Heterotypic mouse models of canine osteosarcoma recapitulate tumor heterogeneity and biological behavior


Milcah C. Scott, Hirotaka Tomiyasu, John R. Garbe, Ingrid Cornax, Clarissa Amaya, M. Gerard O'Sullivan, Subbaya Subramanian, Brad A. Bryan, Jaime F. Modiano "Hetero- typic mouse models of canine osteosarcoma recapitulate tumor heterogeneity and biological behavior",


ABSTRACT

Osteosarcoma (OS) is a heterogeneous and rare disease with a disproportionate impact because it mainly affects children and adolescents. Lamentably, more than half of patients with OS succumb to metastatic disease. Clarification of the etiology of the disease, development of better strategies to manage progression, and methods to guide personalized treatments are among the unmet health needs for OS patients. Progress in managing the disease has been hindered by the extreme heterogeneity of OS; thus, better models that accurately recapitulate the natural heterogeneity of the disease are needed. For this study, we used cell lines derived from two spontaneous canine OS tumors with distinctly different biological behavior (OS-1 and OS-2) for heterotypic in vivo modeling that recapitulates the heterogeneous biology and behavior of this disease. Both cell lines demonstrated stability of the transcriptome when grown as orthotopic xenografts in athymic nude mice. Consistent with the behavior of the original tumors, OS-2 xenografts grew more rapidly at the primary site and had greater propensity to disseminate to lung and establish microscopic metastasis. Moreover, OS-2 promoted formation of a different tumor-associated stromal environment than OS-1 xenografts. OS-2-derived tumors comprised a larger percentage of the xenograft tumors than OS-1-derived tumors. In addition, a robust pro-inflammatory population dominated the stromal cell infiltrates in OS-2 xenografts, whereas a mesenchymal population with a gene signature reflecting myogenic signaling dominated those in the OS-1 xenografts. Our studies show that canine OS cell lines maintain intrinsic features of the tumors from which they were derived and recapitulate the heterogeneous biology and behavior of bone cancer in mouse models. This system provides a resource to understand essential interactions between tumor cells and the stromal environment that drive the progression and metastatic propensity of OS.

 

INTRODUCTION

Osteosarcoma (OS) is the most common malignant pediatric tumor of bone (Kansara and Thomas, 2007; Mirabello et al., 2009). Standard therapy for OS comprises neoadjuvant chemotherapy, surgery and adjuvant chemotherapy (Jaffe, 2014). The 5-year survival rates of OS patients with localized and operable OS is 60-70%, but the outcome of patients with non-resectable or metastatic OS is poor (Bielack et al., 2002; Kumta, Jan-Apr, 2016). These collective statistics belie the extreme heterogeneity of OS (Martin et al., 2012; Tan et al., 2009). Neither the histological appearance nor the propensity of the tumor cells to elaborate bone, cartilage or collagen matrices are predictive of behavior and, although recurrent molecular events have been described (Sarver et al., 2013; Scott et al., 2011; Thayanithy et al., 2012), these are yet to be adopted as prognostic or predictive biomarkers for this disease. Thus, a better understanding of the events that underlie OS tumor heterogeneity and contribute to disease progression is needed to develop effective strategies to manage OS and to improve outcomes.

OS is also the most common primary malignant tumor of bone in dogs, and it is particularly prevalent in large and giant breeds (Morello et al., 2011). In contrast to humans, OS occurs most commonly in older dogs (Fenger et al., 2014; Varshney et al., 2016). Within the extensive heterogeneity that is characteristic of both canine and human OS, important clinical and pathological features are conserved between the two species (Fenger et al., 2014; Varshney et al., 2016; Withrow et al., 1991). Adding to the weight of evidence for spontaneous OS as a homologous cellular and molecular disease of humans and dogs, we have uncovered prognostically significant gene and microRNA expression signatures that are evolutionarily conserved in human and canine OS (Sarver et al., 2013; Scott et al., 2011; Thayanithy et al., 2012).

Understanding the heterogeneous biology and behavior of OS is important to fully elucidate the pathogenesis of this disease. Most studies to date in the area of OS have been focused on studying genetic alterations. Tumor-associated stroma has remained an underrepresented area of OS research, but has been described in recent years as being complicit in the progression of other tumor types and is also an important consideration of recent anti-tumor strategies (Kang, 2016; Kawada, 2016; Sleeman, 2012; Wang et al., 2016). Robust experimental animal models that recapitulate the natural heterogeneity of OS are essential to gain insights into tumor–stromal interactions that might contribute to tumor progression, and to discover ways in which these interactions might be countered. A number of syngeneic, autochthonous and xenograft models of OS have been established in laboratory mice (Chaffee and Allen, 2013; Coomer et al., 2009; Jaroensong et al., 2012; Kanaya et al., 2011; Mohseny et al., 2012; Sampson et al., 2013; Sottnik et al., 2011; Wolfe et al., 2011), and, although many of these have examined OS pathogenesis and the effects of specific therapeutic regimens in vivo, few have addressed OS heterogeneity and biological behavior in the context of tumor-associated stroma and tumor–stromal interactions.

Here, we document that orthotopic canine OS xenografts preserve the biological, molecular and heterotypic biology observed in the tumors from which they were derived. Moreover, transcriptome analysis of xenograft tumors revealed a strong OS-cell-specific stromal response, which provides evidence that intrinsic genetic tumor characteristics and crosstalk between tumor and stromal cells might underlie heterogeneity of biological behavior in individuals with OS. These data provide insight into tumor–host interactions and identify targets that could play a role in treatment strategies for OS patients.

