075 2.20 0.101 6.09 0.003
NM_053923 phosphatidylinositol 3-kinase, C2 domain containing, gamma polypeptide Pic3c2g 0.50 0.014 1.15 0.630 0.30 0.002
NM_031541 scavenger receptor class B, member 1 Scarb1 0.52 0.002 1.29 0.151 0.77 0.152
NM_053523 homocysteine-inducible, endoplasmic reticulum stress-inducible, ubiquitin-like domain member 1 Herpud 1 0.53 0.017 1.42 0.186 0.47 0.015
NM_013063 poly (ADP-ribose) polymerase family, member 1 Parp12_predicted 0.56 0.011 0.87 0.539 1.02 0.951
NM_017206 solute carrier family 6 (neurotransmitter transporter, taurine), member 6 Slc6a6 0.56 0.078 0.81 0.539 1.60 0.185
AA799421 protein kinase C, epsilon Prkce 0.58 0.002 0.79 0.119 1.50 0.019
NM_022185 phosphatidylinositol 3-kinase, regulatory subunit, polypeptide 2 Pik3r2 0.59 0.056 1.18 0.575 1.33 0.348
AI177631 bifunctional apoptosis regualtor bflar 0.59 0.086 0.93 0.857 1.52 0.230
NM_017322 mitogen-activated protein kinase 9 Mapk9 0.60 0.011 1.15 0.468 1.24 0.289
NM_017094 growth hormone receptor Ghr 0.61 0.072 1.15 0.650 0.60 0.096
AI716502 spkingosine kinase 2 Sphk2 0.64 0.046 1.44 0.119 1.06 0.837
BG378230 mitochondrial ribosomal protein S30 (predicted) Mrps30 0.64 0.016 1.10 0.611 0.46 0.002
NM_019364 sec 1 family domain containing 1 Scfd1 0.65 0.025 1.28 0.189 1.05 0.798
BM385237 annexin A4 Anxa5 0.66 0.087 1.49 0.120 2.32 0.007
BI289543 zicn finger, MYND domain containing 11 Zmynd11 0.67 0.006 1.05 0.744 1.24 0.137
U68544 peptidylprolyl isomerase F (cyclophilin F) Ppif 0.67 0.003 1.07 0.587 0.88 0.348
NM_031053 mutL homolog 1 (E. coli) /// hypothetical gene supported by NM_031053 Mlhl 0.68 0.45 1038 0.103 1.30 0.200
X02904 glutathione-S-transferase, pi 1///glutathione S-transferase, pi 2 Gstpl 0.71 0.012 1.22 0.136 0.79 0.089
NM_053777 mitogen activated protein kinase 8 interacting protein Mapk8ip 0.71 0.033 1.29 0.117 0.86 0.387
BE110607 similar to apoptosis related protein APR-3; p18 (predicted) RGD1311605_predicted 0.72 0.072 1.29 0.170 0.69 0.070
NM_012803 protein C Sftpc 0.72 0.005 1.18 0.123 0.70 0.007
U69109 protein tyrosine kinase 2 beta Ptk2b 0.74 0.081 1.11 0.561 1.21 0.321
AA819804 Zinc finger protein 91 Zfp91 0.75 0.090 1.16 0.420 1.26 0.216
BG673589 paxillin Pxn 0.75 0.028 1.16 0.267 1.17 0.258
BF419646 retimoic acid receptor, beta Ram 0.75 0.086 1.14 D.468 2.37 0.001
NM_053739 beclin 1 (coiled-coil, myosin-like BCL2-interacting protein) Becn1 0.75 0.037 1.17 0.258 1.11 0.470
AI171615 chromosome segregation 1-like (S. cerevisiae) (predicted) Csell_predicted 0.78 0.062 1.26 0.104 1.45 0.019
NM_053864 valosin-containing protein Vcpip1 0.78 0.010 1.06 0.501 1.14 0.176
AIl70385 SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 2 Smarca2 0.80 0.089 1.21 0.170 0.99 0.923
NM_012660 eukaryotic translation elongation factor 1 alpha 2 Eefl2a 0.81 0.090 1.02 0.931 0.86 0.276
NM_133409 integrin linked kinase Ilk 0.83 0.087 1.07 0.582 1.27 0.054
AA800669 protein phosphatase 2 (formerly 2A), regulatory subunit A (PR 65), alpha isoform Ppp2f1a 0.88 0.091 1.00 0.979 1.23 0.024
NM_013149 aryl hydrocarbon receptor Ahr 0.32 0.001 1.21 0.502 0.92 0.776
