Skip to main content
Physiological Reports logoLink to Physiological Reports
. 2013 Sep 17;1(4):e00073. doi: 10.1002/phy2.73

Candidate genes for limiting cholestatic intestinal injury identified by gene expression profiling

Samuel M Alaish 1, Jennifer Timmons 1, Alexis Smith 1, Marguerite S Buzza 2, Ebony Murphy 1, Aiping Zhao 3, Yezhou Sun 4, Douglas J Turner 5, Terez Shea-Donahue 2,3, Toni M Antalis 1,2, Alan Cross 3, Susan G Dorsey 6
PMCID: PMC3808870  NIHMSID: NIHMS518056  PMID: 24179676

Abstract

The lack of bile flow from the liver into the intestine can have devastating complications including hepatic failure, sepsis, and even death. This pathologic condition known as cholestasis can result from etiologies as diverse as total parenteral nutrition (TPN), hepatitis, and pancreatic cancer. The intestinal injury associated with cholestasis has been shown to result in decreased intestinal resistance, increased bacterial translocation, and increased endotoxemia. Anecdotal clinical evidence suggests a genetic predisposition to exaggerated injury. Recent animal research on two different strains of inbred mice demonstrating different rates of bacterial translocation with different mortality rates supports this premise. In this study, a microarray analysis of intestinal tissue following common bile duct ligation (CBDL) performed under general anesthesia on these same two strains of inbred mice was done with the goal of identifying the potential molecular mechanistic pathways responsible. Over 500 genes were increased more than 2.0-fold following CBDL. The most promising candidate genes included major urinary proteins (MUPs), serine protease-1-inhibitor (Serpina1a), and lipocalin-2 (LCN-2). Quantitative polymerase chain reaction (qPCR) validated the microarray results for these candidate genes. In an in vitro experiment using differentiated intestinal epithelial cells, inhibition of MUP-1 by siRNA resulted in increased intestinal epithelial cell permeability. Diverse novel mechanisms involving the growth hormone pathway, the acute phase response, and the innate immune response are thus potential avenues for limiting cholestatic intestinal injury. Changes in gene expression were at times found to be not only due to the CBDL but also due to the murine strain. Should further studies in cholestatic patients demonstrate interindividual variability similar to what we have shown in mice, then a “personalized medicine” approach to cholestatic patients may become possible.

Keywords: Cholestasis, growth hormone, intestine, lipocalin, microarray

Introduction

Cholestasis, defined as little or no bile flow from the liver into the intestine, is a complex pathologic condition that can develop from either functional etiologies, such as hepatic parenchymal disease secondary to hepatitis, or mechanical etiologies, such as an obstructing pancreatic cancer or biliary stricture. In the pediatric population, cholestasis resulting from prolonged parenteral nutrition is by far the most common etiology. Cholestatic injury has not only a hepatic component but also an intestinal one. Failure of the intestinal barrier with decreased intestinal resistance, increased bacterial translocation, and increased episodes of sepsis has been well described (Campillo et al. 1999; Pascual et al. 2003; Frances et al. 2004); however, the exact mechanisms remain poorly understood.

Common bile duct ligation (CBDL) is a standard model of cholestasis in the literature (Georgiev et al. 2008). CBDL in mice leads to both hepatic and intestinal injuries which are precisely interrelated. We have previously found differences in the systemic inflammatory responses and outcome following CBDL between two inbred mouse strains, C57BL/6J (B6) and A/J, suggesting a genetic contribution (Alaish et al. 2005). In particular, B6 mice were significantly more likely to develop ascites following 1 week of CBDL (Alaish et al. 2005). In concordance with this observation, the frequency of mortality after CBDL was significantly higher in B6 mice compared to A/J mice on days following CBDL (Alaish et al. 2005). Interestingly, although both strains demonstrated markedly elevated plasma liver function tests following CBDL, no difference was noted in liver histology between the two ligated strains. More recently, our laboratory has shown decreased intestinal resistance and increased bacterial translocation following CBDL in these same two strains of inbred mice. Furthermore, we found genetic variation in the intestinal resistance and bacterial translocation rates, which correlated with mortality following CBDL in different strains of inbred mice (Alaish et al. 2013). Further analysis implicated an IFN-γ-mediated apoptotic-independent mechanism of tight junction disruption, which has been well described in vitro (Madara and Stafford 1989; Marano et al. 1998; Youakim and Ahdieh 1999; Bruewer et al. 2003; Clayburgh et al. 2004), as a mechanism possibly responsible for the genetic variation. Nevertheless, the 2.5-fold changes in IFN-γ gene expression following CBDL, albeit significant, did not seem monumental enough to fully explain the striking genetic influence on mortality following CBDL in the mice. In order to uncover other potential mechanisms including novel pathways, we embarked on a whole-genome microarray analysis of jejunal tissue in these two different strains of inbred mice following either a sham operation or CBDL. The differentially expressed genes reported here constitute a resource of candidate genes for roles in cholestatic intestinal injury.

Material and Methods

Animals

Male A/J and C57BL/6J (B6) mice (8 weeks old) were obtained from the Jackson Laboratory (Bar Harbor, ME) and maintained in identical environmental conditions in a pathogen-free animal facility with 12-h light–dark cycles. All mice weighed 18–25 g at the time of operation. Matriptase (St14) hypomorphic C57BL/6J mice (List et al. 2007) were bred in the Antalis laboratory. Animal studies were conducted according to protocols reviewed and approved by the University of Maryland School of Medicine Institutional Animal Care and Use Committee and adhered to guidelines promulgated by the National Institutes of Health. In accordance with these guidelines, we used the minimum number of animals to meet the rigor necessary for this series of experiments.

Experimental design

CBDL operative procedure

Mice were anesthetized by inhaled isoflurane anesthesia. The abdomen was clipped and then prepared in sterile fashion with 70% ethyl-ethanol followed by betadine. A transverse upper abdominal incision was performed. The CBD was dissected away from the portal vein and was ligated near its junction with the duodenum using aneurysm clips engineered with a precisely standardized opening/closing mechanism. The abdominal wall was then closed in a two-layer fashion using absorbable sutures. Sham-operated mice were treated identically but without dissection or ligation of the CBD. Postoperatively, animals were resuscitated with warmed subcutaneous injections of saline (1 mL) to replace losses. Mice were returned to clean cages where food and water were provided ad libitum. Buprenorphine, 0.05–0.1 mg/kg was given subcutaneously at the time of surgery and then every 8–12 h to treat postoperative pain for 48–72 h.

RNA extraction

Seven days following the surgery, the mice underwent deep general anesthesia and euthanasia by thoracotomy and cardiac exsanguination. Postoperative day 7 was chosen because this time point corresponded to our earlier finding of decreased intestinal transepithelial electrical resistance (TEER) after CBDL (Alaish et al. 2013). In addition, this time point exhibited differences in TEER based on the genetic background of the mouse (Alaish et al. 2013). These TEER findings were found in both the jejunum and ileum and correlated with differences in bacterial translocation and mortality. Further studies on jejunal tissue demonstrated differences in tight junction protein expression between CBDL and sham animals and between the strains (Alaish et al. 2013). Therefore, in this study, we chose jejunum once again; the intestinal tissue was harvested under sterile conditions. RNA extraction and purification were performed as we have previously described (Dorsey et al. 2009).

Microarray data analysis

Microarray expression profiling was performed according to the manufacturer protocols (Affymetrix, Santa Clara, CA). Briefly, total RNA was used to prepare biotinylated cRNA, followed by fragmentation and hybridization to Affymetrix arrays (Genechip Mouse 430 2.0; Affymetrix, Santa Clara, CA). The arrays were incubated for approximately 16 h, washed, stained, and scanned per Affymetrix. Differential gene expression through microarray was then performed. We utilized.cel files generated from Affymetrix profiling process for analysis. Arrays were normalized by GCRMA method implemented in gcrma R package (Bioconductor, an open source collection of software packages). Differential expression analysis was performed using limma R package (Bioconductor). First, a linear model was fitted to expression data for each gene. Empirical Bayes method was then used to assess differential expression between two conditions. A cutoff of FDR less than 0.05 was used to select significant probes. A complete data set has been submitted to the NCBI Gene Expression Omnibus (NCBI GEO #GSE47099 and NCBI Tracking System #16793295).

qPCR verification of promising candidate genes

The identification of significant changes in expression of promising candidate genes (major urinary proteins [MUPs], serine protease-1-inhibitor [Serpina1a] and lipocalin-2 [LCN2]) through microarray analysis was validated using a quantitative polymerase chain reaction (qPCR) technique. Total RNA was isolated from homogenized jejunal samples that were stored in TRIzol (Invitrogen, Grand Island, NY). The total RNA was isolated from TRIzol samples as per the manufacturer's instructions. The pellet was allowed to air dry, and the total RNA was resuspended in an appropriate volume of RNAse-free water. RNA concentrations were calculated using a NanoDrop 1000 spectrophotometer (Thermo Scientific, Waltham, MA). Single-stranded cDNA was synthesized from 2 μg of total RNA using random hexamer primer and the First-Strand cDNA Synthesis Kit (MBI Fermentas, Hanover, MD). The specific primer sequences were designed using Beacon Designer 7.0 (Premier Biosoft International, Palo Alto, CA) and synthesized by the University of Maryland School of Medicine Biopolymer/Genomics Core. qPCR reactions were set up using iQ SYBR Green Supermix (Bio-Rad, Hercules, CA) in a total volume of 25 μL. Amplification conditions were as follows: 95°C for 3 min, 50 cycles of 95°C for 15 sec, 60°C for 15 sec, and 72°C for 20 sec. All reactions were performed using Bio-Rad iCycler instrumentation and software. All samples were normalized with 18s rRNA housekeeping gene levels with subsequent calculation of fold change in mRNA expression. Analysis was carried out in GraphPad Prism5 (San Diego, CA, USA).

Mortality following CBDL in matriptase hypomorphic B6 mice

We conducted a mortality study following CBDL in wild-type C57BL/6J mice (n = 8) and matriptase (St14) hypomorphic C57BL/6J mice (n = 11). Sham-operated mice of each strain served as controls.

Cells

Cdx2-intestinal epitheial cells (Cdx2-IEC), a transformed rat crypt IEC-6 cell line which maintains a stable differentiated phenotype upon future passages, were obtained from Dr. J.-Y. Wang (University of Maryland, Baltimore, MD). Cdx2-IEC cells were maintained at 37°C in a humidified incubator with 10% CO2 in DMEM containing 5% (v:v) fetal bovine serum (FBS), 0.5% (v:v) ITS + liquid media supplement, 0.1 million units/L penicillin, 100 mg/L streptomycin, and 4 mmol/L sopropylthio-β-d-galactoside, which served as an inducer.

Transfection of Cdx2-IEC cells with MUP-1 siRNA and FITC-dextran permeability assay

Following their sixth passage, Cdx2-IEC cells were transfected with either MUP-1 siRNA (Thermo Scientific, Inc.) or Acell Control Non-Targeting siRNA (Thermo Scientific, Inc.) as described previously (Rao et al. 2006). The siRNAs used were as follows: 80 nmol/L Control siRNA; and 20 nmol/L, 40 nmol/L, and 60 nmol/L MUP-1 siRNA. Silencing of MUP-1 in the cells was confirmed by Western blot analysis using MUP (F-3) mouse monoclonal antibody (Santa Cruz Biotechnology, Inc., Paso Robles, CA, USA). Six-well transwell plates with 12-mm-diameter inserts (Costar 3407; Corning, Inc., Kennebunk, ME, USA) were used to perform the permeability studies following the transfection. The cells were incubated on the inserts with control media for 24 h to allow proper attachment to the membrane prior to dextran administration. After 24 h, TEER was measured in both control and transfected Cdx2-IEC cells for the formation of the monolayer as described previously (El Asmar et al. 2002). The media were removed. 4-kDa FITC-dextran in control media was placed onto the apical side (top chamber); control media alone was placed on the basolateral side. The TEER was monitored for 2 h and 100-μL aliquots of the basolateral medium were collected after each 30-min time period. A sample from the top compartment at the time of the last sampling of the bottom compartment was used to normalize the samples to account for possible differences in the total fluorescence added at the beginning of the experiment. Fluorescence of the samples was quantified in a multiplate fluorescence reader in black 96-well plates; the excitation wavelength was 485 nm and emission wavelength was 538 nm.

Statistical analysis

The microarray data (reported as percent change) were analyzed using repeated measures analyses of variance (ANOVA) with false discovery rate correction to control multiple testing errors. Post hoc testing was done using Tukey's honestly significant difference (HSD). qPCR data were analyzed using ANOVA with the Bonferroni posttest. Graph Pad Prism 5 software was used. For the matriptase hypomorphic B6 mouse CBDL experiment, a Kaplan–Meier survival curve was generated with P < 0.05 considered as significant.

Results

Microarray data

As shown in Figure 1A, more than 500 genes were significantly differentially regulated in the CBDL mice compared with sham. When examining changes across strains, although there are shared genes, there are a significant number of differentially regulated genes that are unique to each strain (Fig. 1B). Table 1 shows the number of genes in each category. The first two lines of data in Table 1 demonstrate that many more genes are differentially expressed in A/J mice as compared to B6 mice following CBDL (582 vs. 137). The last two lines of data in Table 1 demonstrate that there are more genes undergoing expression changes following CBDL compared to sham (882 vs. 766). In Figure 1C, the heat map shows clustering of all differentially expressed genes by genotype. A list of all differentially expressed genes can be found in Table A1. In Figure 1D, the top 53 significantly regulated genes across the two strains are shown in a separate heat map (For both heat maps, red = upregulated genes; green = downregulated genes). Note the disparate gene expressions in the two strains.

Figure 1.

Figure 1

Venn diagram and heat map depicting the number of statistically significant differentially expressed genes by experimental condition and across murine strains. The venn diagram in (A) depicts the number of genes that are differentially expressed in the surgical group versus sham. In (B) we show the number of differentially expressed genes across two strains, A/J and B6. (C) The heat map shows all differentially expressed genes between strains. (D) A heat map which shows the top 53 significantly regulated genes across two strains. For both heat maps, red = upregulated genes; green = downregulated genes.

Table 1.

Differentially expressed genes with FDR < 0.05

Total Up Down
A/J CBDL versus sham 582 404 178
B6 CBDL versus sham 137 76 61
Sham A/J versus B6 766 276 490
CBDL A/J versus B6 882 291 591

We next examined significantly enriched canonical signaling pathways. First, we evaluated those that were unique within a strain in the CBDL condition compared with sham. As shown in Figure 2A, coagulation pathways were highly upregulated in A/J CBDL mice compared with sham. In contrast, coagulation pathways were not significantly regulated in B6 CBDL versus sham (Fig. 2B). In Figure 2C, we show that there are a number of signaling pathways that are differentially regulated in A/J CBDL compared with B6 CBDL, demonstrating differential pathway activation in CBDL across these two inbred strains of mice.

Figure 2.