 

RESULTS

Differential growth rates at the primary site in orthotopic canine OS-1 and OS-2 xenografts

Development and progression of primary tumors were examined using in vivo imaging starting 6 h after orthotopic cell injections and then weekly for the duration of the study (Fig. 1A). Luciferase activity was detectable within 6 h in virtually all of the mice receiving OS-1 or OS-2 cells, and all of the mice showed disease progression over time. Expansion of tumor cells can be inferred from the increased luciferase emission over time; Fig. 1B shows that OS-2 intratibial xenografts had grown significantly faster than OS-1 intratibial xenografts by day 22, and this difference persisted until day 50. The results in Fig. 1C encompass a more complex process, because the physical size of the tumors in the proximal tibia would be influenced by infiltrating host stromal cells and swelling. The data confirm that OS-2 intratibial xenografts grew significantly faster than OS-1 intratibial xenografts, albeit that the effect was delayed (detectable by day 29), with this relative difference persisting until day 50 (Fig. 1B,C, Table S1). It is worth noting that neither the indirect imaging measurements nor the direct physical measurements can account for tumor invasion and loss of periosteal integrity, as is described below. Nevertheless, the data shown in Fig. 1 and Table S1 allowed us to determine that disease progression was significantly faster in animals harboring OS-2 xenografts than in animals harboring OS-1 xenografts.

 

Fig. 1.: Orthotopic canine OS-1 and OS-2 xenografts show differential growth rates at the primary site. Athymic nude mice were injected with canine OS-1 or OS-2 cells orthotopically in the left tibia and tumor progression at the primary site was monitored by in vivo imaging and caliper measurements. (A) Representative examples of luciferase activity at the orthotopic site in five mice at 6 h (day 1), 4 weeks (day 29) and 8 weeks (day 57) after injection with OS-1 or OS-2 cells. Time exposures from the images for each group and from each week were different, but the radiance was adjusted to show equivalent scales in the composite. Data from the same mice that received OS-1 are shown in this figure and in Fig. 2A for day 1, but the light emission scale (in radiance=photons/sec) is adjusted in this figure to appreciate luminescence from the tumors in bone (tibiae). (B) Scatter plot showing luciferase activity for the mice in the experiment shown in panel A over time. (C) Scatter plot showing the volume of the orthotopic tumor in the left proximal tibia (minus to the volume of the unaffected, contralateral tibia) for all of the mice with orthotopic canine OS xenografts (16 mice injected with OS-1 cells and 32 mice injected with OS-2 cells) over time. Mice in B and C that received OS-1 are represented by the light symbols, and those that received OS-2 are represented by the dark symbols. The findings were analyzed with Student's t-test and the Holm–Sidak approach was used for multiple comparisons. Two-tailed test P-values are given. Significantly different growth rates between groups are denoted by *P< 0.01, **P< 0.001, ***P< 0.0001.

 

Differential metastatic propensity in orthotopic canine OS-1 and OS-2 xenografts

We observed luciferase activity in the lungs of mice receiving intratibial OS-2 cells, but not in mice injected with OS-1 cells, within 6 h of injections (Fig. 2A). We interpreted this as evidence of systemic dissemination of OS-2 cells with accumulation in the lungs. The luciferase signal disappeared from the lungs within 1 week after tumor administration, but the presence of OS-2 cells was evident focally in the lungs of one mouse from this group again within 2 weeks after tumor administration, and the luciferase activity in this area continued to increase until the last day imaging was done for the experiment (day 49; Fig. 2B). When the mice from all the experiments were considered together, OS-2 cells achieved metastatic dissemination more rapidly than OS-1 cells (by 15, 22 and 29 days), although the rate of microscopic and macroscopic metastasis between the two groups when the experiments were terminated were not different based on imaging on day 49 (P=0.35) or histopathology on day 57 (P=0.77; Table 1).

 

Fig. 2.:Orthotopic canine OS-1 and OS-2 xenografts have differential metastatic propensity. (A) Representative examples of luciferase activity at the primary site and in the lungs of five mice at both 6 h (day 1) and 8 days after intratibial injection of OS-1 and OS-2 cells. The time exposure for each image was different but, in every case, the radiance represents the same scale in the composite figure. Data from the same mice that received OS-1 are shown in Fig. 1A and in this figure for day 1, but the light emission scale (in radiance=photons/sec) is adjusted in this figure to appreciate luminescence from the tumors in lungs. A different group of mice than in Fig. 1A is shown in this figure to represent the transit of OS-2 cells to the lung. (B) Luciferase activity in the same mice shown in panel A 1 week, 2 weeks and 7 weeks after injection. The time exposure for each image was different but, in every case, the radiance represents the same scale in the composite figure. Light emission in the radii of one mouse with OS-1 and three mice with OS-2 was due to reflections from the tibial tumors; no dissemination of osteosarcoma cells was detected in the radii of any mice by histopathological examination. Data from the same mice that received OS-1 are shown in panel A and panel B for day 8, but the light emission scale in panel B is adjusted to appreciate if there was an increase in luminescence from tumors in the lungs. Different groups of mice receiving OS-2 are shown in panels A and B. The signal in the lungs at 6 h was seen consistently in mice receiving intratibial OS-2, but only one of 32 mice developed visible metastasis to the lungs by day 15. The light emission scale in panel B is adjusted to appreciate the increase in luminescence from tumors in the lungs.

 

Primary and metastatic tumors derived from orthotopic implantation of OS-1 and OS-2 cells show histological features and organization that are characteristic of canine OS

All of the mice injected with OS-1 or OS-2 cells had evidence of gross tumor burden in the proximal tibia at necropsy on the eighth week after injection (Fig. 3A,B). Histologically, OS-1-derived tumor xenografts were characterized by relatively well-differentiated, polygonal to spindle-shaped cells that had round to oval nuclei, mild to moderate anisocytosis and anisokaryosis, and infrequent mitotic activity (Fig. 3C,E). These tumors contained organized osteoid ribbons and showed limited destruction of cortical bone and epiphyseal invasion (Fig. 3C).

 

Fig. 3.: Primary and metastatic tumors derived from orthotopic implantation of OS-1 and OS-2 cells show histological features and organization that are characteristic of canine OS. (A,B) Images show the gross appearance of the legs from one representative mouse receiving either OS-1 (A) or OS-2 (B) cells. (C,D) Images show low-power photomicrographs of representative tumors at the primary site formed by OS-1 and OS-2 cells, respectively. The open star in C denotes an example of organized osteoid ribbons, and in D marks an example of necrosis, and arrows denote destruction of cortical bone with invasion of the epiphysis in both images. (E,F) Images show high-power photomicrographs of the tumors in C and D. Arrowheads denote mitotic figures. (G,H) Images show low-power photomicrographs of the lungs from mice receiving OS-1 (G) or OS-2 (H) cells. Areas of extensive necrosis and the pale eosinophilic matrix characteristic of OS-2 tumors are indicated by the asterisk and by the open star in H. (I,J) Images show high-power photomicrographs of the lungs from G and H. The open star denotes the pale, eosinophilic matrix characteristic of OS-2 tumors, and arrowheads in J indicate mitotic figures. Scale bars: 250 µm (C,D), 50 µm (E,F,I,J) and 500 µm (G,H).