NM_022180 hepatocyte nuclear factor 4, alpha Hrf4a 0.33 0.000 1.35 0.119 1.45 0.066
BI297236 Transformation related protein 53 inducible nuclear protein 1 Trp53inp1 0.49 0.053 1.47 0.317 1.06 0.894
BE1080508 Procollagen, type XVIII, alpha 1 Coll8a1 0.49 0.045 1.56 0.217 0.67 0.283
AB019366 poly (ADP-ribose) glycohydrolase Parg 0.52 0.004 1.12 0.611 1.49 0.072
NM_012498 aldo-keto reductase family 1,member B4 (aldose reductase) Akr1b4 053 0.003 1.33 0.138 1.10 0.660
NM_053307 methionine sulfoxide reductase A Msra 0.60 0.002 1.26 0.125 0.47 0.001
BF400782 protein kinase, DNA activated, catalytic polypeptide (predicted) Prkdc_predicted 0.60 0.005 1.06 0.733 0.89 0.506
NM_017079 CDldl antigen Cdldl 0.60 0.031 1.46 0.118 1.28 0.317
NM_053448 histone deacetylase 3 Hdac3 0.61 0.028 1.39 0.147 1.24 0.367
A1408078 BCL2-associated transcription factor 1 Bclaf1 0.64 0.028 0.93 0.748 0.72 0.130
BF283688 tumor necrosis factor ligand superfamily member 12 Tnflfl2 0.65 0.069 1.46 0.123 1.19 0.497
B1286396 PERP, TP53 apoptosis affector (predicted) Perp 0.69 0.042 1.13 0.540 0.51 0.005
BE107162 high mobility group box 1 Hmgb1 0.69 0.005 1.17 0.199 0.89 0.374
BF408447 programmed cell death 5 (predicted) Pdcd8 0.70 0.013 1.25 0.123 1.19 0.236
U06230 protein S (alpha) Prosl 0.70 0.019 1.14 0.405 0.70 0.034
A1227743 Fas-activated serine/threonine kinase Fastk 0.72 0.017 1.00 0.989 1.11 0.459
AIl76519 immediate early response 3 Ier3 0.73 0.051 1.32 0.110 1.16 0.396
NM_130406 Fas-associated factor 1 Fafl 0.74 0.061 1.16 0.389 1.51 0.030
BM389034 TNF receptor-associated protein 1 Trap 1 0.74 0.036 1.08 0.636 0.91 0.554
AA800180 thioredoxin 2 Txn2 0.75 0.029 1.10 0.465 1.18 0.230
D14592 mitogen activated protein kinase kinase 2 Map2k2 0.75 0.033 1.23 0.132 1.37 0.037
BG665132 mitochondrial carrier homolog 1 (C. elegans) Mtch1 0.77 0.022 1.08 0.537 1.17 0.207
BM390196 thioredoxin domain containig 5 (predicted) Txndc5_predicted 0.77 0.043 1.22 0.126 0.65 0.008
BF284481 SH3-domain GRB2-like B1 (endophilin) Sh3glbl 0.77 0.087 0.93 0.673 1.23 0.213
BM383722 NCK-assocaited protein 1 Nell1 0.80 0.070 1.06 0.646 0.95 0.719
NM_031978 proteasome (prosome, macropain) 26S subunit, non-ATPase, 1 Psmd1 0.81 0.093 1.14 0.336 1.33 0.052
AA946474 Calpastatin Cast 0.82 0.089 0.96 0.781 1.15 0.288
GROUP 3 GENES INCREASED IN SBR50 vs. SHAM AND INCREASED IN SBR50/IL-6 vs. SBR50
B1278231 heat shock 70kD protein 1B (mapped) Hspalb 51.80 0.002 7.75 0.072 0.06 0.026
NM_031971 heat shock 70kD protein 1A /// heat shock 70kD protein 1B (mapped) Hspala 45.41 0.002 7.32 0.061 0.08 0.026
AI236601 heat shock 105kDa/110kDa protein 1 Hsph1 8.49 0.006 3.36 0.097 0.43 0.264
B1288619 Jun oncogene Jun 5.64 0.001 2.68 0.021 0.51 0.102
NM_031327 cysteine rich protein 61 Cyr61 3.48 0.003 4.43 0.004 0.52 0.105
AA818262 angiopoietin-like 4 Angpt14 2.98 0.000 1.50 0.028 1.37 0.089
NM_012935 crystallin, alpha B Cryab 1.82 0.016 1.84 0.021 0.88 0.619
AF077354 heat shock protein 4 Hspa4 1.50 0.037 l.46 0.063 1.20 0.381
A1599423 growth arrest and DNA-damage-inducible 45 gamma Gadd45g 8.66 0.000 2.50 0.005 0.80 0.411
A1231792 Bc12-associated athanogene 3 Bag3 3.76 0.018 3.16 0.049 0.16 0.007
*