Figure 2

Significantly enriched canonical signaling pathways from differentially expressed gene sets. As denoted by the blue arrows in (A), coagulation pathways were highly upregulated in CBDL AJ mice compared with sham. In contrast, coagulation pathways were not significantly regulated in B6 CBDL versus sham (B). In (C), we show that there are a number of signaling pathways that are differentially regulated in AJ CBDL compared with B6 CBDL, demonstrating differential pathway activation in CBDL across these two inbred strains of mice.

qPCR verifies microarray results of candidate genes

The following genes struck us as particularly promising as candidate genes to limit cholestatic injury: MUPs, Serpina1a, and LCN2. They all had significant changes in gene expression following CBDL. MUPs and Serpina1a also had disparate strain expressions which could account for the phenotypic differences we see following ligation. LCN2 was chosen because it is known to play a protective role in the intestinal barrier (Berger et al. 2006). Although we did not see strain differences in LCN2, we did find striking differences between the sham and CBDL mice, which we believe is an important finding. This study not only illustrates strain differences following CBDL but also serves to illustrate intestinal gene expression changes following CBDL independent of strain. qPCR did, indeed, verify our results for MUPs, Serpina1a, and LCN2. (Fig. 3A–C).

Figure 3.

Figure 3

Serpina 1a, MUPs, and LCN2 gene expression changes determined by qPCR are consistent with microarray findings. (A) demonstrates a marked increase in Serpina 1a mRNA following CBDL in both strains. Also, there is a strain difference with increased Serpina 1a mRNA expression in B6 as compared to A/J mice. (B) demonstrates marked strain differences in MUP mRNA expression in the sham animals with disparate responses to CBDL. A/J sham MUP mRNA expression is low and expression increases following CBDL; whereas, in the B6 strain, MUP mRNA expression is very high in the sham group but falls in the CBDL group. (C) shows low expression of LCN2 mRNA in sham animals with marked increases following CBDL in both strains.

Matriptase (St14) hypomorphic B6 mice do not have significantly increased mortality following CBDL compared to wild-type B6 mice

Serpina1a is a serine protease inhibitor, suggesting that decreased intestinal serine protease activity could contribute to the decreased intestinal resistance associated with CBDL. Matriptase is a serine protease whose loss results in decreased intestinal resistance as measured by decreased intestinal TEER (Buzza et al. 2010). We hypothesized that when coupled with the loss of matriptase, the increase in Serpina1a which follows CBDL would result in such a large drop in intestinal resistance that mortality would increase. Although there appeared to be a trend toward increased mortality in the matriptase (St14) hypomorphic B6 mice early on following CBDL, this difference never became significant and did not persist, as can be seen in the Kaplan–Meier Survival Curve (Fig. 4).

Figure 4.

Figure 4

Kaplan–Meier survival curve for matriptase hypomorphic B6 and wild-type B6 mice following CBDL. The early trend toward increased mortality in the matriptase hypomorphic B6 mice is not statistically significant.

MUP-1 siRNA transfection increases permeability in Cdx2-IEC cells

Growth hormone treatment has beneficial effects on the intestine by normalizing intestinal permeability (Liu et al. 2003). We hypothesize that increased expression of MUPs by growth hormone activation (Kuhn et al. 1984) could contribute to this beneficial effect. We inhibited MUP-1 protein expression using siRNA technology. Following confirmation of inhibition of MUP-1 protein expression (Fig. 5), we performed a FITC-dextran permeability assay on Cdx2-IEC cells alone, Cdx2-IEC cells exposed to calcium-free media, Cdx2-IEC cells exposed to control siRNA, and Cdx2-IEC cells exposed to two different concentrations of MUP-1 siRNA (Fig. 6). As expected, cells exposed to calcium-free media resulted in a marked increase in cell permeability compared to Cdx2-IEC cells in control media (*P < 0.04). Cells treated with Control siRNA at 80 nmol/L and MUP-1 siRNA at 40 nmol/L concentrations were similar to untreated cells; whereas, cells treated with MUP-1 siRNA at 60 nmol/L concentration had significantly increased cell permeability compared to Cdx2-IEC cells in control media (**P < 0.0002). Indeed, permeability of cells treated with MUP-1 siRNA at 60 nmol/L concentration was similar to cells in calcium-free media.

Figure 5.

Figure 5

Western blot demonstrating silencing of MUP-1. Following their sixth passage, Cdx2-IEC cells were transfected with either MUP-1 siRNA (Thermo Scientific, Inc.) or Acell Control Non-Targeting siRNA (Thermo Scientific, Inc.). Western blot analysis was performed using MUP (F-3) mouse monoclonal antibody (Santa Cruz Biotechnology, Inc.).

Figure 6.

Figure 6

FITC-dextran permeability assay in Cdx2-IEC cells. Cells exposed to calcium-free (Ca-free) media resulted in a marked increase in cell permeability compared to Cdx2-IEC cells in control media (*P < 0.04). Cells treated with Control siRNA at 80 nmol/L and MUP-1 siRNA at 40 nmol/L concentrations were similar to untreated cells; whereas, cells treated with MUP-1 siRNA at 60 nmol/L concentration had significantly increased cell permeability compared to Cdx2-IEC cells in control media (**P < 0.0002). Indeed, permeability of cells treated with MUP-1 siRNA at 60 nmol/L concentration was similar to cells in calcium-free media.

Discussion

Cholestasis can arise from a multitude of conditions in both the adult and pediatric population. In broad terms, cholestasis may result from biliary tract obstruction or hepatic parenchymal disease. Causes of biliary tract obstruction in the adult include pancreatic carcinoma, duodenal carcinoma and cholangiocarcinoma, benign biliary strictures, and choledochal cyst; whereas, biliary atresia and choledochal cyst are seen in infants and children. Causes of hepatic parenchymal disease in both age groups include hepatitis, total parenteral nutrition, sepsis, and other more rare cholestatic syndromes. In the neonatal population, cholestasis is commonly associated with a variety of intestinal pathologies, including necrotizing enterocolitis, intestinal atresias, midgut volvulus, gastroschisis, and Hirschsprung disease. In these conditions, a baby may not be able to tolerate much or any nutrition enterally. Total parenteral (intravenous) nutrition is required. As mentioned earlier, this type of nutrition causes cholestasis and the lack of enteral nutrition worsens the cholestasis; whereas, initiation of enteral nutrition and discontinuation of parenteral nutrition result in improvement of cholestatic liver dysfunction (Javid et al. 2005).

The goal of this study was to use microarray technology to generate candidate genes from both existing and novel pathways which play a role in the development of intestinal injury following cholestasis as modeled by CBDL. Using two different inbred strains of mice, A/J and C57Bl/6J, the design of this study had the benefit of not only discovering strain-independent but also strain-dependent changes in intestinal gene expression following cholestasis. More than 500 genes were increased by more than 2.0-fold following CBDL, and the following were identified as promising candidate genes for further study: MUPs, Serpina1a, and LCN-2.

Major urinary proteins (MUPs) comprise the lipocalin superfamily of lipophilic ligand carriers which, until recently, were thought to participate exclusively in pheromone function. In a seminal article from 2009, MUP1 was shown to be a regulator for glucose and lipid metabolism, in addition to its action as a pheromone ligand to mediate chemical signaling in mice (Zhou et al. 2009). MUP production is known to be increased by testosterone, thyroid hormone, and growth hormone (GH) (Kuhn et al. 1984). The latter hormone is well known to play a significant role in intestinal healing following injury. Increased growth of the small bowel mucosa has been demonstrated in mice overexpressing bovine growth hormone (Ulshen et al. 1993). Moreover, recombinant growth hormone significantly attenuated intestinal mucosal injuries and bacterial translocation in septic rats (Yi et al. 2007), possibly through a mechanism involving the rhGH inhibition of apoptosis of intestinal mucosa cells. Endotoxemia was also reduced after GH administration in jaundiced rats (Scopa et al. 2000). Some insight into the mechanism(s) by which GH exerts it effects have been elucidated in animal models. Prophylactic treatment with growth hormone promoted IgA secretion by B lymphocytes in the plasma and in the intestine in stressed postoperative rats (Ding et al. 2004). Similar effects have been seen in patients. GH was found to attenuate the depression in cellular immunity following surgical stress in a randomized, double-blind, controlled trial of 20 patients undergoing abdominal surgery (Liu et al. 2003). In addition, GH contributes to intestinal adaptation and has been documented to enable both adult and pediatric short gut syndrome patients to graduate from total parenteral nutrition (TPN) supplementation (Byrne et al. 1995; Velasco et al. 1999; Weiming et al. 2004). Growth hormone was also shown to reduce the increase in intestinal permeability seen following abdominal surgery (Liu et al. 2003). Consequently, when our microarray analysis demonstrated MUPs 1, 2, 3, 4, 7, 11, and 20 to be significantly increased following CBDL, we sought to explore this possible mechanism further. qPCR validated the microarray results and demonstrated a >200-fold increase in MUP expression in B6 sham mice compared to A/J sham mice. Most interestingly, the two strains of mice had different responses to CBDL. MUP expression increased >5-fold in A/J mice following CBDL; however, MUP expression in B6 mice decreased greatly following CBDL, such that there was no significant difference between two strains following CBDL. We speculate that GH rises in A/J mice following CBDL to increase MUP expression which, in turn, leads to increased intestinal resistance, strengthens the intestinal barrier, and helps limit injury; whereas, in B6 mice, GH reserves and MUP expression have been depleted and simply cannot keep up with the injury. Further work is needed to clarify this.

We documented baseline expression of MUP-1 in intestinal epithelial cells. Following transfection of these cells with MUP-1 siRNA at 60 nmol/L concentration, we noticed a marked increase in cell permeability. This finding implicates increased MUP expression leading to increased intestinal resistance as a mechanism potentially responsible for the beneficial effects of growth hormone therapy.

Serpina1a is a gene on Chromosome 14 that encodes alpha-1 antitrypsin, which is a type of serine protease inhibitor, serpin. Our microarray analysis demonstrated a markedly elevated level of Serpina1a gene expression in the B6 sham mice, as well as dramatic increases in Serpina1a gene expression following CBDL in both strains. qPCR confirmed these findings. B6 sham mice had approximately 50-fold increased levels of expression compared to A/J sham mice. Furthermore, both strains exhibited approximately 100-fold increased levels of expression following CBDL when compared to the A/J sham mice.

A novel mechanism was suggested by the work of Bacher et al. (1992). They found that in gastrointestinal cell lines, the formation of tight junctions involves cellular proteases which are susceptible to protease inhibitors. Moreover, the recent work by Buzza and colleagues (2010) showing that Matriptase, a membrane-type serine protease-1, regulated epithelial barrier formation and permeability in the intestine intrigued us. Using matriptase (St14) hypomorphic mice which express less than 1% of the gene product, Buzza and colleagues (2010) demonstrated these mice to have a leaky intestinal barrier with decreased TEER and increased paracellular permeability. This raised the hypothesis that matriptase (St14) hypomorphic mice would have a higher mortality following CBDL than wild-type B6 mice. The increased expression of Serpina1a, coupled with the decreased intestinal resistance that we had found following CBDL, suggested to us that matriptase (St14) hypomorphic mice with decreased intestinal resistance and leaky tight junctions would not tolerate CBDL and the resultant further inhibition of serine protease-1 as well as the wild-type B6 mice. To our surprise, we did not find a striking effect on mortality. This might be explained in part by the work of Beliveau et al. (2009) which showed that under in vitro conditions, Serpina1a was an impotent inhibitor of matriptase compared to other serpins, such as antithrombin III. Antithrombin III may very well inhibit matriptase in vivo; however, further experiments would be necessary to confirm this. Whether antithrombin III increases mortality in matriptase hypomorphic mice following CBDL might be confounded by the known salutary effects of antithrombin III in sepsis, including the attenuation of both hepatocyte apoptosis (Huang et al. 2010) and endotoxemia-induced healing impairment in the colon (Diller et al. 2009), as well as preserved mucosal thickness and villus height following CBDL (Caglikulekci et al. 2004). Nevertheless, our results suggest that matriptase does not play a significant role in the mortality following CBDL.

A potential mechanism behind the role of Serpina1a in the cholestatic intestine might be akin to that seen in studies of lung inflammation. Alpha-1 antitrypsin inhibits the enzyme, neutrophil elastase, which is released from neutrophils and macrophages during inflammation; neutrophil elastase destroys both bacteria and host tissue. Neutrophil elastase has also been shown to play a significant role in the pathogenesis of lung injury in pulmonary fibrosis (Yamanouchi et al. 1998). Moreover, alpha-1 antitrypsin was shown to induce hepatocyte growth factor (HGF) production by human lung fibroblasts and function as an anti-inflammatory and regenerative factor in addition to its role in protease inhibition (Kikuchi et al. 2000). HGF has previously been shown to increase intestinal epithelial cell mass and function in vivo (Kato et al. 1997) and also stimulate DNA content and protein content beyond the normal adaptive response following massive small intestinal resection (Kato et al. 1998). Our data suggest that increased expression of Serpina1a would increase HGF and provide a therapeutic approach to limit the intestinal injury following cholestasis, analogous to a recent study, in which alpha-1 antitrypsin therapy was shown to decrease intestinal permeability and ameliorate acute colitis and chronic ileitis in a murine model (Collins et al. 2013).

Lipocalin-2 is an iron-sequestering protein in the antibacterial innate immune response. Upon encountering invading bacteria, the Toll-like receptor 4 (TLR4) on immune cells stimulates the transcription, translation, and secretion of lipocalin-2 (Flo et al. 2004). It binds to siderophores secreted by pathogenic bacteria, including enterochelin secreted by E. coli, and prevents bacterial iron uptake (Flo et al. 2004). LCN2−/− mice have decreased survival following E. coli infection compared to wild-type mice (Berger et al. 2006). Neutrophils isolated from LCN2−/− mice showed significantly less bacteriostatic activity compared with wild-type controls. The bacteriostatic property of the wild-type neutrophils was abolished by the addition of exogenous iron, indicating that the main function of lipocalin-2 is to limit this essential element (Berger et al. 2006). Similarly, lipocalin-2 resistance confers an advantage to Salmonella enterica serotype Typhimurium for growth and survival in the inflamed intestine (Rafatellu et al. 2009). Our microarray analysis demonstrated significant increases in LCN2 following CBDL. qPCR confirmed this finding and is consistent with a strain-independent response to CBDL. LCN2 expression increased more than 20-fold and 40-fold, respectively, in A/J and B6 mice following CBDL. Increased LCN2 gene expression in the intestine following CBDL appears to be a mechanism whereby the host limits injury and mortality. This appears analogous to the increase in LCN2 gene expression coincident with an increase in epithelial cell apoptosis associated with intestinal adaptation following massive small bowel resection (Wildhaber et al. 2003). This is in agreement with earlier work by Devireddy et al. (2001) demonstrating lipocalin-2 to induce apoptosis in hematopoietic cells by an autocrine pathway. Thus, lipocalin-2 is a regulatory factor of intestinal growth.

In summary, our study has generated a resource of candidate genes for intestinal injury secondary to cholestasis. For Serpina1a, MUPs, and LCN2, some of the genes whose expression was most dramatically affected by CBDL, we confirmed the microarray results with qPCR. Novel mechanisms implicated by our results involve the growth hormone pathway, the acute phase response, and the innate immune response. More research is needed to further define these mechanisms; however, future therapeutic strategies might include: overexpression of Serpina1a and increased levels of HGF, upregulation of the GH pathway and increased MUP expression, and increased LCN2 expression to limit intestinal injury following cholestasis.

Appendix

Table A1.