 

In contrast, OS-2 tumors had a more aggressive appearance, with spindle-shaped, anaplastic cells that had round to elongate nuclei, moderate anisocytosis and anisokaryosis, and frequent mitotic activity (Fig. 3D,F). The cells in these tumors were embedded in a poorly organized, pale eosinophilic matrix and they showed extensive necrosis with marked destruction of cortical bone and epiphyseal invasion (Fig. 3D).

The different metastatic propensities of OS-1 and OS-2 were confirmed histologically (Table 1, Fig. 3G-J). Fewer than 20% of the mice injected with OS-2 and 7% of the mice injected with OS-1 developed metastasis by day 36 (an example of lungs without metastasis from a mouse injected orthotopically with OS-1 is illustrated by the photomicrographs shown in Fig. 3G,I). When lung metastasis was present, the histological appearance of the metastatic tumors recapitulated that of the parent tumors (Fig. 3D,F), as illustrated by the photomicrographs on one mouse receiving OS-2 orthotopically in Fig. 3H and J. In these animals, the morphology and mitotic activity of the cells and their residence in a poorly organized, pale eosinophilic matrix with extensive areas of necrosis and frequent mitotic activity were comparable to that seen in the primary tumors.

 

Gene signatures of tumor cells in OS xenografts resemble those of parent cell lines

One obstacle to using xenograft models to understand the heterogeneity of genetically complex tumors is the presumption that these tumors are unstable and will drift rapidly as they adapt to the host microenvironment. Indeed, previous data suggest that altered genomic signatures due to tumor cell plasticity and/or harsh clonal selection lead to unpredictable behavior of tumor cell lines after being transplanted into mice (Creighton et al., 2003; He et al., 2010; Hollingshead et al., 2014; Shin et al., 2013). Here, we used RNA sequencing to examine the stability of key transcriptomic properties between the parental OS cell lines and their corresponding tumor xenografts. The tumor xenografts were more similar to their corresponding parent cell lines than to each other or to the alternative cell line based on principal components analysis (data not shown) and by unsupervised clustering (Fig. 4), where tumor xenografts were assigned to the same group as their corresponding parent cell line based on the expression signatures from canine genes. When dog and mouse genes were analyzed together, expression of mouse-specific genes was not detected in the canine cell lines (Fig. S1), indicating that the mouse genes present in the tumor xenograft tissues could be accurately differentiated from the dog genes using our comparative bioinformatics approach. Furthermore, significantly larger numbers of mouse genes were detectable in OS-2 than in OS-1 xenografts, suggesting that the former tumors were more heavily infiltrated by host stroma (Fig. S1).

 

Fig. 4.: Gene signatures of parent tumor cell lines maintained in OS-1 and OS-2 xenograft tumors. 24,579 total canine genes were filtered to remove genes that did not have a log2 counts per million (CPM) mean-centered value ≥1 in at least two samples. 13,141 genes remained after filtering. The heatmap represents clustered gene-level counts with lower than mean (blue), higher than the mean (red), and mean (gray) levels of expression. Each row represents a single gene. The dendrogram represents the distance or dissimilarity between sample clusters, calculated using unsupervised hierarchical clustering on CPM values for the 13,141 filtered genes. In this dendrogram, there are two sample clusters as two branches that occur at about the same vertical distance. One of the sample clusters consists of four OS-1 (blue) xenograft tumors (black) and two parental cell line replicates (yellow), and one of these clusters consists of four OS-2 (gray) xenograft tumors (black) and two parental cell line replicates (yellow). All replicates are biological replicates.

 

OS-1 and OS-2 xenografts promote distinct tumor-associated stromal environments

To determine the nature of the stromal interactions and the identity of the infiltrating cells in the xenografts, we performed pairwise Fisher's exact test comparisons, with trimmed mean of M-values (TMM) normalization of gene counts, to identify the differentially expressed murine genes in tumors from each group (OS-1 and OS-2). Using a false discovery rate (FDR)-adjusted P-value of <0.005 and log2 fold change >2, we identified 482 genes that were expressed at significantly different levels between the two groups (Fig. 5A; Table S2). Pathway analysis of these 482 differentially expressed murine genes was done by MetaCore software. The top ten most enriched pathways suggest immune and inflammatory themes that modulate IL-17, TGF-β signaling, the complement system, and patterning behavior and cytoskeletal remodeling with involvement of Rho GTPases (Table 2).

 

Fig. 5.: Differentially expressed genes in OS xenografts uncover a propensity for differential stromal cell infiltrates. EdgeR was used for pair-wise Fisher's exact test comparisons, with TMM normalization, to identify differentially expressed murine genes in tumor xenografts. Four biological replicates were used for each OS subtype. Common dispersion across all genes was calculated as 0.079 and the biological coefficient of variation (BCV) as 0.23. Mean tag-wise dispersion (individual dispersion for each gene) was calculated as 0.095. Using statistical significance criteria of FDR-adjusted P<0.005 and log2 fold change >2, 482 differentially expressed murine genes were identified. After identifying differentially expressed genes (DEGs), log-transformed and mean-centered counts per million (CPM) values for 47,997 canine and murine genes were generated. The Pearson distance similarity metric and average linkage clustering method was used for hierarchical clustering of log2 CPM values for the 482 differentially expressed murine genes. See Table S1 for detailed gene lists. (A) Heatmap shows clustered gene-level counts with lower than mean (blue), higher than the mean (red) and mean (gray) levels of expression. Each row represents a single gene. The dendrogram of the horizontal axis of the heatmap shows two sample clusters; OS-1 (blue) and OS-2 (gray) xenografts are in separate sample groups. The rows of the heatmap (vertical axis) cluster into two highly correlated groups. Rows colored in red in the vertical dendrogram are murine genes that are upregulated in OS-2 xenografts, whereas rows colored in blue are downregulated relative to OS-1 xenografts. (B,C) Enriched pathway and functional classification analyses of DEGs were performed using IPA according to row cluster designation. (B) Upregulated genes, red; (C) downregulated genes, blue.