Genes listed in regular type are anti-apoptotic, while genes listed in italics are pro-apoptotic.

FDR: False discovery rate.

Figure 5.

Figure 5

Effect of trauma/HS without or with IL-6 treatment on liver apoptosisrelated gene expression; impact of Stat3 inhibition on the IL-6 effect. In panel A, a heat map of apoptosis pathway genes is shown containing those genes whose expression is altered within the 4 groups. Columns represent samples from the 4 groups examined as indicated (S, Sham; P, placebo-treated SBR50; I, IL-6-treated SBR50/IL-6; and G, animals pre-treated with G-quartet ODN prior to HS and IL-6 treatment, SBR50/IL-6/G). Rows represent genes as listed in Table 1. Red indicates a level of expression above the mean expression of a gene within the experimental group. White indicates a level of expression at the mean within the experimental group while blue indicates a level of expression below the mean within the experimental groups. Log2-fold changes in expression levels of subsets of apoptosis-related genes are shown in panels B and C comparing SBR50 vs. sham (open bars), SBR50/IL-6 vs. SBR50 (gray bars) and SBR50/IL-6/G vs. SBR50/IL-6/N (stippled bars). In panel B, the 308 apoptosis-related genes whose expression levels were changed in SBR50 vs. sham were separated into those genes whose transcript levels were increased in SBR50 vs. sham (134 genes; left side of panel) and those whose transcript levels were decreased in SBR50 vs. sham (90 genes; right side of the panel). Bars shown represent mean ± SD of the Log2-fold change in gene expression levels for each comparison. In panel C, the overall effect of trauma/HS in transcript levels of pro- and antiapoptotic genes is shown. In the left side of the panel, the mean ± SD of the Log2-fold change in gene expression levels of 87 proapoptotic genes whose expression was increased in the SBR50 vs. sham comparison is shown (open bar). The expression of 74 of 87 of these genes was decreased in the SBR50/IL-6 vs. SBR50 comparison (gray bar). In the right side of the panel, the mean ±SD of the Log2-fold change in gene expression levels of 68 anti-apoptotic genes whose expression was decreased in the SBR50 vs. sham comparison is shown (open bar). The expression of 63 of these genes was increased in the SBR50/IL-6 vs. SBR50 comparison (gray bar).

One hundred and twenty-five of the 214 genes altered in the SBR50 vs. sham comparison, and normalized in the SBR50/IL-6 vs. SBR50 comparison, were also altered in the SBR50/IL-6 vs. SBR50/IL-6/G comparison. Fifty seven of these 125 genes (46%) were altered in the opposite direction as the SBR50/IL-6 vs. SBR50 comparison, consistent with the hypothesis that IL-6 normalizes the trauma/HS-induced lung apoptosis transcriptome in part through activation of Stat3.

Apoptosis-related genes consist of those encoding proteins that prevent apoptosis (anti-apoptotic genes) and those encoding proteins that induce apoptosis (pro-apoptotic genes). To identify candidate apoptosis-related genes whose altered expression caused trauma/HS- induced AEC apoptosis, we focused on anti-apoptotic genes whose transcript levels were decreased by trauma/HS and on pro-apoptotic genes whose transcript levels were increased by trauma/HS. Among the genes differentially expressed in the SBR50 vs. sham comparison, 69 anti-apoptotic genes were decreased and 90 pro-apoptotic genes were increased (Figure 5C). Expression levels of 65 out of 69 (94%) anti-apoptotic genes decreased by trauma/HS were increased by IL-6 treatment; 76 of 90 (84%) of the pro-apoptotic genes that were increased by trauma/HS, were decreased by IL-6 treatment. Finally, the expression of 46% of anti-apoptotic genes increased by IL-6 treatment were decreased by pre-treatment with T40214; conversely 63% of pro-apoptotic genes decreased by IL-6 treatment were increased by T40214 pre-treatment (Figure 5C).

Pro-apoptotic genes whose expression was increased ≥ 6-fold by trauma/HS (≥2.5-fold upregulated) were EGL nine homolog 3 (Egln3; 17.3-fold), dual specificity phosphatase 6 (Dusp6; 15.5-fold), tumor necrosis factor receptor superfamily 12a (Tnfrsf12a; 9.8- fold), interferon regulatory factor 1 (Irf1; 9.3- fold), oxidized low density lipoprotein receptor 1 (Oldlr1; 9.0-fold), growth arrest and DNA- damage-inducible 45 alpha (Gadd45a; 6.3- fold), and BCL2 adenovirus E1B interacting protein 3 (Bnip3; 6.1-fold, Table 2). The expression of each was decreased in the IL-6- treated group by 1.7–14.3 fold (Table 2).