List of all differentially expressed genes

Probe set ID Gene accession Gene symbol Gene description Cytoband AJ CBDL vs sham FC AJ CBDL vs sham FDR B6 CBDL vs sham FC B6 CBDL vs sham FDR
10566477 NM_017371 Hpx hemopexin 7 F1 37.4012 1.00E−04 4.7408 0.0435
10581605 NM_017370 Hp haptoglobin 8 D3|8 55.0 cM 34.152 3.00E−04 6.662 0.0445
10505438 NM_008768 Orm1 orosomucoid 1 4 B3|4 31.4 cM 18.2062 2.00E−04 4.3826 0.0404
10414192 NM_133653 Mat1a methionine adenosyltransferase I, alpha 14 C1 17.981 0.001 7.268 0.0306
10511886 NM_016668 // NM_016668 Bhmt // Bhmt betaine-homocysteine methyltransferase // betaine-homocysteine methyltransferase 13 D1 // 13 D1 17.9485 0.0014 10.4211 0.0197
10457114 NM_016668 Bhmt betaine-homocysteine methyltransferase 13 D1 15.6549 5.00E−04 8.3998 0.0109
10578352 NM_145594 Fgl1 fibrinogen-like protein 1 8 A4 13.8891 1.00E−04 4.1148 0.0178
10575349 NM_146214 Tat tyrosine aminotransferase 8 D3 12.4752 3.00E−04 4.323 0.0294
10449452 NM_010220 Fkbp5 FK506 binding protein 5 17 A3.3|17 13.0 cM 11.8521 0.0015 6.359 0.0294
10490989 NM_001042611 Cp ceruloplasmin 3 D 11.1162 0 3.3852 0.0109
10481627 NM_008491 Lcn2 lipocalin 2 2 A3|2 27.0 cM 9.7186 8.00E−04 8.0914 0.0098
10360328 NM_011318 Apcs serum amyloid P-component 1 H3|1 94.2 cM 9.2166 0 3.1253 0.0123
10451953 NM_029796 Lrg1 leucine-rich alpha-2-glycoprotein 1 17 D|17 10.0 cM 6.9919 6.00E−04 5.0051 0.0109
10574023 NM_008630 Mt2 metallothionein 2 8 C5|8 45.0 cM 6.9697 0.0206 8.4549 0.0307
10515187 NM_007822 Cyp4a14 cytochrome P450, family 4, subfamily a, polypeptide 14 4 D1|4 49.5 cM 4.6609 0.0378 9.8282 0.015
10418434 NM_008407 Itih3 inter-alpha trypsin inhibitor, heavy chain 3 14 A2-C1 4.2071 9.00E−04 4.0594 0.0078
10543017 NM_013743 Pdk4 pyruvate dehydrogenase kinase, isoenzyme 4 6 A1|6 0.63 cM 3.5193 0.0145 3.492 0.0368
10503520 NM_015767 Ttpa tocopherol (alpha) transfer protein 4 A3|4 22.7 cM 3.3338 0.003 2.5756 0.0354
10492748 NM_010196 Fga fibrinogen alpha chain 3 F1|3 44.8 cM 3.1077 0.0013 2.2512 0.0307
10496727 NM_026993 Ddah1 dimethylarginine dimethylaminohydrolase 1 3 H3 3.0694 0.001 2.967 0.0098
10583732 NM_010700 Ldlr low density lipoprotein receptor 9 A3|9 5.0 cM 3.0527 0 2.0755 0.0024
10507177 NM_001100181 Cyp4a32 cytochrome P450, family 4, subfamily a, polypeptide 32 4 D1 2.7853 0.0347 3.3932 0.0344
10527920 NM_020010 Cyp51 cytochrome P450, family 51 5 A2|5 1.2 cM 2.7708 0.001 2.3088 0.015
10362073 NM_001161845 Sgk1 serum/glucocorticoid regulated kinase 1 10 A3 2.7412 0.0073 3.4689 0.0109
10379153 NM_009657 Aldoc aldolase C, fructose-bisphosphate 11 B5|11 44.98 cM 2.699 0.0096 2.6246 0.0301
10420730 NM_010191 Fdft1 farnesyl diphosphate farnesyl transferase 1 14 D1|14 3.0 cM 2.6151 0.001 2.0228 0.0228
10515201 NM_007823 Cyp4b1 cytochrome P450, family 4, subfamily b, polypeptide 1 4 D1|4 49.5 cM 2.5614 0.001 1.7862 0.0445
10568001 NM_133670 Sult1a1 sulfotransferase family 1A, phenol-preferring, member 1 7 F3|7 4.0 cM 2.5199 1.00E−04 1.7899 0.0109
10520362 NM_153526 Insig1 insulin induced gene 1 5 B1 2.4631 0.0014 1.836 0.0451
10412909 NM_010191 Fdft1 farnesyl diphosphate farnesyl transferase 1 14 D1|14 3.0 cM 2.4306 6.00E−04 2.072 0.0109
10424349 NM_009270 Sqle squalene epoxidase 15 D1 2.3683 0.0023 1.9609 0.0307
10478048 NM_008489 Lbp lipopolysaccharide binding protein 2 H1|2 83.0 cM 2.3404 6.00E−04 1.8161 0.0194
10499483 NM_134469 Fdps farnesyl diphosphate synthetase 3 F1|3 42.6 cM 2.2316 0.0044 2.2171 0.017
10412466 NM_145942 Hmgcs1 3-hydroxy-3-methylglutaryl -Coenzyme A synthase 1 2.213 0.0012 1.7749 0.0294
10364194 NM_146006 Lss lanosterol synthase 10 C1|10 41.1 cM 2.1256 0.0051 1.9021 0.0346
10378793 NM_025655 Tmigd1 transmembrane and immunoglobulin domain containing 1 11 B5 2.0643 0.0316 3.0221 0.0109
10365830 2.0533 0.0032 1.9576 0.0178
10542470 NM_019946 Mgst1 microsomal glutathione S-transferase 1 6 G1 1.9951 0.0124 1.9203 0.0416
10506571 NM_053272 Dhcr24 24-dehydrocholesterol reductase 4 C7 1.8836 0.0014 1.8709 0.0109
10450242 NM_009780 C4b complement component 4B (Childo blood group) 17 B1|17 18.8 cM 1.8607 0.0124 1.849 0.0333
10585942 NM_001033498 Gramd2 GRAM domain containing 2 9 B 1.8264 0.0037 1.622 0.0375
10493548 NM_026784 Pmvk phosphomevalonate kinase 3 F1 1.7852 0.0105 1.808 0.0273
10492964 NM_009690 Cd5l CD5 antigen-like 3 F1 1.6284 0.0195 1.8539 0.0169
10524555 NM_023556 Mvk mevalonate kinase 5 F|5 64.0 cM 1.6043 0.0144 1.63 0.0307
10600082 NM_010941 Nsdhl NAD(P) dependent steroid dehydrogenase-like X A7.3|X 28.87 cM 1.5727 0.009 1.7157 0.0116
10573626 NM_173866 Gpt2 glutamic pyruvate transaminase (alanine aminotransferase) 2 8 C3 1.5293 0.0438 1.6633 0.0425
10526630 NM_015799 Trfr2 transferrin receptor 2 5 G2 1.5175 0.0197 1.5238 0.0435
10456699 NM_177470 Acaa2 acetyl-Coenzyme A acyltransferase 2 (mitochondrial 3-oxoacy l-Coenzyme A thiolase) 18 E2|18 45.0 cM 1.5095 0.029 1.6537 0.0273
10393970 NM_007988 Fasn fatty acid synthase 11 E2|11 72.0 cM 1.4769 0.004 1.4846 0.015
10525893 NM_030210 Aacs acetoacetyl-CoA synthetase 5 F 1.4673 0.0242 1.4896 0.0456
10425695 NM_033218 Srebf2 sterol regulatory element binding factor 2 15 E1 1.4633 0.0095 1.4826 0.0233
10569972 NM_026058 Lass4 LAG1 homolog, ceramide synthase 4 8 A1.2 1.4318 0.0145 1.5 0.0228
10570437 NM_025785 Fbxo25 F-box protein 25 8 A1.1 −1.3809 0.0356 −1.4953 0.0294
10447239 NM_026180 Abcg8 ATP-binding cassette, sub-family G (WHITE), member 8 17 E4|17 54.5 cM −1.5584 0.013 −1.8003 0.0109
10425945 NM_010180 Fbln1 fibulin 1 15 E-F −1.5672 0.013 −1.601 0.0287
10453318 NM_031884 Abcg5 ATP-binding cassette, sub-family G (WHITE), member 5 17 E4|17 54.5 cM −1.6829 0.0069 −1.6878 0.0224
10390032 NM_153807 Acsf2 acyl-CoA synthetase family member 2 11 D −1.8821 0.0041 −2.228 0.0078
10492426 NM_153807 Acsf2 acyl-CoA synthetase family member 2 11 D −1.9567 0.0045 −2.1236 0.0109
10518674 NM_022020 Rbp7 retinol binding protein 7, cellular 4 E2 −2.0739 0.0021 −1.8152 0.0258
10462442 NM_001164724 Il33 interleukin 33 19 C2 −2.0935 0.0078 −1.9454 0.0354
10575833 NM_008290 Hsd17b2 hydroxysteroid (17-beta) dehydrogenase 2 8 E1 −2.2055 0.0025 −1.9459 0.026
10577655 NM_008324 Ido1 indoleamine 2,3-dioxygenase 1 8 A2 −2.4459 0.0461 −3.4593 0.0249
10463005 NM_028089 Cyp2c55 cytochrome P450, family 2, subfamily c, polypeptide 55 19 C3 −2.5559 0.0458 −14.6416 4.00E−04
10373334 NM_013786 Hsd17b6 hydroxysteroid (17-beta) dehydrogenase 6 10 D3 −2.6186 2.00E−04 −1.7122 0.0241
10502214 NM_027816 Cyp2u1 cytochrome P450, family 2, subfamily u, polypeptide 1 3 H1 −3.4307 1.00E−04 -1.6652 0.0425
10514912 NM_007860 Dio1 deiodinase, iodothyronine, type I 4 C7|4 48.7 cM −4.5064 0.0012 −3.8938 0.0109
10480155 NM_001081084 Cubn cubilin (intrinsic factor -cobalamin receptor) 2 A1|2 9.0 cM −4.8123 3.00E−04 −2.7793 0.016
10585749 NM_009992 Cyp1a1 cytochrome P450, family 1, subfamily a, polypeptide 1 9 B|9 31.0 cM −5.6507 0.001 −3.3135 0.0301
10513420 NM_001134675 Mup7 major urinary protein 7 4 B3|4 127.8618 0.001
10513455 NM_008647 Mup2 major urinary protein 2 4 B3 126.9789 0.001
10513437 NM_001164526 Mup11 major urinary protein 11 4 B3 117.1801 0.001
10513428 NM_001045550 Mup2 major urinary protein 2 4 B3 113.5703 0.001
10513472 NM_008647 Mup2 major urinary protein 2 4 B3 104.5563 0.001
10513504 NM_001045550 Mup2 major urinary protein 2 4 B3 100.7876 0.001
10513497 NM_001045550 Mup2 major urinary protein 2 4 B3 94.2911 0.001
10513521 NM_001012323 Mup20 major urinary protein 20 4 B3 93.1978 0.0021
10513467 NM_001045550 Mup2 major urinary protein 2 4 B3 90.6275 0.001
10513512 NM_001163011 Mup1 major urinary protein 1 4 B3|4 27.8 cM 73.9991 0.001
10523062 NM_009654 Alb albumin 5 E1|5 50.0 cM 61.2531 4.00E−04
10434689 NM_013465 Ahsg alpha-2-HS-glycoprotein 16 B1|16 15.0 cM 58.1197 3.00E−04
10531149 NM_008096 Gc group specific component 5 E1|5 44.0 cM 52.3423 1.00E−04
10492735 NM_133862 Fgg fibrinogen gamma chain 3 E3|3 41.3 cM 38.7526 1.00E−04
10498981 NM_181849 Fgb fibrinogen beta chain 3 E3|3 48.2 cM 38.2336 1.00E−04
10402406 NM_009245 Serpina1c serine (or cysteine) peptidase inhibitor, clade A, member 1C 12 E|12 51.0 cM 30.3389 3.00E−04
10402399 NM_009243 Serpina1a serine (or cysteine) peptidase inhibitor, clade A, member 1A 12 E|12 51.0 cM 23.9693 3.00E−04
10558673 NM_021282 Cyp2e1 cytochrome P450, family 2, subfamily e, polypeptide 1 7 F5|7 68.4 cM 21.9342 0.0012
10562169 NM_032541 Hamp hepcidin antimicrobial peptide 7 B1|7 11.0 cM 21.1349 0.004
10434719 NM_001102411 Kng1 kininogen 1 16 B1 19.6656 7.00E−04
10402409 NM_009247 Serpina1e serine (or cysteine) peptidase inhibitor, clade A, member 1E 12 E 18.9172 3.00E−04
10548207 NM_007376 Pzp pregnancy zone protein 6 F1-G3|6 62.0 cM 18.3515 3.00E−04
10402390 NM_009244 Serpina1b serine (or cysteine) preptidase inhibitor, clade A, member 1B 12 E|12 51.0 cM 15.5285 5.00E−04
10498921 NM_019911 Tdo2 tryptophan 2,3-dioxygenase 3 E3 13.9226 0.0023
10596148 NM_133977 Trf transferrin 9 F1-F3|9 56.0 cM 13.3568 4.00E−04
10382189 NM_013475 Apoh apolipoprotein H 11 D|11 63.0 cM 13.1193 0.001
10553274 NM_011314 Saa2 serum amyloid A 2 7 B4|7 23.5 cM 10.9174 0.015
10541480 NR_027619 Mug-ps1 murinoglobulin, pseudogene 1 6 F1 10.2599 0.0013
10496825 NM_009474 Uox urate oxidase 3 H2|3 75.0 cM 10.1685 0.0046
10497337 NM_009799 Car1 carbonic anhydrase 1 3 A1|3 10.5 cM 9.9909 0.0175
10454192 NM_013697 Ttr transthyretin 18 A2|18 7.0 cM 9.9844 0.0025
10463037 NM_010003 Cyp2c39 cytochrome P450, family 2, subfamily c, polypeptide 39 19 C3 9.7038 1.00E−04
10398060 NM_011458 Serpina3k serine (or cysteine) peptidase inhibitor, clade A, member 3K 12 E|12 51.5 cM 9.5497 0.0042
10541448 NR_027619 Mug-ps1 murinoglobulin, pseudogene 1 6 F1 9.4334 0.0027
10541410 NM_008645 Mug1 murinoglobulin 1 6 F1 9.3764 0.001
10379190 NM_011707 Vtn vitronectin 11 B5|11 45.09 cM 8.9998 0.0014
10431915 NM_027052 Slc38a4 solute carrier family 38, member 4 15 F1 8.9956 0.0027
10351546 NM_013474 Apoa2 apolipoprotein A-II 1 H3|1 92.6 cM 8.8013 8.00E−04
10467410 NM_145499 Cyp2c70 cytochrome P450, family 2, subfamily c, polypeptide 70 19 C3 8.4844 0.0014
10467319 NM_001159487 Rbp4 retinol binding protein 4, plasma 19 D1|19 38.0 cM 7.858 0.0021
10542983 NM_011134 Pon1 paraoxonase 1 6 A2|6 0.5 cM 7.6708 0.021
10560618 NM_007469 Apoc1 apolipoprotein C-I 7 A3|7 4.0 cM 7.6693 0.0065
10398075 NM_009252 Serpina3n serine (or cysteine) peptidase inhibitor, clade A, member 3N 12 F1 7.4314 0.0115
10368343 NM_007482 Arg1 arginase, liver 10 A4 7.3671 9.00E−04
10563611 NM_009117 Saa1 serum amyloid A 1 7 B4|7 23.5 cM 7.1769 0.0095
10434709 NM_053176 Hrg histidine-rich glycoprotein 16 B1|16 14.1 cM 7.0319 0.0028
10513529 NM_001039544 Mup3 major urinary protein 3 4 B3 6.9857 0.0286
10549154 BC151094 Gm766 predicted gene 766 6 G3 6.9004 0.0267
10413615 NM_018746 Itih4 inter alpha-trypsin inhibitor, heavy chain 4 14 B|14 11.75 cM 6.7951 0.0012