 

We looked at upstream regulators of these 482 differentially expressed murine genes by Ingenuity Pathway Analysis (IPA). The most significant, predicted activated upstream regulators in OS-2 (worse prognosis), relative to OS-1 tumor xenografts, were CEBPB and NFKB1 (P-value 5.54E–10 and 3.94E–09, respectively), whereas the most significant predicted inhibited upstream regulator was MEF2C (P-value 2.54E–23) (Table S3). The retinoblastoma tumor suppressor gene (RB1) was also among the predicted significant upstream regulators (P-value 1.25E–04) showing inactivation in OS-2 xenograft tumors, as we would have predicted based on our previous work (Scott et al., 2015) (Table S3).

To better understand the unique differences between OS-1 and OS-2, we considered the upregulated and downregulated murine genes in OS-2 as separate lists and used IPA to identify enriched biological functions and transcription factors that regulate these genes. The 482 differentially expressed murine genes included 240 that were upregulated (Fig. 5B) and 242 that were downregulated (Fig. 5C) in OS-2 tumor xenografts relative to OS-1 tumor xenografts. The most upregulated murine gene in the OS-2 xenografts was Mcpt1 (+11.25 fold), whereas the most downregulated murine gene was Nkx2-1 (–10.97 fold) (Table S2).

Based on biological function and processes, the most upregulated murine genes in OS-2 tumors were proteases, metallopeptidases, cytokines and chemokines involved in cell movement, leukocyte migration, inflammation and angiogenesis (Fig. 5C, Table S2). By contrast, the most downregulated genes in OS-2 tumor xenografts were transcriptional regulators of cellular differentiation and cell cycle involved in the formation and morphology of muscle (Fig. 5C, Table S2).

Upstream regulators predicted to modulate expression and activity of the 240 upregulated expressed murine genes in the OS-2 tumor xenografts included the T-helper cell type-17 (Th17)-activating cytokines TGF-β (P-value 1.26E–27), IL-1β (P-value 9.07E–25) and IL-6 (P-value 9.03E–22) (Table S4).

The top upstream regulators predicted to modulate expression and activity of the 244 downregulated murine genes in the OS-2 xenografts were MEF2C and MYOD1 (P-value 1.15E–24 and 2.16E–15, respectively) (Table S5). MEF2C and MYOD1, both predicted as being inhibited in OS-2 xenografts and activated in OS-1 tumors, are important in promoting transcription of muscle-specific target genes and play a role in muscle differentiation.

 

DISCUSSION

In this study, we established a novel approach using mouse xenografts to study the heterogeneity and biological behavior of OS in vivo. Specifically, this approach creates opportunities to examine tumor-intrinsic properties, as well as organotypic tumor–stromal interactions that influence tumor progression.

We injected cells at the orthotopic site to simulate the biology of the spontaneous disease. A comprehensive review by Talmadge et al., (2007) described the advantages of orthotopic xenografts over subcutaneous xenografts for solid tumors. Importantly, the anatomical site of implantation needs to be considered carefully because the biological behavior of tumors is dependent on the intrinsic properties of both tumor cells and host factors (which differ between tissues and organs). The microenvironment in subcutaneous xenografts consists of desmoplastic mouse stromal cells that do not resemble the organization seen in autochthonous tumors (Delitto et al., 2015). These properties also apply to OS: Rosol and colleagues showed that orthotopic canine OS xenografts in nude mice produced osteoid matrix and metastasized spontaneously, whereas subcutaneous xenografts did not (Wolfe et al., 2011).

The applicability of cell lines to understand tumor heterogeneity has similarly been challenged (Choi et al., 2014; McIntyre et al., 2015). However, our data show that heterogeneity of biological behavior (including metastatic propensity) can be recapitulated to a limited extent in tumors from cell lines, but more readily by utilizing multiple cell lines that cover the spectrum of tumor behavior. Another impediment that has been articulated is genetic drift, where cell lines that adapt to grow in culture no longer resemble the genetic makeup of the parental tumors (Daniel et al., 2009). Yet, our data show that the major genetic drivers that distinguish the two canine OS cell lines in vitro were retained in the orthotopic xenografts. In addition to stability of the transcriptome, the cell lines show stable morphology from the primary canine tumors to the primary orthotopic tumors (Scott et al., 2015), and to the metastatic tumors. Confirmation of this remarkable genetic and morphologic stability over many passages was essential to validate the utility of our model to understand OS tumor heterogeneity.

As predicted from the original behavior of the spontaneous tumors in the dogs and from their gene and microRNA expression signatures (Sarver et al., 2013; Scott et al., 2011; Thayanithy et al., 2012), the logarithmic expansion phase of OS-2 primary xenografts was faster than that of OS-1 primary xenografts. However, both cell lines seemed to reach the tumor endpoints at approximately the same time. We believe that two factors might account for this. First, the tumors are growing within a cavity surrounded by bone and, despite the fact that OS-2 xenografts showed greater epiphyseal destruction and invasion, the bone constrains the maximum size achievable by the primary tumors within the experimental time frame. Second, mice with OS-2 xenografts did not show greater morbidity than mice with OS-1 xenografts, determined by the absence of lameness, ambulatory deficits and other behaviors associated with chronic pain. This could be due to adaptive behavior of prey species to hide pain (Arras et al., 2007); however, previous work has shown unequivocally that painful intramedullary bone tumors produce behavioral changes in mice (Pacharinsak and Beitz, 2008). We should note that these cell lines accurately represent the biological behavior of the tumors from which they were originally derived, and more broadly the classification of more aggressive and less aggressive tumors (Moriarity et al., 2015; Sarver et al., 2013; Scott et al., 2011, 2015; Thayanithy et al., 2012). Furthermore, such properties have been verified independently by other groups using one of these cell lines (Wolfe et al., 2011), and they generally extend to human and murine osteosarcoma (Moriarity et al., 2015) (M.C.S., unpublished). Still, data from two cell lines should be interpreted with caution because osteosarcomas are extremely heterogeneous, both genetically and biologically (Varshney et al., 2016). It will thus be important to document the fidelity with which this model is able to recapitulate such heterogeneity.