Discussion

In these studies, we demonstrated that trauma/HS-induced liver apoptosis occurs within 1 hr of resuscitation from trauma/HS and depends on the severity of shock with the degree of apoptosis increasing exponentially with increased duration of shock. There was an absolute requirement for resuscitation in order for apoptosis to occur; the finding that no apoptosis occurred in the absence of resuscitation suggested that complete prevention could be achieved by an appropriate intervention introduced at the start of resuscitation. IL-6 administration at the start of resuscitation completely prevented trauma/HS-induced liver apoptosis and was accompanied by increased levels of Stat3 activity within the liver. Pharmacological inhibition of Stat3 using the G-quartet oligodeoxynucleotide (GQ-ODN) T40214 markedly attenuated IL-6-mediated Stat3 activation and prevention of liver apoptosis. Mice deficient in Stat3β, an endogenous naturally occurring, dominant negative isoform of Stat3, were protected from trauma/HS- induced liver apoptosis confirming a role for Stat3, especially Stat3α, in protection against trauma/HS-induced liver apoptosis. Liver microarray analysis showed that 48% of known apoptosis–related genes were altered in trauma/HS. IL-6 “normalized” the expression of 69% of these genes; Stat3 was responsible for this normalization in 46% of the cases. Further examination of the microarray results indicated that the effect of IL-6 in the apoptosis transcriptome was two- fold; IL-6 increased levels of 96% of the anti-apoptotic gene transcripts whose levels were decreased by trauma/HS and also decreased transcript levels of 84% of the pro-apoptotic genes whose levels were increased by trauma/HS.

The liver is susceptible to injury following insults such as hemorrhagic shock. Since it is responsible for maintaining energy homeostasis, hepatic injury and dysfunction associated with hemorrhagic shock can affect other organs and lead to multiple organ failure and death [4042]. We have previously demonstrated in mice that trauma/HS induces liver apoptosis detected 24 hrs after the resuscitation [6]. In the current study, using our rat model of trauma/HS we found that liver apoptosis occur as early as 1 hour after reperfusion, and for the first time, that its severity depends on the duration of hypotension and requires resuscitation. Liver apoptosis due to ischemia/reperfusion injury has been shown to occur in humans and in experimental models of liver transplantation [43], as well as during hemorrhagic shock [41, 42]. In these studies, release of ROS during reperfusion was implicated in the pathogenesis of liver apoptosis during reperfusion in models of liver transplantation [42, 43], and may be contributing to liver apoptosis in our model.

Interventions aiming at preventing liver apoptosis have been shown to prevent parenchymal injury and improve animal survival in ischemic injury during liver transplant [43]. We previously demonstrated that exogenous administration of IL-6 decreased liver apoptosis in mice following trauma/HS [6], however, the mechanism for the anti-apoptotic effects of IL-6 was not determined. Kovalovich et al. provided evidence that IL-6 protects hepatocytes from carbon tetrachloride-induced apoptosis [44] as well as from Fas-mediated apoptosis [45] through prevention of rapid degradation of the anti-apoptotic proteins Bcl-2, Bcl-xL and FLIP in the livers. While this may contribute to the anti-apoptotic effect of IL-6 administration in our rat trauma/HS model, our finding that the IL-6 preventive effect was blocked by a specific Stat3 inhibitor, T40214, and could be replicated in mice by genetic deletion of Stat3β, a naturally occurring, dominant-negative isoform of Stat3, indicates that a large portion of the effect of IL-6 in our trauma/HS model is being mediated by Stat3 which acts at the transcriptional and not the post-translational level.

IL-6-mediated activation of Stat3 has been shown to protect against toxin-mediated hepatocyte apoptosis through mechanisms that involve increased levels of anti-apoptotic proteins such as Bcl-xL and Bcl-2 in the liver [46, 47]. Stat3 activation mediates increased transcription of anti-apoptotic genes like Bcl- xL, Bcl-2, and Mcl-1, as well as decreased transcription of pro-apoptotic genes, including Bad, Bnip3 and Casp3 [29, 3239]. Microarray analysis of the liver apoptosis transcriptome revealed that the expression levels of the pro- apoptotic genes Bnip3 and Casp3 were increased by 6- and 2-fold, respectively, following trauma/HS and were decreased by 5.4- and 4.1-fold respectively after IL-6 treatment. However, only the expression of Casp3 was increased by Stat3 pharmacological inhibition with T40214. None of the other apoptosis-related genes known to be modulated by Stat3 to promote apoptosis protection in other settings of liver injury were affected by IL-6 treatment or by pre-treatment with T40214, suggesting that other apoptosis-related genes are involved in the anti-apoptotic effects of IL-6-mediated Stat3 activation in our model of trauma/HS.

A complete assessment of the liver apoptosis transcriptome using oligonucleotide microarray analysis determined that trauma/HS altered the expression of 48% of apoptosis-related genes, of which 56% were anti-apoptotic and 44% were pro-apoptotic genes (Figure 5B). The overall effect of trauma/HS in the liver apoptosis transcriptome was to increase the expression of apoptosis-related genes. IL-6 administration induced the opposite effect by decreasing the expression of apoptosis-related genes whose expression was increased by trauma/HS, and by increasing the expression of apoptosis-related genes whose expression was decreased by trauma/HS (Figure 5C), suggesting a “normalizing” effect of IL-6 in the trauma/HS-induced liver apoptosis transcriptome. Stat3 inhibition, by GQ-ODN administration, reversed the IL-6 “normalizing” effect on gene expression in 46% of the apoptosis-related genes (Figure 5C). These results suggest that IL-6 administration promotes protection from trauma/HS-induced liver apoptosis by opposing the effects of trauma/HS in the liver apoptosis transcriptome in part by Stat3 activation. Anti- and pro-apoptotic subsets of genes were analyzed separately to determine the effect of trauma/HS in each subset of transcripts. Gene transcript levels in both, anti- and pro- apoptotic subsets were increased by trauma/HS. Interestingly, IL-6 normalized the expression of genes in both subsets of genes, an effect that was reversed by pre-treatment with Stat3 inhibitor (Figure 5C).