10441753 NM_008877 Plg plasminogen 17 A1|17 7.3 cM 6.266 0.0022
10401289 NM_001177561 Slc10a1 solute carrier family 10 (sodium/bile acid cotransporter family), member 1 12 D1|12 37.0 cM 6.0785 0.0028
10485027 NM_010168 F2 coagulation factor II 2 E1|2 47.5 cM 6.048 0.0096
10402394 NM_009246 Serpina1d serine (or cysteine) peptidase inhibitor, clade A, member 1D 12 E|12 51.0 cM 6.0378 0.0135
10363860 NM_025807 Slc16a9 solute carrier family 16 (monocarboxylic acid transporters), member 9 10 B5.3 5.8753 0.0065
10434698 NM_021564 Fetub fetuin beta 16 B|16 14.1 cM 5.7467 0.0098
10580635 NM_053200 Ces3 carboxylesterase 3 8 C5 5.6566 0.0021
10526712 NM_013478 Azgp1 alpha-2-glycoprotein 1, zinc 5 G2|5 78.0 cM 5.4824 0.0232
10506125 NM_013913 Angptl3 angiopoietin-like 3 4 C6|4 48.0 cM 5.2732 0.0124
10580624 NM_007954 Es1 esterase 1 8 C5|8 43.0 cM 5.1268 0.0355
10513630 NM_007443 Ambp alpha 1 microglobulin/bikunin 4 C1-C3|4 30.6 cM 5.1009 0.0025
10351852 NM_007768 Crp C-reactive protein, pentraxin-related 1 H3|1 94.2 cM 4.9882 0.0025
10363541 NM_007494 Ass1 argininosuccinate synthetase 1 2 B|2 20.0 cM 4.9534 0.0057
10480003 NM_010582 Itih2 inter-alpha trypsin inhibitor, heavy chain 2 2 A1|2 1.0 cM 4.9513 0.0059
10542615 NM_020495 Slco1b2 solute carrier organic anion transporter family, member 1b2 6 G1|6 67.0 cM 4.89 0.0212
10367221 NM_133997 Apof apolipoprotein F 10 D3|10 73.0 cM 4.8785 0.0025
10551287 NM_133657 Cyp2a12 cytochrome P450, family 2, subfamily a, polypeptide 12 7 A3 4.8046 0.0096
10494643 NM_008256 Hmgcs2 3-hydroxy-3-methylglutaryl -Coenzyme A synthase 2 3 F2.2|3 48.0 cM 4.5815 0.0144
10563602 NM_011316 Saa4 serum amyloid A 4 7 B4|7 23.5 cM 4.5245 0.001
10471154 NM_007494 Ass1 argininosuccinate synthetase 1 2 B|2 20.0 cM 4.4979 0.0069
10357516 NM_007576 C4bp complement component 4 binding protein 1 E4|1 67.6 cM 4.4217 0.001
10416451 NM_019775 Cpb2 carboxypeptidase B2 (plasma) 14 D2 4.3546 0.0036
10551293 NM_007817 Cyp2f2 cytochrome P450, family 2, subfamily f, polypeptide 2 7 A3 4.2794 0.0394
10398069 NM_009253 Serpina3m serine (or cysteine) peptidase inhibitor, clade A, member 3M 12 E 4.2775 0.0231
10438681 NM_201375 Kng2 kininogen 2 16 B1 4.2464 0.0051
10367215 NM_133996 Apon apolipoprotein N 10 D3|10 76.0 cM 4.2277 0.0021
10351015 NM_080844 Serpinc1 serine (or cysteine) peptidase inhibitor, clade C (antithrombin), member 1 1 H2.1|1 84.6 cM 4.1985 0.0034
10403322 NM_030611 Akr1c6 aldo-keto reductase family 1, member C6 13 A2|13 8.0 cM 4.1493 0.0498
10452316 NM_009778 C3 complement component 3 17 E1-E3|17 34.3 cM 4.1151 0.0339
10596718 NM_023805 Slc38a3 solute carrier family 38, member 3 9 F1|9 63.0 cM 4.1024 0.0239
10501555 NM_007446 Amy1 amylase 1, salivary 3 F3|3 50.0 cM 4.1021 0.0069
10519497 NM_054098 Steap4 STEAP family member 4 5 A1 3.9917 0.0063
10496001 NM_007686 Cfi complement component factor i 3 G3|3 66.6 cM 3.9698 0.0014
10475532 NM_021507 Sqrdl sulfide quinone reductase-like (yeast) 2 F2 3.9595 0
10593225 NM_001033324 Zbtb16 zinc finger and BTB domain containing 16 9 A5.3|9 23.0 cM 3.8909 0.018
10458828 NM_033037 Cdo1 cysteine dioxygenase 1, cytosolic 18 C|18 23.0 cM 3.7452 0.0025
10514763 NM_146148 C8a complement component 8, alpha polypeptide 4 C6 3.7375 0.0096
10358339 NM_009888 Cfh complement component factor h 1 F|1 74.1 cM 3.6868 0.0032
10514532 NM_010007 Cyp2j5 cytochrome P450, family 2, subfamily j, polypeptide 5 4 C5|4 46.5 cM 3.6645 0.0256
10450038 NM_020581 Angptl4 angiopoietin-like 4 17 B1 3.5851 0.0062
10511375 NM_007824 Cyp7a1 cytochrome P450, family 7, subfamily a, polypeptide 1 4 A1 3.5797 0.0076
10423002 NM_207216 Ugt3a1 UDP glycosyltransferases 3 family, polypeptide A1 15 A1 3.5454 0.0345
10483410 NM_021022 Abcb11 ATP-binding cassette, sub-family B (MDR/TAP), member 11 2 C2|2 38.4 cM 3.4306 0.0445
10447885 NM_153151 Acat3 acetyl-Coenzyme A acetyltransferase 3 17 A1|17 7.55 cM 3.3695 0.0023
10537169 NM_009731 Akr1b7 aldo-keto reductase family 1, member B7 6 B1|6 14.0 cM 3.3685 0.0032
10395273 BC052902 Gdap10 ganglioside-induced differentiation-associated-protein 10 12 A3 3.3561 0.0043
10503502 NM_015767 Ttpa tocopherol (alpha) transfer protein 4 A3|4 22.7 cM 3.2169 0.0089
10585015 NM_080434 Apoa5 apolipoprotein A-V 9 B 3.2153 0.0026
10581388 NM_008490 Lcat lecithin cholesterol acyltransferase 8 D3|8 53.0 cM 3.2103 0.0014
10482004 BC094504 AI182371 expressed sequence AI182371 2 B 3.1837 0.0281
10574027 NM_013602 Mt1 metallothionein 1 8 C5|8 45.0 cM 3.1654 0.0374
10379736 NM_183249 1100001G20Rik RIKEN cDNA 1100001G20 gene 11 C 3.1049 0.0151
10420899 NM_178747 Gulo gulonolactone (L-) oxidase 14 D1 3.0186 0.0345
10435626 NM_013547 Hgd homogentisate 1, 2-dioxygenase 16 B3|16 27.3 cM 2.9656 0.0296
10345065 NM_001077353 Gsta3 glutathione S-transferase, alpha 3 1 A4|1 15.0 cM 2.9309 0.0145
10356145 NM_030556 Slc19a3 solute carrier family 19 (sodium/hydrogen exchanger), member 3 1 C5|1 51.0 cM 2.9123 0.0301
10533612 NM_008277 Hpd 4-hydroxyphenylpyruvic acid dioxygenase 5 F|5 67.0 cM 2.8602 0.0373
10451451 NM_010321 Gnmt glycine N-methyltransferase 17 C|17 10.0 cM 2.8319 0.0461
10543333 NM_013930 Aass aminoadipate-semialdehyde synthase 6 A3.1|6 4.5 cM 2.8069 0.0115
10596166 NM_027918 1300017J02Rik RIKEN cDNA 1300017J02 gene 9 F1 2.7756 0.0308
10406646 NM_028772 Dmgdh dimethylglycine dehydrogenase precursor 13 C3 2.7336 0.0244
10500547 NM_153193 Hsd3b2 hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 2 3 F2.2|3 49.1 cM 2.6399 0.0032
10570717 NM_001177522 Gm14850 predicted gene 14850 8 A2 2.5973 0.0148
10361234 NM_008288 Hsd11b1 hydroxysteroid 11-beta dehydrogenase 1 1 H6 2.5957 0.0096
10507171 NM_201640 Cyp4a31 cytochrome P450, family 4, subfamily a, polypeptide 31 4 D1 2.575 0.0484
10570732 NM_001177528 Gm15315 predicted gene 15315 8 A2|8 2.5592 0.0098
10599826 NM_007979 F9 coagulation factor IX X A6-A7|X 22.0 cM 2.5474 0.0143
10578916 NM_025436 Sc4mol sterol-C4-methyl oxidase-like 8 B3.1 2.5379 0.0023
10399365 NM_177802 Slc7a15 solute carrier family 7 (cationic amino acid transporter, y+ system), member 15 12 A1.1 2.5357 0.0361
10570693 NM_007851 Defa5 defensin, alpha, 5 8 A2 2.5181 0.0129
10497590 NM_007963 Mecom MDS1 and EVI1 complex locus 3 A3|3 14.4 cM 2.5047 0.0091
10545881 NM_053096 Cml2 camello-like 2 6 C3 2.5013 0.027
10478523 2.4962 0.0021
10454944 NM_201256 Eif4ebp3 eukaryotic translation initiation factor 4E binding protein 3 18 B2 2.4951 0.0117
10386683 NM_026183 Slc47a1 solute carrier family 47, member 1 11 B2 2.4534 0.0067
10365559 NM_010512 Igf1 insulin-like growth factor 1 10 C1|10 48.0 cM 2.4375 0.0041
10500555 NM_001161742 Hsd3b3 hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 3 3 F2.2|3 49.1 cM 2.4177 0.0094
10513412 NM_008648 Mup4 major urinary protein 4 4 B3|4 27.8 cM 2.3986 5.00E−04
10491846 2.3936 0.0128
10537306 NM_145364 Akr1d1 aldo-keto reductase family 1, member D1 6 B1 2.3924 0.0045
10589099 NM_029634 Ip6k2 inositol hexaphosphate kinase 2 9 F2 2.3776 0.0021
10594825 NM_022026 Aqp9 aquaporin 9 9 D 2.3723 0.0412
10519578 NM_008830 Abcb4 ATP-binding cassette, sub-family B (MDR/TAP), member 4 5 A1|5 1.0 cM 2.3672 0.0297
10590957 NM_010242 Fut4 fucosyltransferase 4 9 A2|9 3.0 cM 2.3611 0.0019
10447317 NM_010137 Epas1 endothelial PAS domain protein 1 17 E4 2.2998 0.0142
10352000 NM_133809 Kmo kynurenine 3-monooxygenase (kynurenine 3-hydroxylase) 1 H4 2.2966 0.0301
10360840 NM_001081361 Mosc1 MOCO sulphurase C-terminal domain containing 1 2.2607 0.0105
10570660 NM_001177481 Gm10104 predicted gene 10104 8 A2|8 2.2377 0.0163
10386460 NM_008819 Pemt phosphatidylethanolamine N-methyltransferase 11 B1.3|11 31.0 cM 2.2274 0.0265
10576901 NM_011388 Slc10a2 solute carrier family 10, member 2 8 A1.1|8 2.0 cM 2.2212 0.0253
10530319 NM_001038999 Atp8a1 ATPase, aminophospholipid transporter (APLT), class I, type 8A, member 1 5 C3.1 2.1827 0.0152
10502050 NM_027808 Alpk1 alpha-kinase 1 3 H1 2.1604 0.04
10381096 NM_010517 Igfbp4 insulin-like growth factor binding protein 4 11 D 2.1452 0.0025
10408689 NM_153529 Nrn1 neuritin 1 13 A3.3 2.1125 0.0301
10587266 NM_010295 Gclc glutamate-cysteine ligase, catalytic subunit 9 D-E|9 42.0 cM 2.1067 0.0473
10429588 NM_001039720 9030619P08Rik RIKEN cDNA 9030619P08 gene 15 D3 2.1009 0.0025
10537146 NM_008012 Akr1b8 aldo-keto reductase family 1, member B8 6 B1|6 13.0 cM 2.0926 0.0397
10576774 NM_029465 Clec4g C-type lectin domain family 4, member g 8 A1.1 2.0888 0.0367
10542592 NR_033555 Gm10400 predicted gene 10400 2.0729 0.0104
10496605 NM_173763 Ccbl2 cysteine conjugate-beta lyase 2 3 H1 2.0685 0.0222
10358299 NM_001029977 EG214403 predicted gene, EG214403 1 F 2.0607 0.0386
10494023 NM_011281 Rorc RAR-related orphan receptor gamma 3 F2 2.0501 0.0013
10357363 NM_001081756 Nckap5 NCK-associated protein 5 1 E3 2.0191 0.023
10506452 AY512949 AY512949 cDNA sequence AY512949 2.0161 0.0416
10512895 NM_007519 Baat bile acid-Coenzyme A: amino acid N-acyltransferase 4 B1|4 22.7 cM 2.0089 0.0204
10388430 NM_011340 Serpinf1 serine (or cysteine) peptidase inhibitor, clade F, member 1 11 B5 2.0036 0.0181
10454198 NM_026301 Rnf125 ring finger protein 125 18 A2 2.003 0.0053
10606989 NM_001077364 Tsc22d3 TSC22 domain family, member 3 X F1 1.9909 0.0197
10418455 NM_008406 Itih1 inter-alpha trypsin inhibitor, heavy chain 1 14 A2-C1 1.9848 0.0374
10365482 NM_011595 Timp3 tissue inhibitor of metalloproteinase 3 10 C1-D1|10 47.0 cM 1.9692 0.0355
10388440 NM_008878 Serpinf2 serine (or cysteine) peptidase inhibitor, clade F, member 2 11 B5 1.9646 0.043
10372682 NM_029875 Slc35e3 solute carrier family 35, member E3 10 D2 1.9641 0.0062
10571657 NM_007981 Acsl1 acyl-CoA synthetase long-chain family member 1 8 B2 1.9589 0.0041
10581650 NM_011998 Chst4 carbohydrate (chondroitin 6/keratan) sulfotransferase 4 1.9469 0.039
10394990 NM_026037 Mboat2 membrane bound O-acyltransferase domain containing 2 12 A1.3 1.9401 0.0244
10596072 NM_001161362 Ppp2r3a protein phosphatase 2, regulatory subunit B'', alpha 9 E4 1.9318 0.0021
10411332 NM_008255 Hmgcr 3-hydroxy-3-methylglutaryl-Coenzyme A reductase 13 D1|13 49.0 cM 1.9292 0.0013
10571840 NM_008278 Hpgd hydroxyprostaglandin dehydrogenase 15 (NAD) 8 B3.2 1.9235 0.0025
10435787 1.9234 0.0109
10555438 NM_199012 Fchsd2 FCH and double SH3 domains 2 7 E3 1.9147 0.0156
10436100 NM_181596 Retnlg resistin like gamma 16 B5|16 32.5 cM 1.9126 0.0028
10542594 AY512955 Gm10210 predicted gene 10210 1.9107 0.047
10554693 NM_023377 Stard5 StAR-related lipid transfer (START) domain containing 5 7 D3 1.903 0.0204
10391744 AK135410 Gpatch8 G patch domain containing 8 11 E1 1.9025 0.0148
10560624 NM_009696 Apoe apolipoprotein E 7 A3|7 4.0 cM 1.8965 0.0301
10542880 BC048711 4833442J19Rik RIKEN cDNA 4833442J19 gene 6 G3 1.8917 0.0144
10461150 1.8816 0.0397