Beyond growth at the primary site, biological behavior can be quantified by metastatic propensity and successful spread to distant sites. Again, the predictions from the original spontaneous tumors were confirmed experimentally in our models. OS-2 cells were a representative example from a group of highly aggressive tumors (worse prognosis) that showed high expression of cell-cycle- and DNA-damage-repair-associated genes, with concomitant reduced expression of a complement of genes that defined ‘microenvironment interactions’ (Scott et al., 2011). This reduced expression of molecules that mediate local cell communication could explain, at least in part, the observation that cells injected intratibially achieved rapid systemic distribution, spreading to the lungs within 6 h; i.e. there was nothing to hold the cells in place, and they had no preference to remain in the local bone environment.

We cannot completely exclude the possibility that metastasis is driven by selection of cells that acquire (or previously harbored) mutations or epigenetic events that favor dissemination to and survival in the lung, and, in fact, both mechanisms (niche conditioning and selection) could be operative in this model. If this were the case, however, and given the reproducibility of results over multiple experiments and the low probability that cells would acquire the same stochastic mutations repeatedly, one would have to argue that such mutations or epigenetic changes pre-exist in a subset of cells within these cell lines.

A previous study (Garimella et al., 2013) used in vivo imaging to show tumor cells in the lungs within 2 weeks after orthotopic implantation of OS cells; however, we are not aware of any previous studies showing dissemination of OS cells into the lungs on the day of implantation. In our experiments, the luciferase signals disappeared from the lungs in all of the mice receiving OS-2 xenografts within 24 h, and they were only visible again in one of the 32 mice within 2 weeks and eventually in six of 32 mice that received OS-2 cells by the end of the experiment. This suggests that the lung niche required prior conditioning by OS-2 tumors in order to become receptive for metastatic colonization. Furthermore, our results suggest that, even though both OS-1 and OS-2 cell lines can establish a metastatic niche, they do so with different kinetics, creating a suitable model to study intrinsic differences in metastatic propensity, as well as host-related factors that contribute to the metastatic niche in OS.

Based on these observations, we could propose two distinct mechanisms for the different metastatic potential of OS-1 and OS-2 xenografts. One noted above is that OS-2 cells might have greater metastatic potential due to their interaction with the local microenvironment in the bone, which leads to reduced retention, and potentially to an increased capability to condition the distant site. The alternative possibility is that, as shown in Fig. 2, OS-2 cells seed the lungs shortly after inoculation and, even though many of these cells might leave the lungs or die, accounting for the loss of luciferase signal by 24 h, some cells remain and eventually form the pulmonary lesions (i.e. equivalent to seeding or colonization by intravenous inoculation). We favor the first possibility because preliminary experiments suggest that OS-1 and OS-2 cells have low efficiency of pulmonary colonization upon intravenous injection. Nevertheless, additional experiments will be necessary to formally exclude the second possibility.

Our results differ from those of Rosol's group (Wolfe et al., 2011), which showed development of multiple lung metastases in all of the mice that received orthotopic injections with canine OSCA-40 cells (OS-2). This is almost certainly due to the fact that we terminated our experiments after 8 weeks, whereas Rosol et al. continued their experiments for up to 12 weeks. We thus surmise that OS-2 and other aggressive OS cell lines can be used to investigate therapeutic interventions to delay or prevent OS metastasis in the minimal residual disease setting, whether attained through amputation or through administration of cytoreductive chemotherapy. Future experiments could further investigate the intrinsic biology of the tumors and mechanisms of drug resistance, as well as preclinical interventions to delay or prevent metastatic dissemination, especially by modeling the current standards of care, which combine surgery with neoadjuvant and adjuvant chemotherapy, and particularly expanding the model to leverage multiple available human and canine OS cell lines (Lauvrak et al., 2013; Mohseny et al., 2011; Scott et al., 2011).

Finally, genomic stability might be a peculiar feature of OS xenografts. Consistent with our previous study (Scott et al., 2015), our present results indicate that functional RB can be stably maintained in OS cells, and that its loss in the tumor cell compartment is associated with a more aggressive phenotype of rapid growth and increased metastatic propensity. In addition, other studies have shown that gene expression patterns and copy number alterations were preserved in patient-derived OS cell lines and xenograft tumors (Kuijjer et al., 2011; Mayordomo et al., 2010). Yet, we are not aware of any previous studies describing the relationship between intrinsic gene signatures of OS tumor cells with distinctly different biological behaviors and host stromal cells.

Highly expressed mouse genes present in the OS-2 xenografts were associated with B-cell signaling, inflammation and immune response, whereas mouse genes in the OS-1 cells xenografts were associated with patterning, and especially with muscle formation. Increased expression of myogenic regulators in mouse stromal cells in OS-1 xenografts raises interesting questions regarding possible effects of OS-1 tumor cells on marrow-derived mesenchymal stromal cells. Interestingly, myogenic regulators have been implicated in human oncogenesis. For instance, expression of MYOD1 can predict patient survival in lung cancer patients (Jiang et al., 2015) and high expression of MEF2C is associated with poor outcome in acute myeloid leukemia (AML) patients (Laszlo et al., 2015). Moreover, myogenic transcription factors have been shown to regulate metastasis in soft-tissue sarcomas, suggesting that further investigation of these factors be done in other sarcomas, including OS (Dodd et al., 2016). Importantly, MEF2C was recently identified as a candidate tumor suppressor gene in OS in a forward genetic screen (Moriarity et al., 2015).