In our model of trauma/HS, pro-apoptotic genes upregulated by trauma/HS are likely to be mediators of trauma/HS-induced liver apoptosis, which is likely prevented by the IL-6 normalizing effect on their expression through Stat3 activation. Among the transcripts most up-regulated following trauma/HS and downregulated in the IL-6 treatment group were Egln3, Dusp6, Tnfrsf12, Irf1, Oldlr1, Gadd45a, and Bnip3. Egln3 is a prolyl hydroxylase which is induced in sympathetic neurons after nerve growth factor withdrawal, and induces apoptosis when overexpressed in pheochromocytoma cells [48]. Dusp6 is a cytosolic phosphatase with pancreatic tumor suppressive properties and mediates apoptosis and cell growth arrest by specifically inactivating extracellular signalregulated kinase (ERK) [49]. Tnfrs12 is known to sensitize melanoma cells to chemotherapy-induced apoptosis [50]. Irf1 is activated by serum from human patients with sepsis, and mediates apoptosis in fetal myocytes [51]. Oldlr1 is expressed in highly vascularized organs such as the lung and placenta, and in endothelial cells, smooth muscle cells, cardiomyocytes and activated macrophages. Oldlr1 is a type II glycoprotein and acts as a receptor for oxidized low-density lipoprotein (ox-LDL). Interaction with ox- LDL induces ROS, reduces NO and activates NFκB. It also increases expression of Bax and decreases expression of Bcl-2. Oldlr1 is known to induce apoptosis of vascular endothelial cells and vascular smooth muscle cells in a model of cardiac ischemia reperfusion [52]. Gadd45g belongs to a family of proteins involved in DNA damage response and cell growth arrest. It is ubiquitously expressed in all normal adult and fetal tissues. Gadd45g activates MTK1 kinase activity in response to environmental stresses leading to apoptosis through the p38/c-Jun kinase pathway. Downregulation of Gadd45g prevents apoptosis of cancer cells [53].

The protective effects of IL-6 in trauma/HS-induced liver injury were mediated in large measure by Stat3 through its ability to normalize the apoptosis transcriptome. Our findings provide evidence that support the use of IL-6 as a potential therapeutic agent to protect against liver injury and dysfunction by blocking apoptosis early after reperfusion. Such an intervention has the potential to prevent multiple organ failure and improve survival in the setting of severe hemorrhagic shock.

Acknowledgments

This work was supported, in part, by grant HL07619 (DJT) and T32-HL66991 (AM) from the National Heart, Lung and Blood Institute of the National Institutes of Health and H48839 (AM) from the American Lung Association.