10585982 NM_173018 Myo9a myosin IXa 9 B 1.8764 0.019
10547469 NM_001037155 Hsn2 hereditary sensory neuropathy, type II 6 F1 1.8743 0.0152
10482762 NM_145360 Idi1 isopentenyl-diphosphate delta isomerase 13 A1 1.8722 0.0143
10411147 NM_022884 Bhmt2 betaine-homocysteine methyltransferase 2 13 C3|13 49.0 cM 1.8637 0.0317
10364038 NM_133995 Upb1 ureidopropionase, beta 10 C1 1.8607 0.0099
10382888 AK171830 2810008D09Rik RIKEN cDNA 2810008D09 gene 1.8559 0.0272
10485170 NM_009963 Cry2 cryptochrome 2 (photolyase-like) 2 E 1.8553 0.0324
10594988 NM_015806 Mapk6 mitogen-activated protein kinase 6 9 D|9 38.0 cM 1.8546 0.0327
10535938 NM_133898 N4bp2l1 NEDD4 binding protein 2-like 1 5 G3 1.8517 0.0189
10556769 NM_016870 Acsm3 acyl-CoA synthetase medium-chain family member 3 7 F3 1.8511 0.0207
10401149 NM_013738 Plek2 pleckstrin 2 12 D2 1.8429 0.0098
10599192 NM_028894 Lonrf3 LON peptidase N-terminal domain and ring finger 3 X A2 1.8374 0.035
10391746 NM_001159492 Gpatch8 G patch domain containing 8 11 E1 1.8331 0.0248
10403413 NM_145360 Idi1 isopentenyl-diphosphate delta isomerase 13 A1 1.8228 0.0272
10580955 1.8221 0.0277
10396730 1.8218 0.0374
10508800 AY140896 Gm3579 predicted gene 3579 4 D2.3|4 1.8214 0.0197
10573865 AY140896 Gm3579 predicted gene 3579 4 D2.3|4 1.8214 0.0197
10558961 NM_053082 Tspan4 tetraspanin 4 7 F5 1.8183 0.0132
10350753 NM_008131 Glul glutamate-ammonia ligase (glutamine synthetase) 1.817 0.0285
10351043 NR_028543 Snord47 small nucleolar RNA, C/D box 47 1|1 1.8156 0.0327
10377439 NM_001159367 Per1 period homolog 1 (Drosophila) 11 B 1.8092 0.0359
10520388 NM_028234 Rbm33 RNA binding motif protein 33 5 B1 1.8056 0.0379
10370931 NM_021462 Mknk2 MAP kinase-interacting serine/threonine kinase 2 10 C1 1.7989 0.0023
10582658 NM_007428 Agt angiotensinogen (serpin peptidase inhibitor, clade A, member 8) 8 E2|8 68.0 cM 1.7834 0.0217
10472860 NM_019688 Rapgef4 Rap guanine nucleotide exchange factor (GEF) 4 2 C3 1.7824 0.0118
10505489 NM_021362 Pappa pregnancy-associated plasma protein A 4 C1|4 32.2 cM 1.7809 0.0012
10377751 NM_009714 Asgr1 asialoglycoprotein receptor 1 11 B3|11 37.0 cM 1.7793 0.0146
10351224 NM_007976 F5 coagulation factor V 1 H2.2|1 86.6 cM 1.7735 0.0277
10372988 NM_011391 Slc16a7 solute carrier family 16 (monocarboxylic acid transporters), member 7 10 D3 1.7729 0.0311
10351259 NM_054087 Slc19a2 solute carrier family 19 (thiamine transporter), member 2 1 H2.2|1 87.0 cM 1.7698 0.0242
10409876 NM_007796 Ctla2a cytotoxic T lymphocyte-associated protein 2 alpha 13 B2|13 36.0 cM 1.7598 0.0424
10410877 NM_001081176 Polr3g polymerase (RNA) III (DNA directed) polypeptide G 13 C3 1.7388 0.0345
10424686 BC025446 BC025446 cDNA sequence BC025446 15 D3 1.7378 0.0152
10351533 NM_009803 Nr1i3 nuclear receptor subfamily 1, group I, member 3 1 H3|1 92.6 cM 1.7285 0.0341
10533050 NM_030704 Hspb8 heat shock protein 8 5 F|5 59.0 cM 1.716 0.0394
10496872 NM_133222 Eltd1 EGF, latrophilin seven transmembrane domain containing 1 3 H3-H4 1.7157 0.0233
10518743 NM_173371 H6pd hexose-6-phosphate dehydrogenase (glucose 1-dehydrogenase) 4 E2|4 78.4 cM 1.7094 0.0165
10487021 NM_011774 Slc30a4 solute carrier family 30 (zinc transporter), member 4 2 E5|2 69.0 cM 1.7066 0.0161
10595622 1.7056 0.0342
10595636 1.7056 0.0342
10595626 1.7034 0.029
10446425 1.7003 0.0214
10473160 NM_080558 Ssfa2 sperm specific antigen 2 2 D 1.6908 0.0159
10375229 1.6902 0.0112
10592938 NM_138951 Ttc36 tetratricopeptide repeat domain 36 9 A5.2 1.6901 0.04
10394699 NM_009072 Rock2 Rho-associated coiled-coil containing protein kinase 2 12 A3 1.6898 0.0376
10354085 NM_019570 Rev1 REV1 homolog (S. cerevisiae) 1 B 1.6849 0.0097
10462922 NM_019588 Plce1 phospholipase C, epsilon 1 19 D1 1.6829 0.032
10395259 NM_021524 Nampt nicotinamide phosphoribosyltransferase 12 B1 1.676 0.0284
10597875 NM_010012 Cyp8b1 cytochrome P450, family 8, subfamily b, polypeptide 1 9 F4|9 71.0 cM 1.6755 0.013
10344809 NM_026493 Cspp1 centrosome and spindle pole associated protein 1 1 A2 1.6735 0.0484
10347748 NM_028817 Acsl3 acyl-CoA synthetase long-chain family member 3 1 C4|1 24.1 cM 1.6733 0.019
10375216 NM_145962 Pank3 pantothenate kinase 3 11 A4 1.6733 0.013
10545877 NM_023455 Nat8 N-acetyltransferase 8 (GCN5-related, putative) 6 C3 1.6673 0.0378
10374453 NM_008131 Glul glutamate-ammonia ligase (glutamine synthetase) 1.666 0.0345
10515690 NM_198170 Szt2 seizure threshold 2 4 D2.1 1.6601 0.0207
10591612 NM_177030 Dock6 dedicator of cytokinesis 6 9 A3 1.6594 0.0225
10389214 NM_011338 Ccl9 chemokine (C-C motif) ligand 9 11 C|11 47.4 cM 1.6564 0.0355
10587688 1.6554 0.0277
10595620 1.6554 0.0277
10366275 1.6481 0.0095
10607089 NM_207625 Acsl4 acyl-CoA synthetase long-chain family member 4 X F2 1.647 0.0484
10379820 NM_133360 Acaca acetyl-Coenzyme A carboxylase alpha 11 C 1.6452 0.0014
10414537 NM_001161731 Ang angiogenin, ribonuclease, RNase A family, 5 14 B-C1|14 18.0 cM 1.6406 0.0094
10505779 NM_139306 Acer2 alkaline ceramidase 2 4 C4 1.6403 0.0141
10564183 AF241256 Snord116 small nucleolar RNA, C/D box 116 cluster 7 C|7 29.0 cM 1.6393 0.0126
10544573 NM_027852 Rarres2 retinoic acid receptor responder (tazarotene induced) 2 6 B2.3 1.6174 0.0361
10472436 NM_020283 B3galt1 UDP-Gal:betaGlcNAc beta 1,3-galactosyltransferase, polypeptide 1 2 C3 1.6168 0.0376
10515399 NM_013807 Plk3 polo-like kinase 3 (Drosophila) 4 D1 1.616 0.039
10407876 NM_138654 5033411D12Rik RIKEN cDNA 5033411D12 gene 13 A2 1.6108 0.0317
10478847 NM_001081005 1500012F01Rik RIKEN cDNA 1500012F01 gene 2 H3 1.6077 0.0445
10362896 NM_009846 Cd24a CD24a antigen 10 B2|10 26.0 cM 1.6059 0.0272
10535956 NM_001163493 Stard13 StAR-related lipid transfer (START) domain containing 13 5 G3 1.6025 0.0225
10371400 NM_007771 Cry1 cryptochrome 1 (photolyase-like) 10 C|10 46.0 cM 1.5989 0.0109
10540122 NM_009320 Slc6a6 solute carrier family 6 (neurotransmitter transporter, taurine), member 6 6 D1|6 38.2 cM 1.5937 0.0136
10580953 1.5913 0.0232
10356082 NM_011636 Plscr1 phospholipid scramblase 1 9 E3.3 1.5899 0.036
10453272 NM_001001806 Zfp36l2 zinc finger protein 36, C3H type-like 2 17 E4 1.5897 0.013
10488322 NM_001033348 Ralgapa2 Ral GTPase activating protein, alpha subunit 2 (catalytic) 2 G1 1.5885 0.0019
10462406 NM_001081319 C030046E11Rik RIKEN cDNA C030046E11 gene 19 C1 1.5858 0.0471
10382797 NM_176902 Fam100b family with sequence similarity 100, member B 11 E2 1.583 0.023
10346328 XR_031547 Gm8292 predicted gene 8292 1 C1.2 1.5828 0.0066
10459766 NR_028560 Scarna17 small Cajal body-specific RNA 17 18|18 1.5813 0.0232
10461642 NR_028560 Scarna17 small Cajal body-specific RNA 17 18|18 1.5813 0.0232
10474545 NM_133649 Slc12a6 solute carrier family 12, member 6 2 E3 1.5812 0.0361
10471503 BC110660 Taf1d TATA box binding protein (Tbp)-associated factor, RNA polymerase I, D 9 A3 1.5797 0.0352
10572865 NM_027837 Isx intestine specific homeobox 8 C2 1.5776 0.0224
10357345 NM_172484 Nckap5 NCK-associated protein 5 1 E3 1.5726 0.0374
10501494 NM_001042711 Amy2a5 amylase 2a5 3 F3|3 50.0 cM 1.5715 0.015
10558295 ENSMUST00000106157 Zranb1 zinc finger, RAN-binding domain containing 1 7 F3 1.5699 0.0362
10553829 NR_003376 Gm7367 1110014K08Rik pseudogene 7 C|7 1.5663 0.0376
10553831 NR_003376 Gm7367 1110014K08Rik pseudogene 7 C|7 1.5663 0.0376
10557233 NM_144925 Tnrc6a trinucleotide repeat containing 6a 7 F3 1.5644 0.0447
10439667 NM_145389 BC016579 cDNA sequence, BC016579 16 B5 1.5633 0.0355
10354168 NM_018775 Tbc1d8 TBC1 domain family, member 8 1 B 1.5603 0.0205
10418169 ENSMUST00000079800 // ENSMUST00000079800 1700054O19Rik // 1700054O19Rik RIKEN cDNA 1700054O19 gene // RIKEN cDNA 1700054O19 gene 14 A3 // 14 A3 1.5539 0.0023
10509063 NM_178257 Il22ra1 interleukin 22 receptor, alpha 1 4 D3|4 62.0 cM 1.5466 0.0232
10391732 NM_001159492 Gpatch8 G patch domain containing 8 11 E1 1.5421 0.0168
10399198 NM_019744 Ncoa4 nuclear receptor coactivator 4 14 B 1.5412 0.0444
10512774 NM_178893 Coro2a coronin, actin binding protein 2A 4 B1 1.5393 0.0307
10485633 ENSMUST00000099651 Gm10796 predicted gene 10796 1.5366 0.0286
10350758 AK080751 A930039A15Rik RIKEN cDNA A930039A15 gene 1.5361 0.044
10427461 NM_001136079 Ptger4 prostaglandin E receptor 4 (subtype EP4) 15 A1|15 6.4 cM 1.5341 0.0233
10417887 NM_029104 Zmynd17 zinc finger, MYND domain containing 17 14 B 1.531 0.0494
10436209 NM_001033238 Cblb Casitas B-lineage lymphoma b 16 B5 1.5287 0.0345
10587792 NM_011636 Plscr1 phospholipid scramblase 1 9 E3.3 1.5275 0.0242
10469300 NM_177268 Ankrd16 ankyrin repeat domain 16 2 A1 1.5231 0.0313
10497487 1.5217 0.0217
10510286 NM_027985 Mad2l2 MAD2 mitotic arrest deficient-like 2 (yeast) 4 E1 1.52 0.0152
10475866 NM_207680 Bcl2l11 BCL2-like 11 (apoptosis facilitator) 2 F3-G1 1.5187 0.0194
10497421 NM_080634 Hps3 Hermansky-Pudlak syndrome 3 homolog (human) 3 A2|3 12.5 cM 1.5153 0.0272
10538356 NM_001163640 Chn2 chimerin (chimaerin) 2 6 B3 1.5142 0.0215
10475946 NM_178404 Zc3h6 zinc finger CCCH type containing 6 2 F1 1.5139 0.0465
10584350 NM_009429 Tpt1 tumor protein, translationally-controlled 1 14 D3 1.5134 0.0362
10523856 1.5131 0.0416
10564159 AF241256 Snord116 small nucleolar RNA, C/D box 116 cluster 7 C|7 29.0 cM 1.5126 0.0148
10559916 1.5073 0.0309
10410513 NM_001040692 Slc6a18 solute carrier family 6 (neurotransmitter transporter), member 18 13 C1|13 42.0 cM 1.506 0.0469
10413839 NM_001033988 Ncoa4 nuclear receptor coactivator 4 14 B 1.5054 0.048
10459768 1.5005 0.0316
10562234 NM_001110252 Hpn hepsin 7 B1 1.4972 0.0454
10407370 NM_001113550 4833420G17Rik RIKEN cDNA 4833420G17 gene 13 D2.3 1.4954 0.0245
10517703 1.4948 0.0146
10497441 NM_013755 Gyg glycogenin 3 A2|3 12.5 cM 1.4893 0.0056
10470959 NM_172267 Phyhd1 phytanoyl-CoA dioxygenase domain containing 1 2 B 1.4891 0.0376
10601755 NM_019865 Rpl36a ribosomal protein L36A X E3 1.4875 0.0384
10355050 NM_001045513 Raph1 Ras association (RalGDS/AF-6) and pleckstrin homology domains 1 1 C2 1.4843 0.0308
10514500 NM_001004141 Cyp2j11 cytochrome P450, family 2, subfamily j, polypeptide 11 4 C5 1.483 0.045
10497399 NM_001122759 Pde7a phosphodiesterase 7A 3 A2|3 7.0 cM 1.4697 0.0106
10532040 NM_026856 Zfp644 zinc finger protein 644 5 E5|5 56.0 cM 1.4674 0.0484
10421972 NM_019865 Rpl36a ribosomal protein L36A X E3 1.467 0.0489
10399478 NM_015763 Lpin1 lipin 1 12 A1.1|12 9.0 cM 1.4665 0.0214
10438358 NM_213614 5-Sep septin 5 16 A3|16 11.42 cM 1.4654 0.0468
10544219 NM_139294 Braf Braf transforming gene 6 B1|6 15.5 cM 1.4639 0.0078
10358894 NM_146126 Sord sorbitol dehydrogenase 2 E5|2 66.0 cM 1.4618 0.0277
10401997 NM_011877 Ptpn21 protein tyrosine phosphatase, non-receptor type 21 12 F1 1.4616 0.0405
10475437 NM_146126 Sord sorbitol dehydrogenase 2 E5|2 66.0 cM 1.4599 0.0325
10379034 NM_026708 Tlcd1 TLC domain containing 1 11 B5 1.4516 0.0339
10497935 1.4454 0.0361