Intriguingly, the most downregulated murine gene in the OS-2 xenografts was the transcription factor Nkx2-1, which is known to regulate lung epithelial cell morphogenesis and differentiation. Downregulation of NKX2-1 has been shown to precede dissemination of lung adenocarcinoma cells (Caswell et al., 2014). NKX2-1 amplification has been reported in one human OS patient but there are no reports of downregulation or loss of NKX2-1 in OS patients (Egas-Bejar et al., 2014).

Activated TGF-β, IL-6 and IL-1β were the most significant upstream regulators of highly expressed genes from stromal cells in the OS-2 xenografts. This is especially intriguing because this would normally be associated with a pro-inflammatory Th17 response. Such a response cannot happen in athymic nude mice, which lack T cells; in fact, it is important to recognize that a limitation of this model is the fact that the full complement of the T-cell immune response cannot be studied in the immunocompromised mouse strains that provide receptive hosts for tumor xenotransplantation. This then creates a gap that will have to be addressed using syngeneic or autochthonous, immunocompetent animal models.

In conclusion, we have developed xenograft models that recapitulated the heterogeneous biological behavior of OS. These models will be useful to understand the mechanisms that drive progression and metastasis of OS because they are expandable into additional cell lines to represent a wider spectrum of disease.

 

MATERIALS AND METHODS

Cells and culture conditions

Two canine OS cell lines representing previously described ‘less aggressive’ and ‘highly aggressive’ molecular phenotypes (OS-1 and OS-2, respectively), were used in this study (Scott et al., 2011, 2015). OS-1 and OS-2 are derivatives of the OSCA-32 and OSCA-40 cell lines (Scott et al., 2011, 2015). Specifically, OS-1 represents a subline that successfully established tumors after orthotopic implantation, because the parental OSCA-32 did not establish heterotopic or orthotopic tumors on every occasion. OS-2 represents the parental OSCA-40, which reliably formed tumors after orthotopic implantation in every experiment done (Scott et al., 2015; Wolfe et al., 2011).

Cell lines were validated using short tandem repeat (STR) profiles by DNA Diagnostics Center (DDC Medical) (Fairfield, OH). OS-1 and OS-2 cells were modified to stably express green fluorescent protein (GFP) and firefly luciferase as described (Scott et al., 2015) and used for orthotopic injections in mice. After transfection and selection, we confirmed that the GFP/luciferase construct was stably integrated in each cell line by fluorescence in situ hybridization, and we corroborated that the two cell lines had approximately equivalent luciferase activity on a per cell basis using conventional luciferase assays (Scott et al., 2015). All cell lines were grown in DMEM (Gibco, Grand Island, NY) containing 5% glucose and L-glutamine, supplemented with 10% fetal bovine serum (Atlas Biologicals, Fort Collins, CO), 10 mM 4-(2-hydroxyethyl)-1-piperazine ethanesulphonic acid buffer (HEPES) and 0.1% Primocin (InvivoGen, San Diego, CA), and cultured at 37°C in a humidified atmosphere of 5% CO2. Canine OS cell lines are available for distribution through Kerafast, Inc. (Boston, MA). Each cell line was passaged more than 15 times before the experiments when they were inoculated into mice.

 

Mice

Six-week-old female, athymic nude mice (strain NCrnu/nu) were obtained from Charles River Laboratories (Wilmington, MA). The University of Minnesota Institutional Animal Care and Use Committee approved protocols for mouse experiments of this study (protocol no.: 1307-30806A).

 

Tumor xenografts

Eight animals per group provide >95% power to identify a 15% change in the median time to tumor when the σ for both populations is <2.0 and the acceptable α error is 5% (P<0.05). Experimental replicates increased statistical robustness, accounting for the expected heterogeneity.

Four replicate experiments were done to assess orthotopic growth and metastatic dissemination of OS-1 and OS-2 cells. For the first pilot experiment, groups of three mice were used to validate the approach. All of the mice receiving OS-1 xenografts showed successful implantation, but only two of the three mice receiving OS-2 xenografts showed successful implantation. For the second experiment, groups of 16 mice were used to establish significance. In this experiment, all of the mice receiving OS-2 xenografts showed successful implantation, but eight mice injected with OS-1 xenografts had significant adverse effects during anesthesia and were not recovered (i.e. they were humanely euthanatized). For the third experiment, we inoculated nine mice with OS-2 cells to verify the unexpected effects of rapid dissemination to the lung. No mice received OS-1 for this experiment. Finally, for the fourth experiment, we inoculated five mice with each cell line (OS-1 or OS-2) to achieve a biological replicate of experiment two, maintaining the sample size at a number to maximize a positive outcome. Appropriate censoring was used to include all animals in the analyses, only excluding any which succumbed acutely or subacutely during the intratibial injection procedure. Thus, 16 mice inoculated with OS-1 were included in the analyses of tumor growth, and 32 mice inoculated with OS-2 were included in the analyses of tumor growth.

We previously determined that four samples per group approximate the point of minimal returns using large genomic datasets for gene expression profiling (Tamburini et al., 2009), and these estimates hold true from microarrays to RNAseq where the fidelity of replication within samples is high, despite orders of magnitude more data (see analysis of RNA sequencing below).

Animals were assigned to separate cages (four animals each) in random order for each experiment. All of the animals in each cage received the same treatment. OS-1 and OS-2 cells expressing GFP and firefly luciferase were injected intratibially. Mice were anesthetized with xylazine [10 mg/kg body weight, intraperitoneally (IP)] and ketamine (100 mg/kg, IP), and 1×105 cells suspended in 10 µl of sterile PBS were injected into the left tibia using a tuberculin syringe with 29-gauge needle. Buprenorphine (0.075 mg/kg, IP  every 8 h; Buprenex®, Reckitt Benckiser Healthcare, Richmond, VA) was used for pain control over the first 24 h after injection of tumor cells, and prophylactic ibuprofen administrated in the water was used over the next 3 days.

Tumor growth was monitored by measuring width (W) and length (L) of the proximal tibia and the stifle joint weekly using calipers, as well as by in vivo imaging as described (Kim et al., 2014). Bioluminescence imaging (Xenogen IVIS spectrum, Caliper Life Sciences, Hopkinton, MA) was done after injection of D-luciferin (Gold Biotechnology, St Louis, MO) following isoflurane inhalant anesthesia and analyzed with Living Image Software (Caliper Life Sciences). Bone tissue volume (V) was calculated from both tibiae using the equation V=L×W2×0.52 (Banerjee et al., 2013) and tumor volume was estimated by subtracting the normal bone tissue volume of the contralateral unaffected (right) tibia from the volume of the affected (left) tibia.