References

  • 1.Minino AM AR, Fingerhut LA, Boudreault MA, Warner M. Deaths: Injuries 2002. National Vital Statistics Reports. 2006;Vol. 54:1–125. [PubMed] [Google Scholar]
  • 2.Moore FA, Sauaia A, Moore EE, Haenel JB, Burch JM, Lezotte DC. Postinjury multiple organ failure: a bimodal phenomenon. J Trauma. 1996;40:501–10. doi: 10.1097/00005373-199604000-00001. discussion 510–512. [DOI] [PubMed] [Google Scholar]
  • 3.Ciesla DJ ME, Johnson JL, Burch JM, Cothren CC, Sauaia A. The role of the lung in postinjury multiple organ failure. Surgery. 2005;138:749–758. doi: 10.1016/j.surg.2005.07.020. [DOI] [PubMed] [Google Scholar]
  • 4.Jarrar D, Wang P, Cioffi WG, Bland KI, Chaudry IH. Critical role of oxygen radicals in the initiation of hepatic depression after trauma hemorrhage. J Trauma. 2000;49:879–885. doi: 10.1097/00005373-200011000-00015. [DOI] [PubMed] [Google Scholar]
  • 5.Heckbert SR, Vedder NB, Hoffman W, et al. Outcome after hemorrhagic shock in trauma patients. J Trauma. 1998;45:545–549. doi: 10.1097/00005373-199809000-00022. [DOI] [PubMed] [Google Scholar]
  • 6.Arikan AA YB, Mastrangelo MA, Tweardy DJ. Interleukin-6 treatment reverses apoptosis and blunts susceptibility to intraperitoneal bacterial challenge following hemorrhagic shock. Crit Care Med. 2006;34:771–777. doi: 10.1097/01.ccm.0000201901.30292.c2. [DOI] [PubMed] [Google Scholar]
  • 7.Meng ZH, Dyer K, Billiar TR, Tweardy DJ. Essential role for IL-6 in postresuscitation inflammation in hemorrhagic shock. Am J Physiol Cell Physiol. 2001;280:C343–351. doi: 10.1152/ajpcell.2001.280.2.C343. [DOI] [PubMed] [Google Scholar]
  • 8.Hierholzer C, Harbrecht B, Menezes JM, et al. Essential role of induced nitric oxide in the initiation of the inflammatory response after hemorrhagic shock. J Exp Med. 1998;187:917–928. doi: 10.1084/jem.187.6.917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Maritano D, Sugrue ML, Tininini S, et al. The STAT3 isoforms alpha and beta have unique and specific functions. Nat Immunol. 2004;5:401–409. doi: 10.1038/ni1052. [DOI] [PubMed] [Google Scholar]
  • 10.Hierholzer C HB, Menezes JM, Kane J, Macmicking J, Nathan CF, Peitzman AB, Billiar TR, Tweardy DJ. Essential role of induced nitric oxide in the initiation of the inflammatory response after hemorrhagic shock. J Exp Med. 1998;187:917–928. doi: 10.1084/jem.187.6.917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ono M YB, Hardison EG, Mastrangelo MA, Tweardy DJ. Increased susceptibility to liver injury after hemorrhagic shock in rats chronically fed ethanol: role of nuclear factor kappa-B, interlekin-6, and granulocyte colony-stimulating factor. Shock. 2004;21:519–525. doi: 10.1097/01.shk.0000126905.75237.07. [DOI] [PubMed] [Google Scholar]
  • 12.Ono M, Yu B, Hardison EG, Mastrangelo MA, Tweardy DJ. Increased susceptibility to liver injury after hemorrhagic shock in rats chronically fed ethanol: role of nuclear factor-kappa B, interleukin-6, and granulocyte colony-stimulating factor. Shock. 2004;21:519–525. doi: 10.1097/01.shk.0000126905.75237.07. [DOI] [PubMed] [Google Scholar]
  • 13.Jing N, Li Y, Xiong W, Sha W, Jing L, Tweardy DJ. G-quartet oligonucleotides: a new class of signal transducer and activator of transcription 3 inhibitors that suppresses growth of prostate and breast tumors through induction of apoptosis. Cancer Res. 2004;64:6603–6609. doi: 10.1158/0008-5472.CAN-03-4041. [DOI] [PubMed] [Google Scholar]
  • 14.Jing N, Tweardy DJ. Targeting Stat3 in cancer therapy. Anticancer Drugs. 2005;16:601–607. doi: 10.1097/00001813-200507000-00002. [DOI] [PubMed] [Google Scholar]
  • 15.SE SPaA. Mitochondria-associated apoptotic signalling in denervated rat skeletal muscle. J Physiol. 2005;565:309–323. doi: 10.1113/jphysiol.2004.081083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wu JY, Feng L, Park HT, et al. The neuronal repellent Slit inhibits leukocyte chemotaxis induced by chemotactic factors. Nature. 2001;410:948–952. doi: 10.1038/35073616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wang W, Goswami S, Lapidus K, et al. Identification and testing of a gene expression signature of invasive carcinoma cells within primary mammary tumors. Cancer Res. 2004;64:8585–8594. doi: 10.1158/0008-5472.CAN-04-1136. [DOI] [PubMed] [Google Scholar]
  • 18.Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res. 2001;125:279–284. doi: 10.1016/s0166-4328(01)00297-2. [DOI] [PubMed] [Google Scholar]