10389025 NM_177390 Myo1d myosin ID 11 B5|11 46.0 cM 1.443 0.0454
10389022 NM_177390 Myo1d myosin ID 11 B5|11 46.0 cM 1.4428 0.0447
10396956 NM_018814 Pcnx pecanex homolog (Drosophila) 12 D1 1.4386 0.0122
10462346 NM_021525 Rcl1 RNA terminal phosphate cyclase-like 1 19 C1 1.4359 0.0151
10352416 NM_130890 Capn8 calpain 8 1 H4 1.4317 0.0233
10592001 NM_011176 St14 suppression of tumorigenicity 14 (colon carcinoma) 9 A4|9 17.0 cM 1.4289 0.0123
10507110 1.4264 0.0448
10552030 NM_008085 Gapdhs glyceraldehyde-3-phosphate dehydrogenase, spermatogenic 7 B1 1.4162 0.0437
10447429 ENSMUST00000072072 Gm4832 predicted gene 4832 17 E4 1.4137 0.035
10520371 NM_028234 Rbm33 RNA binding motif protein 33 5 B1 1.4117 0.045
10402117 NM_153587 Rps6ka5 ribosomal protein S6 kinase, polypeptide 5 12 E 1.4072 0.048
10481827 NM_001085507 Zbtb34 zinc finger and BTB domain containing 34 2 B 1.4071 0.0194
10526656 NM_146164 Lrch4 leucine-rich repeats and calponin homology (CH) domain containing 4 5 G2 1.404 0.0447
10582811 NM_001164598 Irf2bp2 interferon regulatory factor 2 binding protein 2 8 E2 1.3943 0.0286
10493343 1.3926 0.0441
10474725 NM_013719 Eif2ak4 eukaryotic translation initiation factor 2 alpha kinase 4 1.3874 0.0301
10489377 NM_012032 Serinc3 serine incorporator 3 2 H3 1.3784 0.0274
10480570 NM_001162485 Arrdc1 arrestin domain containing 1 2 A3 1.3734 0.0478
10366446 NM_146010 Tspan8 tetraspanin 8 10 D2 1.3733 0.0324
10346970 NM_011086 Pikfyve phosphoinositide kinase, FYVE finger containing 1 C2 1.3711 0.0175
10384423 NM_172496 Cobl cordon-bleu 11 A1|11 6.5 cM 1.3708 0.048
10476443 NM_013829 Plcb4 phospholipase C, beta 4 2 F3|2 77.0 cM 1.3705 0.0333
10534889 NM_178162 Agfg2 ArfGAP with FG repeats 2 5 G2 1.369 0.0181
10518428 NM_011929 Clcn6 chloride channel 6 4 76.4 cM 1.3681 0.0262
10542264 NM_001163445 2700089E24Rik RIKEN cDNA 2700089E24 gene 6 G1 1.3677 0.045
10497485 1.3662 0.0333
10349065 NM_001122676 Zcchc2 zinc finger, CCHC domain containing 2 1 E2.1 1.3603 0.0286
10477311 NM_001039939 Asxl1 additional sex combs like 1 (Drosophila) 2 H1 1.3592 0.0395
10432122 NM_001170711 Asb8 ankyrin repeat and SOCS box-containing 8 15 F2 1.3504 0.0362
10567229 NM_001031814 Smg1 SMG1 homolog, phosphatidylinositol 3-kinase-related kinase (C. elegans) 7 F2 1.3481 0.0485
10376312 NM_028451 Larp1 La ribonucleoprotein domain family, member 1 11 B2 1.3375 0.0311
10503161 NM_001081417 Chd7 chromodomain helicase DNA binding protein 7 4 A1|4 1.0 cM 1.3312 0.0407
10587150 NM_001081322 Myo5c myosin VC 9 D 1.3133 0.0489
10476033 NM_183262 Stk35 serine/threonine kinase 35 2 F1 1.2947 0.0412
10466530 NM_001163144 Pcsk5 proprotein convertase subtilisin/kexin type 5 19 B|19 18.0 cM 1.2894 0.0362
10478219 NM_021280 Plcg1 phospholipase C, gamma 1 2 H2|2 92.0 cM 1.2708 0.0381
10367919 NM_029075 Stx11 syntaxin 11 10 A1 −1.2794 0.048
10605319 NM_145405 Ubl4 ubiquitin-like 4 X A7.3|X 29.9 cM −1.2895 0.048
10508089 NM_025544 Mrps15 mitochondrial ribosomal protein S15 4 D2.1 −1.2959 0.0479
10591482 NM_016679 Keap1 kelch-like ECH-associated protein 1 9 A3 −1.298 0.0397
10591739 NM_001102404 Acp5 acid phosphatase 5, tartrate resistant 9 A3|9 6.0 cM −1.3007 0.0272
10551159 XR_034166 Gm4607 predicted gene 4607 7 A3|7 −1.3016 0.0479
10413222 NM_134084 Ppif peptidylprolyl isomerase F (cyclophilin F) 14 A3 −1.3037 0.048
10356657 NM_024197 Ndufa10 NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 10 1 D −1.3055 0.0479
10347672 NM_027886 Stk11ip serine/threonine kinase 11 interacting protein 1 C3 −1.3059 0.0438
10439695 NM_019754 Tagln3 transgelin 3 16 B5 −1.3087 0.0387
10593591 NM_144784 Acat1 acetyl-Coenzyme A acetyltransferase 1 9 30.0 cM −1.3115 0.0418
10432866 NM_001003667 Krt77 keratin 77 15 F3 −1.3141 0.0484
10378443 NM_001011851 Olfr412 olfactory receptor 412 11 B5 −1.3165 0.048
10487629 NM_130884 Idh3b isocitrate dehydrogenase 3 (NAD+) beta 2 F3 −1.3214 0.0265
10432636 NM_174992 Smagp small cell adhesion glycoprotein 15 F1 −1.3221 0.0376
10543684 NR_030713 Mir183 microRNA 183 −1.3247 0.0355
10411156 NM_029153 Scamp1 secretory carrier membrane protein 1 13 D1 −1.3284 0.036
10585417 NM_029573 Idh3a isocitrate dehydrogenase 3 (NAD+) alpha 9 A5.3 −1.3357 0.0367
10415742 NM_027436 Mipep mitochondrial intermediate peptidase 14 D1 −1.3368 0.0307
10406205 NM_030711 Erap1 endoplasmic reticulum aminopeptidase 1 13 C1 −1.3442 0.0454
10449940 NM_022434 Cyp4f14 cytochrome P450, family 4, subfamily f, polypeptide 14 17 B1 −1.3479 0.0363
10420225 NM_010780 Cma1 chymase 1, mast cell 14 C3|14 20.0 cM −1.3516 0.0307
10390685 NM_025661 Ormdl3 ORM1-like 3 (S. cerevisiae) 11 D −1.3522 0.0445
10484431 NM_025868 Tmx2 thioredoxin-related transmembrane protein 2 2 D −1.3536 0.0486
10515930 BC076608 AA415398 expressed sequence AA415398 4 D2.1 −1.3575 0.0362
10607643 −1.3579 0.0374
10386416 NM_001011789 Olfr222 olfactory receptor 222 11 B1.3 −1.3596 0.0447
10488291 NM_015754 Rbbp9 retinoblastoma binding protein 9 2 G1-H1 −1.3617 0.032
10447551 NM_027457 5730437N04Rik RIKEN cDNA 5730437N04 gene 17 A1 −1.3687 0.0396
10464128 NM_007611 Casp7 caspase 7 19 D2|19 50.0 cM −1.3699 0.0383
10377286 NM_001081566 Pik3r6 phosphoinositide-3-kinase, regulatory subunit 6 11 B3 −1.3731 0.0233
10539472 NM_019542 Nagk N-acetylglucosamine kinase 6 D1 −1.3736 0.0143
10434191 NM_013711 Txnrd2 thioredoxin reductase 2 16 A3|16 11.2 cM −1.3797 0.0324
10369525 BC024943 2010107G23Rik RIKEN cDNA 2010107G23 gene 10 B4 −1.3815 0.0461
10465826 NM_001160356 AI462493 expressed sequence AI462493 19 A −1.3869 0.0399
10435617 NM_001042499 Rabl3 RAB, member of RAS oncogene family-like 3 16 B3 −1.3924 0.0387
10495285 NM_019972 Sort1 sortilin 1 3 F3 −1.3932 0.035
10447354 NM_025868 Tmx2 thioredoxin-related transmembrane protein 2 2 D −1.3953 0.0338
10575894 NM_026648 Lrrc50 leucine rich repeat containing 50 8 E1 −1.3988 0.0418
10368893 NM_145743 Lace1 lactation elevated 1 10 B2|10 25.5 cM −1.399 0.0265
10470050 NM_007379 Abca2 ATP-binding cassette, sub-family A (ABC1), member 2 2 A2-B|2 12.6 cM −1.4023 0.0316
10566618 NM_206897 Olfr6 olfactory receptor 6 7 E3 −1.4057 0.0416
10526514 NM_021719 Cldn15 claudin 15 5 G2 −1.4077 0.0418
10483679 NM_001080707 Gpr155 G protein-coupled receptor 155 2 C3 −1.4079 0.035
10476301 NM_001177833 Smox spermine oxidase 2 F1 −1.408 0.0478
10365116 NM_133964 Dohh deoxyhypusine hydroxylase/monooxygenase 10 C1 −1.4203 0.0304
10386086 NM_153079 Nmur2 neuromedin U receptor 2 11 B1.3 −1.4289 0.043
10442370 NM_019910 Dcpp1 demilune cell and parotid protein 1 17 A3.3|17 10.0 cM −1.4357 0.043
10593169 NM_023114 Apoc3 apolipoprotein C-III 9 A5.2|9 27.0 cM −1.4359 0.0317
10579508 NM_001164679 Ano8 anoctamin 8 8 B3.3 −1.4375 0.015
10431300 NM_001142357 Alg12 asparagine-linked glycosylation 12 homolog (yeast, alpha-1,6-mannosyltransferase) 15 E3 −1.4389 0.0334
10585874 NM_010421 Hexa hexosaminidase A 9 B|9 29.0 cM −1.4394 0.0245
10571302 NM_026432 Tmem66 transmembrane protein 66 8 A4 −1.4412 0.0307
10519652 NR_003596 Gm6455 predicted gene 6455 5 A1 −1.4455 0.0286
10397975 NM_026790 Ifi27l1 interferon, alpha-inducible protein 27 like 1 12 E|12 51.0 cM −1.4507 0.0331
10362904 NM_130892 Rtn4ip1 reticulon 4 interacting protein 1 10 B2|10 29.0 cM −1.4553 0.047
10352767 NM_010892 Nek2 NIMA (never in mitosis gene a)-related expressed kinase 2 1 H6|1 103.0 cM −1.4555 0.0145
10480699 NM_031843 Dpp7 dipeptidylpeptidase 7 2 A3 −1.4566 0.0363
10529454 ENSMUST00000069741 E130018O15Rik RIKEN cDNA E130018O15 gene 5 B3 −1.4683 0.0429
10600531 −1.4695 0.0218
10426812 NM_010271 Gpd1 glycerol-3-phosphate dehydrogenase 1 (soluble) 15 56.8 cM −1.4746 0.0375
10519688 NR_003596 Gm6455 predicted gene 6455 5 A1 −1.4804 0.0063
10518679 NM_133435 Nmnat1 nicotinamide nucleotide adenylyltransferase 1 4 E2 −1.4814 0.048
10357418 NM_001081078 Lct lactase 1 E4 −1.4902 0.0333
10559590 NM_207270 Ptprh protein tyrosine phosphatase, receptor type, H 7 A1 −1.494 0.0286
10525365 NM_001042489 Hvcn1 hydrogen voltage-gated channel 1 5 F −1.4944 0.0376
10424929 NM_172960 Adck5 aarF domain containing kinase 5 15 D3 −1.4964 0.043
10351473 NM_001033499 Sh2d1b2 SH2 domain protein 1B2 1 H3 −1.4988 0.0061
10547976 NM_145391 Tapbpl TAP binding protein-like 6 F3 −1.5013 0.041
10521391 NM_030721 Acox3 acyl-Coenzyme A oxidase 3, pristanoyl −1.5034 0.0098
10490104 NM_011497 Aurka aurora kinase A 2 H3|2 100.0 cM −1.5077 0.035
10411633 NM_008670 Naip1 NLR family, apoptosis inhibitory protein 1 13 D1-D3|13 54.0 cM −1.5078 0.0232
10450145 NM_013585 Psmb9 proteasome (prosome, macropain) subunit, beta type 9 (large multifunctional peptidase 2) 17 B1|17 18.59 cM −1.5167 0.045
10524681 NM_026933 Triap1 TP53 regulated inhibitor of apoptosis 1 5 F −1.517 0.0058
10490491 NM_008093 Gata5 GATA binding protein 5 2 H4|2 106.0 cM −1.5174 0.0361
10437655 NM_011955 Nubp1 nucleotide binding protein 1 16 A1|16 3.4 cM −1.5217 0.0461
10441530 NM_031395 Sytl3 synaptotagmin-like 3 17 A1 −1.5249 0.0443
10562130 NM_194057 Ffar1 free fatty acid receptor 1 7 B1 −1.526 0.0014
10463704 NM_020577 As3mt arsenic (+3 oxidation state) methyltransferase 19 D1 −1.5357 0.0162
10524878 NM_001033311 Vsig10 V-set and immunoglobulin domain containing 10 5 F −1.5404 0.0046
10393573 NM_011150 Lgals3bp lectin, galactoside-binding, soluble, 3 binding protein 11 E −1.5406 0.0119
10606714 NM_013463 Gla galactosidase, alpha X E-F1|X 53.0 cM −1.5428 0.0277
10458547 NR_028061 Gm8615 glucosamine-6-phosphate deaminase 1 pseudogene 5 G3 −1.5451 0.0246
10384378 NM_016672 Ddc dopa decarboxylase 11 A1-A4|11 7.0 cM −1.5472 0.043
10429638 XR_032493 Gm9568 glyceraldehyde-3-phosphate dehydrogenase pseudogene 15 D3|15 −1.5502 0.0482
10510516 NM_019741 Slc2a5 solute carrier family 2 (facilitated glucose transporter), member 5 4 E2 −1.5555 0.0065
10565634 NM_008663 Myo7a myosin VIIA 7 E2|7 48.1 cM −1.5718 0.0053
10524422 NM_010018 Dao D-amino acid oxidase 5 F|5 65.0 cM −1.575 0.0418
10566583 AK172683 Gm8995 predicted gene 8995 7 E3 −1.5789 0.0371
10391207 NM_030150 Dhx58 DEXH (Asp-Glu-X-His) box polypeptide 58 11 D|11 61.5 cM −1.5797 0.0135
10408879 NM_001033399 Gfod1 glucose-fructose oxidoreductase domain containing 1 13 A4 −1.5847 0.013
10505954 NM_013690 Tek endothelial-specific receptor tyrosine kinase 4 C5|4 43.6 cM −1.5877 0.0422
10538082 NM_133764 Atp6v0e2 ATPase, H+ transporting, lysosomal V0 subunit E2 6 B3 −1.5918 0.0041
10545958 NM_013471 Anxa4 annexin A4 6 D1|6 38.0 cM −1.596 0.0411
10560474 NM_177691 Ppm1n protein phosphatase, Mg2+/Mn2+ dependent, 1N (putative) 7 A3 −1.6066 0.0158
10577792 NM_031257 Plekha2 pleckstrin homology domain-containing, family A (phosphoinositide binding specific) member 2 8 A2 −1.623 0.0343
10455784 NM_026240 Gramd3 GRAM domain containing 3 18 D2 −1.6238 0.0361
10467907 NM_145502 Erlin1 ER lipid raft associated 1 19 C3 −1.6274 0.0115