Mice were observed for up to 8 weeks or until tumor endpoint criteria were reached (ill thrift, tumor reaching 1 cm in the largest diameter, visible lameness, pain or severe weight loss), at which time they were humanely euthanized with pentobarbital sodium and sodium phenytoin solution (Beuthanasia-D Special®, Schering-Plough Animal Health, Union, NJ). Primary bone tumors and lung tissues were dissected and a portion of each was stored at −80°C for RNA extraction. The remaining tissues were fixed in 10% neutral-buffered formalin, and processed for routine histological examination.

Luciferase activity and tumor sizes were compared using multiple t-test and Holm–Sidak method with Prism 6 software (GraphPad). P<0.05 was used as the level of significance.

 

RNA extraction, library preparation and RNA sequencing

Total RNA was extracted from primary intratibial tumors and from cell lines using the miRNeasy Mini Kit (QIAGEN, Valencia, CA). RNA integrity was examined using Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA) and RNA integrity number (RIN) values of all samples were >8.0. Sequencing libraries were prepared with the TruSeq Library Preparation Kit (Illumina, San Diego, CA). RNA sequencing (100-bp paired-end) with HiSeq 2500 (Illumina) was done at the University of Minnesota Genomics Center (UMGC). A minimum of ten-million read-pairs was generated for each sample.

 

Analysis of RNA sequencing data

Initial quality control analysis of RNA sequencing (FASTQ) data for each sample was performed using the FastQC software (version 0.11.2; http://www.bioinformatics.babraham.ac.uk/projects/fastqc). FASTQ data were trimmed with Trimmomatic (Bolger et al., 2014). HISAT2 (Kim et al., 2015) was used to map paired-end reads from eight xenograft tumors (four tumors of OS-1 and four tumors of OS-2) and four parental cell-line samples (two each for OS-1 and OS-2 cell lines). For accurate alignment of sequencing reads to canine and murine genes within xenograft tumors, a HISAT2 index for mapping was built from a multi-sequence fasta file containing both the canine (canFam3) and murine (mm10) genomes. Insertion-size metrics were calculated for each sample using Picard software (version 1.126) (http://picard.sourceforge.net). Samtools (version 1.0_BCFTools_HTSLib) was used to sort and index the bam files (Li et al., 2009). Transcript abundance estimates were generated using the Rsubread featureCounts program for differential gene expression analysis (Liao et al., 2014).

Gene counts for each xenograft sample were imported into RStudio (v. 3.2.3) (http://www.rstudio.com) for differential gene expression (DGE) analysis with EdgeR (Robinson et al., 2010; Zhou et al., 2014). Lowly expressed genes were removed by filtering. A gene was considered expressed if it had log2-transformed read counts per million (CPM) >1 in at least two of the eight xenograft tumors. Biological variation within xenograft sample groups was estimated by common dispersion and biological coefficient of variation (BCV) calculations (Robinson et al., 2010). Pairwise empirical analysis of differential gene expression was performed on sample groups (OS-1 and OS-2) using Fisher's exact test for two-group comparisons with TMM normalization (Robinson and Oshlack, 2010). Tagwise dispersion (individual dispersion for each gene) was used to adjust for abundance differences across biological replicates (n=4) within each xenograft group (OS-1 and OS-2). Gene counts as CPM were imported into Partek Genomic Suite for clustering analysis and visualization. The Pearson similarity metric and average linkage clustering method were used for hierarchical clustering of mean-centered CPM values. Enriched pathway and functional classification analyses of DGEs were performed using QIAGEN's Ingenuity® Pathway Analysis (IPA®, QIAGEN Redwood City, www.qiagen.com/ingenuity). The reference set for all IPA analyses was the Ingenuity Knowledge Base (genes only) and human Entrez gene names were used as the output format. To understand the high-level functions and utilities that each gene identified as differentially expressed between OS-1 and OS-2 was associated with, we utilized MetaCore software (Thompson Reuters) to identify statistically over-represented cellular processes in the dataset.

 

Acknowledgements

The University of Minnesota's Research Animal Resources provided assistance with mouse experiments. We also recognize Lihua Li, Mitzi Lewellen, Ashley Graef, Katie Anderson, and Drs Jong-Hyuk Kim and Keumsoon Im for assistance with in vivo experiments, and Dr Ramesh Kovi for assistance with pathological analyses. We also recognize Drs Aaron Sarver, David Largaespada, Tim O'Brien, Daisuke Ito and Erin Dickerson for helpful discussions.

 

Footnotes

  • Competing interests

    The authors declare that patent “A method for discovery of cell-free nucleic acids as markers of disease” related to this work and listing Milcah C. Scott, John R. Garbe and Jaime F. Modiano as inventors has been filed by the Office of Technology Commercialization of the University of Minnesota.

  • Author contributions

    H.T. conducted the xenograft experiments. M.C.S. applied bioinformatic techniques to synthesize and analyze RNA sequencing data. M.C.S. and H.T. generated, analyzed, interpreted and validated the data, and both contributed equally to writing the initial drafts, reviewing, editing and writing the final draft of the manuscript. J.R.G. provided programming and implementation of computer code and supporting algorithms and testing of code components, and critically reviewed and revised the initial drafts of the manuscript. I.C. and M.G.O. were responsible for gross necropsies, for pathological evaluation of the mice and the xenograft tumors, and for preparation of the photomicrographs used in the figures. C.A. performed the MetaCore analysis. B.A.B. contributed to experimental design, critically reviewed and revised the initial drafts of the manuscript and acquired funding. S.S. contributed to experimental design, critically reviewed and revised the initial drafts of the manuscript, and acquired funding. J.F.M. conceptualized and designed the study, acquired funding, provided study supervision, and assisted with review and editing of the initial drafts of the manuscript. All of the authors reviewed the results and approved the final version of the manuscript.