  • 19.Ishihara K, Hirano T. Molecular basis of the cell specificity of cytokine action. Biochim Biophys Acta. 2002;1592:281–296. doi: 10.1016/s0167-4889(02)00321-x. [DOI] [PubMed] [Google Scholar]
  • 20.Shirogane T, Fukada T, Muller JM, Shima DT, Hibi M, Hirano T. Synergistic roles for Pim-1 and c-Myc in STAT3-mediated cell cycle progression and antiapoptosis. Immunity. 1999;11:709–719. doi: 10.1016/s1074-7613(00)80145-4. [DOI] [PubMed] [Google Scholar]
  • 21.Brazil DP, Park J, Hemmings BA. PKB binding proteins. Getting in on the Akt. Cell. 2002;111:293–303. doi: 10.1016/s0092-8674(02)01083-8. [DOI] [PubMed] [Google Scholar]
  • 22.Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell. 1999;96:857–868. doi: 10.1016/s0092-8674(00)80595-4. [DOI] [PubMed] [Google Scholar]
  • 23.Kops GJ, de Ruiter ND, De Vries-Smits AM, Powell DR, Bos JL, Burgering BM. Direct control of the Forkhead transcription factor AFX by protein kinase B. Nature. 1999;398:630–634. doi: 10.1038/19328. [DOI] [PubMed] [Google Scholar]
  • 24.Wang XJ, Liefer KM, Tsai S, O'Malley BW, Roop DR. Development of geneswitch transgenic mice that inducibly express transforming growth factor beta1 in the epidermis. Proc Natl Acad Sci USA. 1999;96:8483–8488. doi: 10.1073/pnas.96.15.8483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Nicholson KM, Anderson NG. The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal. 2002;14:381–395. doi: 10.1016/s0898-6568(01)00271-6. [DOI] [PubMed] [Google Scholar]
  • 26.Jing N, Li Y, Xu X, et al. Targeting Stat3 with G-quartet oligodeoxynucleotides in human cancer cells. DNA Cell Biol. 2003;22:685–696. doi: 10.1089/104454903770946665. [DOI] [PubMed] [Google Scholar]
  • 27.Jing N, Sha W, Li Y, Xiong W, Tweardy DJ. Rational drug design of G-quartet DNA as anti-cancer agents. Curr Pharm Des. 2005;11:2841–2854. doi: 10.2174/1381612054546761. [DOI] [PubMed] [Google Scholar]
  • 28.Huang Y, Qiu J, Dong S, et al. Stat3 isoforms, alpha and beta, demonstrate distinct intracellular dynamics with prolonged nuclear retention of Stat3beta mapping to its unique C-terminal end. J Biol Chem. 2007;282:34958–67. doi: 10.1074/jbc.M704548200. [DOI] [PubMed] [Google Scholar]
  • 29.Bromberg JF WM, Devgan G, Zhao Y, Pestell RG, Albanese C, Darnell JE. Stat3 as an oncogene. Cell. 1999;98:295–303. doi: 10.1016/s0092-8674(00)81959-5. [DOI] [PubMed] [Google Scholar]
  • 30.Turkson J, Bowman T, Garcia R, Caldenhoven R, Degroot RP, Jove R. Stat3 activation by Src induces specific gene regulation and is required for cell transformation. Molecular and Cellular Biology. 1998 May;18:2545–2552. doi: 10.1128/mcb.18.5.2545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Takeda K, Kishimoto T, Akira S. Signal transducer and activator of transcription protein (STAT): its relation to Th1/Th2-mediated diseases [editorial] Nutrition. 1997;13:987–8. doi: 10.1016/s0899-9007(97)00343-2. [DOI] [PubMed] [Google Scholar]
  • 32.Chen CL HF, Lin J. Systemic evaluation of total Stat3 and Stat3 tyrosine phosphorylation in normal human tissues. Exp Mol Pathol. 2006;80:295–305. doi: 10.1016/j.yexmp.2005.11.003. [DOI] [PubMed] [Google Scholar]
  • 33.Levy DaLC. What does Stat3 do? J Clin Invest. 2002;109:1143–1148. doi: 10.1172/JCI15650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Real P SA, Dejuan A, Segovia J, Lopez-Vega J, Fernandez-Luna J. Resistance to chemotherapy via Stat3-dependent overexpression of Bcl-2 in metastatic breast cancer cells. Oncogene. 2002;21:7611–7618. doi: 10.1038/sj.onc.1206004. [DOI] [PubMed] [Google Scholar]
  • 35.Leslie K LC, Devgan G, Azare J, Berishaj M, Gerald W, Kim YB, Paz K, et al. Cyclin D1 is transcriptionally regulated by and required for transformation by activated signal transducer and activator of transcription 3. Cancer Res. 2006;66:2544–2552. doi: 10.1158/0008-5472.CAN-05-2203. [DOI] [PubMed] [Google Scholar]
  • 36.Catlett-Falcone R, Landowski TH, Oshiro MM, et al. Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells. Immunity. 1999;10:105–115. doi: 10.1016/s1074-7613(00)80011-4. [DOI] [PubMed] [Google Scholar]
  • 37.Gritsko T WA, Tucson J, Kaneko S, et al. Persistent activation of Stat3 induces survivin gene expression and confers resistance to apoptosis in human breast cancer cells. Clin Cancer Res. 2006;12:11–19. doi: 10.1158/1078-0432.CCR-04-1752. [DOI] [PubMed] [Google Scholar]