10492102 NM_144895 Spg20 spastic paraplegia 20, spartin (Troyer syndrome) homolog (human) 3 C −1.6317 0.0301
10377927 NM_027445 Rnf167 ring finger protein 167 11 B4 −1.633 0.0209
10404439 NM_011452 Serpinb9b serine (or cysteine) peptidase inhibitor, clade B, member 9b 13 A3.3|13 12.5 cM −1.6346 0.0217
10566585 ENSMUST00000098144 Gm1966 predicted gene 1966 7 E3 −1.6387 0.0248
10587873 −1.6446 0.037
10439068 NM_145932 Osta organic solute transporter alpha 16 B3 −1.6448 0.035
10376726 NM_001172112 Dhrs7b dehydrogenase/reductase (SDR family) member 7B 11 B2 −1.6476 0.0062
10469951 NM_176834 Rnf208 ring finger protein 208 2 A3 −1.6491 0.0225
10548817 NM_025806 Plbd1 phospholipase B domain containing 1 6 G1 −1.654 0.0163
10473356 NM_019949 Ube2l6 ubiquitin-conjugating enzyme E2L 6 2 E1 −1.6544 0.0048
10501235 NM_026764 Gstm4 glutathione S-transferase, mu 4 3 F2.3 −1.6622 0.0309
10474825 NM_026412 D2Ertd750e DNA segment, Chr 2, ERATO Doi 750, expressed 2 E5 −1.6757 0.0095
10463716 NM_033569 Cnnm2 cyclin M2 19 C3|19 47.0 cM −1.6797 0.0023
10604337 −1.6798 0.0307
10595081 NM_012033 Tinag tubulointerstitial nephritis antigen 9 D −1.6828 0.0065
10524621 NM_011854 Oasl2 2'-5' oligoadenylate synthetase-like 2 5 F −1.687 0.0489
10444810 ENSMUST00000105041 H2-Q1 histocompatibility 2, Q region locus 1 17 B1|17 19.14 cM −1.6883 0.0032
10584334 NM_011734 Siae sialic acid acetylesterase 9 A4|9 19.0 cM −1.699 0.0032
10582985 NM_009807 Casp1 caspase 1 9 A1|9 1.0 cM −1.6991 0.0091
10488185 NM_009751 Bfsp1 beaded filament structural protein 1, in lens-CP94 2 G −1.7014 0.0096
10491091 NM_009425 Tnfsf10 tumor necrosis factor (ligand) superfamily, member 10 3 A3 −1.7131 0.0053
10583920 NM_026189 Eepd1 endonuclease/exonuclease/phosphatase family domain containing 1 9 A4 −1.7418 0.0179
10378572 NM_027249 Tlcd2 TLC domain containing 2 11 B5 −1.7499 0.0309
10383192 −1.7677 0.0205
10559606 NM_023440 Tmem86b transmembrane protein 86B 7 A1 −1.776 0.008
10550740 NM_027839 Ceacam20 carcinoembryonic antigen-related cell adhesion molecule 20 7 A3 −1.7837 0.0219
10486172 NM_001033136 Fam82a2 family with sequence similarity 82, member A2 2 E5 −1.7911 0.0045
10482802 NM_139200 Cytip cytohesin 1 interacting protein 2 C1.1 −1.798 0.0078
10402347 NM_029803 Ifi27l2a interferon, alpha-inducible protein 27 like 2A 12 E −1.807 0.0173
10519196 NM_147776 Vwa1 von Willebrand factor A domain containing 1 −1.8075 0.0079
10488636 NM_001039120 Defb26 defensin beta 26 2 H1 −1.8103 0.041
10542857 NM_178797 Far2 fatty acyl CoA reductase 2 6 G3 −1.8273 0.0308
10376832 NM_007413 Adora2b adenosine A2b receptor 11 B2 −1.8282 0.0194
10558265 NM_029609 Lhpp phospholysine phosphohistidine inorganic pyrophosphate phosphatase 7 F4 −1.8296 0.001
10569203 NM_001142681 Chid1 chitinase domain containing 1 7 F5 −1.8468 0.0112
10571984 NM_001081215 Ddx60 DEAD (Asp-Glu-Ala-Asp) box polypeptide 60 8 B3.1 −1.8805 0.05
10424221 NM_001081396 Wdr67 WD repeat domain 67 15 D1 −1.8879 0.0054
10364134 NM_133184 Slc5a4a solute carrier family 5, member 4a 10 C1 −1.9264 0.0251
10447904 NM_199252 Unc93a unc-93 homolog A (C. elegans) 17 A1|17 7.8 cM −1.9295 0.025
10539186 NM_011259 Reg3a regenerating islet-derived 3 alpha 6 C3|6 33.5 cM −1.9355 0.0286
10451287 NM_015783 Isg15 ISG15 ubiquitin-like modifier 4 E2|4 −1.936 0.013
10447634 NM_001142539 Gm9992 predicted gene 9992 17 A1 −1.9524 0.0175
10389261 NM_001037932 Gm11437 predicted gene 11437 11 C −1.9832 0.0076
10538590 NM_025992 Herc5 hect domain and RLD 5 6 C1|6 −1.9883 0.0089
10510532 NM_001085529 Slc2a7 solute carrier family 2 (facilitated glucose transporter), member 7 4 E2|4 −2.0053 0.0136
10385500 NM_008326 Irgm1 immunity-related GTPase family M member 1 11 B1.2 −2.0184 0.0234
10587871 NM_198414 Paqr9 progestin and adipoQ receptor family member IX 9 E3.3 −2.0207 0.0376
10568568 NM_016978 Oat ornithine aminotransferase 7 F3|7 63.0 cM −2.0471 3.00E−04
10467470 NM_019698 Aldh18a1 aldehyde dehydrogenase 18 family, member A1 19 C3 −2.3775 0.0079
10425287 NM_134090 Kdelr3 KDEL (Lys-Asp-Glu-Leu) endoplasmic reticulum protein retention receptor 3 15 E1 −2.4335 0.0115
10376324 NM_001135115 Gm12250 predicted gene 12250 11 B1.3 −2.4477 0.0311
10443869 NM_024442 Cyp4f16 cytochrome P450, family 4, subfamily f, polypeptide 16 17 B1 −2.5218 0.0265
10566571 NR_030719 Gm8979 very large inducible GTPase 1 pseudogene 7 E3 −2.6198 0.0145
10390748 NM_172564 Tns4 tensin 4 11 D −2.6277 0.0044
10462618 NM_010501 Ifit3 interferon-induced protein with tetratricopeptide repeats 3 19 C3 −2.6278 0.0357
10545200 ENSMUST00000101325 LOC100046894 similar to Igk-C protein −2.6477 0.0473
10499899 NM_009264 Sprr1a small proline-rich protein 1A 3 F1|3 45.2 cM −2.7972 0.0286
10483074 NM_008100 Gcg glucagon 2 C1.3|2 36.0 cM −2.7977 0.0019
10557300 NM_007474 Aqp8 aquaporin 8 7 F3|7 61.0 cM −2.7985 8.00E−04
10566578 NR_030719 Gm8979 very large inducible GTPase 1 pseudogene 7 E3 −2.8066 0.0095
10430006 NM_028064 Slc39a4 solute carrier family 39 (zinc transporter), member 4 15 D3 −2.8188 0.0145
10390691 NM_145434 Nr1d1 nuclear receptor subfamily 1, group D, member 1 11 D −3.298 3.00E−04
10480633 NM_080854 Slc34a3 solute carrier family 34 (sodium phosphate), member 3 2 A3 −3.3614 0.013
10566366 NM_199146 AI451617 expressed sequence AI451617 7 E3 −3.9279 0.0019
10505451 NM_011016 Orm2 orosomucoid 2 4 C1|4 31.4 cM 3.9755 0.0307
10416057 NM_013492 Clu clusterin 14 D1|14 28.0 cM 3.6483 0.0393
10452854 NM_053188 Srd5a2 steroid 5 alpha-reductase 2 17 E2 3.4167 7.00E−04
10367045 NM_009040 Rdh16 retinol dehydrogenase 16 10 D3 2.3984 0.0273
10523717 NM_009263 Spp1 secreted phosphoprotein 1 5 E5|5 56.0 cM 2.2985 0.0406
10464594 BC034269 BC021614 cDNA sequence BC021614 19 A 2.2581 0.011
10429140 NM_008681 Ndrg1 N-myc downstream regulated gene 1 15 D2 2.087 0.0273
10469609 NR_033225 Gm13375 predicted gene 13375 2 A3 2.0048 0.0294
10507671 NM_008190 Guca2a guanylate cyclase activator 2a (guanylin) 4 D2.1|4 57.0 cM 1.8922 0.0482
10556113 NM_016809 Rbm3 RNA binding motif protein 3 X A1.1|X 2.0 cM 1.8115 0.0271
10359917 NM_010476 Hsd17b7 hydroxysteroid (17-beta) dehydrogenase 7 1 H3 1.7626 0.0467
10358454 NM_001166409 Rbm3 RNA binding motif protein 3 X A1.1|X 2.0 cM 1.7444 0.0294
10540034 NM_027406 Aldh1l1 aldehyde dehydrogenase 1 family, member L1 6 D1 1.7157 0.015
10371784 NM_001163700 Nr1h4 nuclear receptor subfamily 1, group H, member 4 10 C2|10 50.0 cM 1.7001 0.0435
10603469 NM_001166409 Rbm3 RNA binding motif protein 3 X A1.1|X 2.0 cM 1.6579 0.0273
10582310 NM_138656 Mvd mevalonate (diphospho) decarboxylase 8 E1 1.6179 0.0287
10518568 1.5432 0.0336
10443898 NM_134127 Cyp4f15 cytochrome P450, family 4, subfamily f, polypeptide 15 17 B1 1.5311 0.0468
10344713 NM_016661 Ahcy S-adenosylhomocysteine hydrolase 2 H1|2 89.0 cM 1.5215 0.0425
10488816 NM_016661 Ahcy S-adenosylhomocysteine hydrolase 2 H1|2 89.0 cM 1.5127 0.0377
10439762 NM_016661 Ahcy S-adenosylhomocysteine hydrolase 2 H1|2 89.0 cM 1.5089 0.0468
10605055 NM_028633 Haus7 HAUS augmin-like complex, subunit 7 X A7.3 1.5083 0.015
10556169 NM_025344 Eif3f eukaryotic translation initiation factor 3, subunit F 7 E3 1.418 0.0317
10413542 NM_009388 Tkt transketolase 14 B1 1.4043 0.0425
10408850 NM_001111324 Nedd9 neural precursor cell expressed, developmentally down-regulated gene 9 13 A3.3-A4 −1.3853 0.0463
10489107 NM_018851 Samhd1 SAM domain and HD domain, 1 2 H2 −1.4139 0.0456
10516620 NM_010693 Lck lymphocyte protein tyrosine kinase 4 D2.2|4 59.0 cM −1.4259 0.0348
10513256 NM_010336 Lpar1 lysophosphatidic acid receptor 1 4 B3|4 16.0 cM −1.434 0.0294
10604057 NR_033443 6-Sep septin 6 X A2 −1.4532 0.0468
10588464 NR_029526 Mirlet7g microRNA let7g −1.464 0.0301
10432362 NM_026967 Rhebl1 Ras homolog enriched in brain like 1 15 F2 −1.49 0.017
10350173 NM_001130174 Tnnt2 troponin T2, cardiac 1 E4|1 60.0 cM −1.4944 0.0425
10547906 NM_008479 Lag3 lymphocyte-activation gene 3 6 F2 −1.5108 0.0346
10420659 NM_025697 6330409N04Rik RIKEN cDNA 6330409N04 gene 14 D1 −1.5451 0.0354
10352097 NM_027077 1700016C15Rik RIKEN cDNA 1700016C15 gene 1 H3 −1.5503 0.0468
10409579 NM_019568 Cxcl14 chemokine (C-X-C motif) ligand 14 13 B1 −1.5799 0.0377
10355960 NM_009129 Scg2 secretogranin II 1 C4|1 43.6 cM −1.6019 0.0178
10478633 NM_013599 Mmp9 matrix metallopeptidase 9 2 H1-H2|2 96.0 cM −1.6069 0.0228
10501629 NM_001080818 Cdc14a CDC14 cell division cycle 14 homolog A (S. cerevisiae) 3 G1 −1.6217 0.0425
10447056 NM_027455 Qpct glutaminyl-peptide cyclotransferase (glutaminyl cyclase) 17 E3 −1.6326 0.0228
10407940 −1.6681 0.0294
10605355 −1.6826 0.0494
10593015 NM_009850 Cd3g CD3 antigen, gamma polypeptide 9 A5.2|9 26.0 cM −1.6893 0.0161
10464999 NM_028623 Cst6 cystatin E/M 19 A|19 4.0 cM −1.7219 0.0456
10584821 NM_013487 Cd3d CD3 antigen, delta polypeptide 9 A5.2|9 26.0 cM −1.7359 0.0435
10561927 NM_007467 Aplp1 amyloid beta (A4) precursor-like protein 1 7 B1|7 8.0 cM −1.7367 0.0109
10552406 NM_024253 Nkg7 natural killer cell group 7 sequence 7 B2 −1.7501 0.0178
10366881 NM_007837 Ddit3 DNA-damage inducible transcript 3 10 D3 −1.7621 0.0344
10490923 NM_009801 Car2 carbonic anhydrase 2 3 A1|3 10.5 cM −1.7726 0.0189
10428534 NM_032000 Trps1 trichorhinophalangeal syndrome I (human) 15 C|15 30.1 cM −1.793 0.0494
10403821 ENSMUST00000103558 Tcrg-V3 T-cell receptor gamma, variable 3 −1.8213 0.0468
10516093 NM_007558 Bmp8a bone morphogenetic protein 8a 4 D2.2|4 57.4 cM −1.8392 0.0307
10513739 NM_011607 Tnc tenascin C 4 C1|4 32.2 cM −1.853 0.0348
10574166 NM_153507 Cpne2 copine II 8 C5 −1.857 0.0346
10542632 NM_010491 Iapp islet amyloid polypeptide 6 G2|6 62.0 cM −1.8827 0.0301
10541605 NM_020001 Clec4n C-type lectin domain family 4, member n 6 F3|6 55.0 cM −1.889 0.017
10402325 NM_023049 Asb2 ankyrin repeat and SOCS box-containing 2 12 E|12 50.0 cM −1.9087 0.0492
10466200 NM_027836 Ms4a7 membrane-spanning 4-domains, subfamily A, member 7 19 A −1.9359 0.0172
10404913 NM_026056 Cap2 CAP, adenylate cyclase-associated protein, 2 (yeast) 13 A5 −1.9619 0.0346
10420308 NM_013542 Gzmb granzyme B 14 D3|14 20.5 cM −1.9705 0.0294
10412211 NM_010370 Gzma granzyme A 13 D|13 64.0 cM −2.0182 0.0229
10463016 NM_028191 Cyp2c65 cytochrome P450, family 2, subfamily c, polypeptide 65 19 C3 −2.1252 0.0456
10551197 NM_009999 Cyp2b10 cytochrome P450, family 2, subfamily b, polypeptide 10 7 A3|7 6.5 cM −2.5753 0.0138
10512999 NR_033139 AI427809 expressed sequence AI427809 4 B2 −2.5806 0.0109
10405063 NM_008760 Ogn osteoglycin 13 A5 −2.6878 0.0346
10512949 NM_013454 Abca1 ATP-binding cassette, sub-family A (ABC1), member 1 4 A5-B3|4 23.1 cM −2.753 0.0273
10366043 NM_026268 Dusp6 dual specificity phosphatase 6 10 C3 −3.0369 0.0224
10534493 NM_019577 Ccl24 chemokine (C-C motif) ligand 24 5 G1 −4.9474 0.0072
10463023 NM_001011707 Cyp2c66 cytochrome P450, family 2, subfamily c, polypeptide 66 19 C3 −5.082 0.015

Conflict of Interest

None declared.

References

  1. Alaish SM, Torres M, Ferlito M, Sun C, De Maio A. The severity of cholestatic injury is modulated by the genetic background. Shock. 2005;24:412–416. doi: 10.1097/01.shk.0000183392.83272.97. [DOI] [PubMed] [Google Scholar]
  2. Alaish SM, Smith AD, Timmons J, Greenspon J, Eyvazzadeh D, Murphy E, et al. Gut microbiota, tight junction protein expression, intestinal resistance, bacterial translocation and mortality following cholestasis depend on the genetic background of the host. Gut Microbes. 2013;4:1–14. doi: 10.4161/gmic.24706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Bacher A, Griebl K, Mackamul S, Mitreiter R, Muckter H, Ben-Shaul Y. Protease inhibitors suppress the formation of tight junctions in gastrointestinal cell lines. Exp. Cell Res. 1992;200:97–104. doi: 10.1016/s0014-4827(05)80076-x. [DOI] [PubMed] [Google Scholar]
  4. Beliveau F, Desilets A, Leduc R. Probing the substrate specificities of matriptase, matriptase-2, hepsin, and DESC1 with internally quenched fluorescent peptides. FEBS J. 2009;276:2213–2226. doi: 10.1111/j.1742-4658.2009.06950.x. [DOI] [PubMed] [Google Scholar]
  5. Berger T, Togawa A, Duncan GS, Elia AJ, You-Ten A, Wakeham A, et al. Lipocalin-2-deficient mice exhibit increased sensitivity to Escherichia coli infection but not ischemia-reperfusion injury. PNAS. 2006;103:1834–1839. doi: 10.1073/pnas.0510847103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bruewer M, Luegering A, Kucharzik T, Parkos CA, Madara JL, Hopkins AM, et al. Proinflammatory cytokines disrupt epithelial barrier function by apoptosis-independent mechanisms. J. Immunol. 2003;171:6164–6172. doi: 10.4049/jimmunol.171.11.6164. [DOI] [PubMed] [Google Scholar]
  7. Buzza MS, Netzel-Arnett S, Shea-Donahue T, Zhao A, Lin C-Y, List K, et al. Membrane-anchored serine protease matriptase regulates epithelial barrier formation and permeability in the intestine. PNAS. 2010;107:4200–4205. doi: 10.1073/pnas.0903923107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Byrne TA, Persinger RL, Young LS, Ziegler TR, Wilmore DW. A new treatment for patients with short-bowel syndrome: growth hormone, glutamine and a modified diet. Ann. Surg. 1995;222:243–254. doi: 10.1097/00000658-199509000-00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Caglikulekci M, Dirlik M, Scatton O, Cinel I, Ozer I, Cinel L, et al. Effect of antithrombin-III (AT-III) on intestinal epithelium changes related to obstructive icterus: experimental study in rats. Ann. Chir. 2004;129:273–277. doi: 10.1016/j.anchir.2004.01.008. [DOI] [PubMed] [Google Scholar]
  10. Campillo B, Pernet P, Bories PN, Richardet JP, Devanlay M, Aussel C. Intestinal permeability in liver cirrhosis: relationship with severe septic complications. Eur. J. Gastroenterol. Hepatol. 1999;11:755–759. doi: 10.1097/00042737-199907000-00013. [DOI] [PubMed] [Google Scholar]
  11. Clayburgh DR, Shen L, Turner JR. A porous defense: the leaky epithelial barrier in intestinal disease. Lab. Invest. 2004;84:282–291. doi: 10.1038/labinvest.3700050. [DOI] [PubMed] [Google Scholar]
  12. Collins CB, Aherne CM, Ehrentraut SF, Gerich ME, McNamee EN, McManus MC, et al. Alpha-1-antitrypsin therapy ameliorates acute colitis and chronic murine ileitis. Inflamm. Bowel Dis. 2013;19:1964–1973. doi: 10.1097/MIB.0b013e31829292aa. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Devireddy LR, Teodoro JG, Richard FA, Green MR. Induction of apoptosis by a secreted lipocalin that is transcriptionally regulated by IL-3 deprivation. Science. 2001;293:829–834. doi: 10.1126/science.1061075. [DOI] [PubMed] [Google Scholar]
  14. Diller R, Stratmann U, Minin E, Baumer C, von Eiff G, Huismans H, et al. ATIII attenuates endotoxemia induced healing impairment in the colon. J. Surg. Res. 2009;157:4–13. doi: 10.1016/j.jss.2008.10.008. [DOI] [PubMed] [Google Scholar]
  15. Ding L, Li J-S, Li Y-S, Liu FN, Tan L. Prophylactic treatment with growth hormone improves intestinal barrier function and alleviates bacterial translocation in stressed rats. Chin. Med. J. 2004;117:264–269. [PubMed] [Google Scholar]
  16. Dorsey SG, Leitch CC, Renn CL, Lessans S, Smith BA, Wang XM, et al. Genome-wide screen identifies drug-induced regulation of the gene giant axonal neuropathy (Gan) in a mouse model of antiretroviral-induced painful peripheral neuropathy. Biol. Res. Nurs. 2009;11:7–16. doi: 10.1177/1099800409332726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. El Asmar R, Panigrahi P, Bamford P, Berti I, Not T, Coppa GV, et al. Host-dependent zonulin secretion causes the impairment of the small intestine barrier function after bacterial exposure. Gastroenterology. 2002;123:1607–1615. doi: 10.1053/gast.2002.36578. [DOI] [PubMed] [Google Scholar]
  18. Flo TH, Smith KD, Sato S, Rodriquez DJ, Holmes MA, Strong RK, et al. Lipocalin 2 mediates an innate immune response to bacterial infection by sequestering iron. Nature. 2004;432:917–921. doi: 10.1038/nature03104. [DOI] [PubMed] [Google Scholar]
  19. Frances R, Benlloch S, Zapater P, Gonzalez JM, Lozano B, Munoz C, et al. A sequential study of serum bacterial DNA in patients with advanced cirrhosis and ascites. Hepatology. 2004;39:484–491. doi: 10.1002/hep.20055. [DOI] [PubMed] [Google Scholar]
  20. Georgiev P, Jochum W, Heinrich S, Jang JH, Nocito A, Dahm F, et al. Characterization of time-related changes after experimental bile duct ligation. Br. J. Surg. 2008;95:646–656. doi: 10.1002/bjs.6050. [DOI] [PubMed] [Google Scholar]
  21. Huang C-Y, Sheen-Chen S-M, Ho H-T, Tang RP, Eng HL. Antithrombin-III attenuates hepatocyte apoptosis in bile duct ligated rat: a striking cellular change. Surg. Innov. 2010;17:132–135. doi: 10.1177/1553350610366716. [DOI] [PubMed] [Google Scholar]
  22. Javid PJ, Collier S, Richardson D, Iglesias J, Gura K, Lo C, et al. The role of enteral nutrition in the reversal of parenteral-nutrition-associated liver dysfunction in infants. J. Pediatr. Surg. 2005;40:1015–1018. doi: 10.1016/j.jpedsurg.2005.03.019. [DOI] [PubMed] [Google Scholar]
  23. Kato Y, Yu D, Lukish JR, Schwartz MZ. Hepatocyte growth factor enhances intestinal mucosal cell function and mass in vivo. J. Pediatr. Surg. 1997;32:991–994. doi: 10.1016/s0022-3468(97)90384-5. [DOI] [PubMed] [Google Scholar]
  24. Kato Y, Yu D, Schwartz MZ. Enhancement of intestinal adaptation by hepatocyte growth factor. J. Pediatr. Surg. 1998;33:235–239. doi: 10.1016/s0022-3468(98)90438-9. [DOI] [PubMed] [Google Scholar]
  25. Kikuchi T, Abe T, Yaekashiwa M, Tominaga Y, Mitsuhashi H, Satoh K, et al. Secretory leukoprotease inhibitor augments hepatocyte growth factor production in human lung fibroblasts. Am. J. Respir. Cell Mol. Biol. 2000;23:364–370. doi: 10.1165/ajrcmb.23.3.3942. [DOI] [PubMed] [Google Scholar]
  26. Kuhn NJ, Woodworth-Gutai M, Gross KW, Held WA. Subfamilies of the mouse major urinary protein (MUP) multi-gene family: sequence analysis of cDNA clones and differential regulation in the liver. Nucleic Acids Res. 1984;12:6073–6090. doi: 10.1093/nar/12.15.6073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. List K, Currie B, Scharschmidt TC, Szabo R, Shireman J, Molinolo A, et al. Autosomal ichthyosis with hypotrichosis syndrome displays low matriptase proteolytic activity and is phenocopied in ST14 hypomorphic mice. J. Biol. Chem. 2007;282:36714–36723. doi: 10.1074/jbc.M705521200. [DOI] [PubMed] [Google Scholar]
  28. Liu W, Jiang Z, Wang X, Shu H, Cui W, Wilmore DW. Impact of perioperative treatment of recombinant human growth hormone on cell immune function and intestinal barrier function: randomized, double-blind, controlled trial. World J. Surg. 2003;27:412–415. doi: 10.1007/s00268-002-6758-x. [DOI] [PubMed] [Google Scholar]
  29. Madara JL, Stafford J. Interferon-gamma directly affects barrier function of cultured intestinal epithelial monolayers. J. Clin. Invest. 1989;83:724–727. doi: 10.1172/JCI113938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Marano CW, Lewis SA, Garulacan LA, Soler AP, Mullin JM. Tumor necrosis factor-alpha increases sodium and chloride conductance across the tight junction of CACO-2 BBE, a human intestina epithelial cell line. J. Membr. Biol. 1998;161:263–274. doi: 10.1007/s002329900333. [DOI] [PubMed] [Google Scholar]
  31. Pascual S, Such J, Esteban A, Zapater P, Casellas JA, Aparicio JR, et al. Intestinal permeability is increased in patients with advanced cirrhosis. Hepatogastroenterology. 2003;50:1482–1486. [PubMed] [Google Scholar]
  32. Rafatellu M, George MD, Akiyama Y, Hornsby MJ, Nuccio SP, Paixao TA, et al. Lipocalin-2 resistance confers an advantage to Salmonella enterica serotype Typhimurium for growth and survival in the inflamed intestine. Cell Host Microbe. 2009;5:476–486. doi: 10.1016/j.chom.2009.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Rao JN, Platoshyn O, Golovina VA, Liu L, Zou T, Marasa BS, et al. TRPC1 functions as a store-operated Ca2 + channel in intestinal epithelial cells and regulates early mucosal restitution after wounding. Am. J. Physiol. Gastrointest. Liver Physiol. 2006;290:G782–G792. doi: 10.1152/ajpgi.00441.2005. [DOI] [PubMed] [Google Scholar]
  34. Scopa CD, Koureleas S, Tsamandas AC, Spiliopoulou I, Alexandrides T, Filos KS, et al. Beneficial effects of growth hormone and insulin-like growth factor I on intestinal bacterial translocation, endotoxemia, and apoptosis in experimentally jaundiced rats. J. Am. Coll. Surg. 2000;190:423–431. doi: 10.1016/s1072-7515(99)00285-9. [DOI] [PubMed] [Google Scholar]
  35. Ulshen MH, Dowling RH, Fuller CR, Zimmermann EM, Lund PK. Enhanced growth of small bowel in transgenic mice overexpressing bovine growth hormone. Gastroenterology. 1993;104:973–980. doi: 10.1016/0016-5085(93)90263-c. [DOI] [PubMed] [Google Scholar]
  36. Velasco B, Lassaletta L, Gracia R, Tovar JA. Intestinal lengthening and growth hormone in extreme short bowel syndrome: a case report. J. Pediatr. Surg. 1999;34:1423–1424. doi: 10.1016/s0022-3468(99)90027-1. [DOI] [PubMed] [Google Scholar]
  37. Weiming Z, Ning L, Jieshou L. Effect of recombinant human growth hormone and enteral nutrition on short bowel syndrome. JPEN J. Parenter. Enteral Nutr. 2004;28:377–381. doi: 10.1177/0148607104028006377. [DOI] [PubMed] [Google Scholar]
  38. Wildhaber BE, Yang H, Coran AG, Teitelbaum DH. Gene alteration of intestinal intraepithelial lymphocytes in response to massive small bowel resection. Pediatr. Surg. Int. 2003;19:310–315. doi: 10.1007/s00383-003-1001-x. [DOI] [PubMed] [Google Scholar]
  39. Yamanouchi H, Fujita J, Hojo S, Yoshinouchi T, Kamei T, Yamadori I, et al. Neutrophil elastase: alpha-1-proteinase inhibitor complex in serum and bronchoalveolar lavage fluid in patients with pulmonary fibrosis. Eur. Respir. J. 1998;11:120–125. doi: 10.1183/09031936.98.11010120. [DOI] [PubMed] [Google Scholar]
  40. Yi C, Cao Y, Wang SR, Xu YZ, Huang H, Cui YX, et al. Beneficial effect of recombinant human growth hormone on the intestinal mucosa barrier of septic rats. Braz. J. Med. Biol. Res. 2007;40:41–48. doi: 10.1590/s0100-879x2007000100006. [DOI] [PubMed] [Google Scholar]
  41. Youakim A, Ahdieh M. Interferon-gamma decreases barrier function in T84 cells by reducing ZO-1 levels and disrupting apical actin. Am. J. Physiol. 1999;276:G1279–G1288. doi: 10.1152/ajpgi.1999.276.5.G1279. [DOI] [PubMed] [Google Scholar]
  42. Zhou Y, Jiang L, Rui L. Identification of MUP1 as a regulator for glucose and lipid metabolism in mice. J. Biol. Chem. 2009;284:11152–11159. doi: 10.1074/jbc.M900754200. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Physiological Reports are provided here courtesy of Wiley

RESOURCES