  • 38.Fukada T OT, Yoshida Y, Shirogane T, Nishida K, Nakajima K, Hibi M, Hirano T. Stat3 orchestrates contradictory signals in cytokine induced G1 to S cell cycle transition. The EMBO J. 1996;17:6670–6677. doi: 10.1093/emboj/17.22.6670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Taub R. Hepatoprotection via the IL-6/Stat3 pathway. J Clin Invest. 2003;112:978–980. doi: 10.1172/JCI19974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kobelt F, Schreck U, Henrich HA. Involvement of liver in the decompensation of hemorrhagic shock. Shock. 1994;2:281–288. doi: 10.1097/00024382-199410000-00009. [DOI] [PubMed] [Google Scholar]
  • 41.Sundar SV, Li YY, Rollwagen FM, Maheshwari RK. Hemorrhagic shock induces differential gene expression and apoptosis in mouse liver. Biochem Biophys Res Commun. 2005;332:688–96. doi: 10.1016/j.bbrc.2005.05.008. [DOI] [PubMed] [Google Scholar]
  • 42.Ayuste EC, Chen H, Koustova E, et al. Hepatic and pulmonary apoptosis after hemorrhagic shock in swine can be reduced through modifications of conventional Ringer's solution. J Trauma. 2006;60:52–63. doi: 10.1097/01.ta.0000200156.05397.0b. [DOI] [PubMed] [Google Scholar]
  • 43.Rudiger HA GR, Claiven PA. Liver ischemia: apoptosis as a central mechanism of injury. J Invest Surg. 2003;16:149–159. [PubMed] [Google Scholar]
  • 44.Kovalovich K, DeAngelis RA, Li W, Furth EE, Ciliberto G, Taub R. Increased toxin-induced liver injury and fibrosis in interleukin-6-deficient mice. Hepatology. 2000;31:149–159. doi: 10.1002/hep.510310123. [DOI] [PubMed] [Google Scholar]
  • 45.Kovalovich K, Li W, DeAngelis R, Greenbaum LE, Ciliberto G, Taub R. Interleukin-6 protects against Fas-mediated death by establishing a critical level of antiapoptotic hepatic proteins FLIP, Bcl-2, and Bcl-xL. J Biol Chem. 2001;276:26605–26613. doi: 10.1074/jbc.M100740200. [DOI] [PubMed] [Google Scholar]
  • 46.Hong F, Jaruga B, Kim WH, et al. Opposing roles of STAT1 and STAT3 in T cellmediated hepatitis: regulation by SOCS. J Clin Invest. 2002;110:1503–1513. doi: 10.1172/JCI15841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hong F, Kim WH, Tian Z, et al. Elevated interleukin-6 during ethanol consumption acts as a potential endogenous protective cytokine against ethanol-induced apoptosis in the liver: involvement of induction of Bcl-2 and Bcl-x(L) proteins. Oncogene. 2002;21:32–43. doi: 10.1038/sj.onc.1205016. [DOI] [PubMed] [Google Scholar]
  • 48.Lee CK, Raz R, Gimeno R, et al. STAT3 is a negative regulator of granulopoiesis but is not required for G-CSF-dependent differentiation. Immunity. 2002;17:63–72. doi: 10.1016/s1074-7613(02)00336-9. [DOI] [PubMed] [Google Scholar]
  • 49.Furukawa Y, Kawasoe T, Daigo Y, et al. Isolation of a novel human gene, ARHGAP9, encoding a rho-GTPase activating protein. Biochem Biophys Res Commun. 2001;284:643–649. doi: 10.1006/bbrc.2001.5022. [DOI] [PubMed] [Google Scholar]
  • 50.Kokkinakis DM. Methionine-stress: a pleiotropic approach in enhancing the efficacy of chemotherapy. Cancer Lett. 2006;233:195–207. doi: 10.1016/j.canlet.2005.02.034. [DOI] [PubMed] [Google Scholar]
  • 51.Kumar A, Kumar A, Michael P, et al. Human serum from patients with septic shock activates transcription factors STAT1, IRF1, and NF-kappaB and induces apoptosis in human cardiac myocytes. J Biol Chem. 2005;280:42619–42626. doi: 10.1074/jbc.M508416200. [DOI] [PubMed] [Google Scholar]
  • 52.Kataoka K, Hasegawa K, Sawamura T, et al. LOX-1 pathway affects the extent of myocardial ischemia-reperfusion injury. Biochem Biophys Res Commun. 2003;300:656–660. doi: 10.1016/s0006-291x(02)02905-4. [DOI] [PubMed] [Google Scholar]
  • 53.Zerbini LF, Libermann TA. Life and death in cancer. GADD45 alpha and gamma are critical regulators of NF-kappaB mediated escape from programmed cell death. CellCycle. 2005;4:18–20. doi: 10.4161/cc.4.1.1363. [DOI] [PubMed] [Google Scholar]
  • 54.Chakraborty A, Tweardy DJ. Stat3 and G-CS induced myeloid differentiation. Leukemia and Lymphoma. 1998;30:433–442. doi: 10.3109/10428199809057555. [DOI] [PubMed] [Google Scholar]
  • 55.Chakraborty A, White SM, Schaefer TS, Ball ED, Dyer KF, Tweardy DJ. Granulocyte colony-stimulating factor activation of Stat3 alpha and Stat3 beta in immature normal and leukemic human myeloid cells. Blood. 1996;88:2442–2449. [PubMed] [Google Scholar]

Articles from International Journal of Clinical and Experimental Medicine are provided here courtesy of e-Century Publishing Corporation

RESOURCES