  • Funding

    This study was supported by grants from Morris Animal Foundation (D13CA-032 to J.F.M. and D15CA-047 to J.F.M., S.S. and B.A.B.), the American Cancer Society (RSG-13-381-01 to S.S.), Karen Wyckoff Rein in Sarcoma Foundation (2011-1 to J.F.M.), and the Comparative Medicine Signature Program of the College of Veterinary Medicine, University of Minnesota (to J.F.M., S.S. and B.A.B.). The National Institutes of Health (NIH) Comprehensive Cancer Center Support Grant to the Masonic Cancer Center, University of Minnesota (P30 CA077598) provided support for bioinformatics, genomics, bioimaging and comparative pathology services. J.F.M. is supported by the Alvin and June Perlman Chair in Animal Oncology, University of Minnesota College of Veterinary Medicine. The authors also gratefully acknowledge funds from donors to the Animal Cancer Care and Research Program of the University of Minnesota that helped support the project. The automated RNA-sequence analysis pipeline used for this work was made possible by support from the University of Minnesota Informatics Institute, in collaboration with the University of Minnesota Genomics Center and the RIS group at the University of Minnesota Supercomputing Institute.

  • Data availability

    Data associated with this manuscript is available in the GEO database under accession number GSE89835 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE89835).

  • Supplementary information

    Supplementary information available online at http://dmm.biologists.org/lookup/doi/10.1242/dmm.026849.supplemental

  • Received July 8, 2016.
  • Accepted September 23, 2016.
http://creativecommons.org/licenses/by/3.0

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

 

References

Data supplements

DMM026849 Supplementary information

Gene logFC PValue FDR Entrez Gene Name Location Type(s)
Nkx2-1 -10,9716375586 3,67E-43 2,51E-39 NK2 homeobox 1 Nucleus transcription regulator
Zic1 -10,3772096495 2,80E-28 5,47E-25 Zic family member 1 Nucleus transcription regulator
Kcnq2 -7,9364263299 1,90E-18 1,37E-15 potassium channel, voltage gated KQT-like subfamily Q, member 2 Plasma Membrane ion channel
Smpd3 -5,9373983942 4,58E-11 4,93E-09 sphingomyelin phosphodiesterase 3, neutral membrane (neutral sphingomyelinase II) Cytoplasm enzyme
Phex -5,4791366738 3,98E-13 7,78E-11 phosphate regulating endopeptidase homolog, X-linked Cytoplasm peptidase
Fam43b -5,1655158795 7,72E-13 1,37E-10 family with sequence similarity 43, member B Other other
Myoz2 -5,031353137 1,21E-14 3,75E-12 myozenin 2 Other other
Myh2 -5,0162782183 1,42E-10 1,37E-08 myosin, heavy chain 2, skeletal muscle, adult Cytoplasm enzyme
Hoxd13 -5,0056512025 6,42E-16 2,83E-13 homeobox D13 Nucleus transcription regulator
Slc13a5 -4,983558827 3,64E-11 4,15E-09 solute carrier family 13 (sodium-dependent citrate transporter), member 5 Plasma Membrane transporter
Panx3 -4,8243778495 2,74E-06 7,53E-05 pannexin 3 Plasma Membrane other
Tmem145 -4,6027921516 2,02E-10 1,92E-08 transmembrane protein 145 Other other
Lect1 -4,5675141671 5,15E-09 3,25E-07 leukocyte cell derived chemotaxin 1 Extracellular Space other
Asic3 -4,4735941511 2,73E-14 7,19E-12 acid sensing (proton gated) ion channel 3 Plasma Membrane ion channel
Ankrd2 -4,4450481038 2,61E-19 2,23E-16 ankyrin repeat domain 2 (stretch responsive muscle) Nucleus transcription regulator
Fam180a -4,4449197857 1,04E-07 4,35E-06 family with sequence similarity 180, member A Other other
Col9a1 -4,3972952794 2,52E-11 3,08E-09 collagen, type IX, alpha 1 Extracellular Space other
Fabp3 -4,3674922168 2,88E-18 1,97E-15 fatty acid binding protein 3, muscle and heart Cytoplasm transporter
Lyz1 -4,308114309 0,0001025991 0,0016378742 lysozyme Extracellular Space enzyme
Adamts18 -4,303169665 3,57E-09 2,42E-07 ADAM metallopeptidase with thrombospondin type 1 motif, 18 Extracellular Space peptidase
Sp7 -4,2248164906 1,29E-08 7,16E-07 Sp7 transcription factor Nucleus transcription regulator
Omd -4,1860981293 2,89E-17 1,58E-14 osteomodulin Extracellular Space other
1700101I11Rik -4,1551683867 2,75E-07 9,92E-06 RIKEN cDNA 1700101I11 gene Other other
Dlx6 -4,109636472 3,22E-11 3,76E-09 distal-less homeobox 6 Nucleus transcription regulator
Actn2 -4,0698270275 5,15E-17 2,71E-14 actinin, alpha 2 Nucleus transcription regulator
Tceal7 -4,0382675922 4,23E-06 0,0001085007 transcription elongation factor A (SII)-like 7 Nucleus transcription regulator
Xirp2 -4,0148103261 6,23E-10 5,33E-08 xin actin binding repeat containing 2 Other other
Paqr6 -3,961900754 2,42E-10 2,28E-08 progestin and adipoQ receptor family member VI Plasma Membrane other
Csrp3 -3,9364612384 8,12E-11 8,23E-09 cysteine and glycine-rich protein 3 (cardiac LIM protein) Nucleus other
Fcrls -3,8106497911 2,99E-37 1,36E-33 Fc receptor-like S, scavenger receptor Plasma Membrane other
Ckmt2 -3,8064250766 2,10E-09 1,55E-07 creatine kinase, mitochondrial 2 (sarcomeric) Cytoplasm kinase
Myl6b -3,7875440342 9,99E-09 5,82E-07 myosin, light chain 6B, alkali, smooth muscle and non-muscle Cytoplasm other
Gli1 -3,7400401264 9,38E-17 4,58E-14 GLI family zinc finger 1 Nucleus transcription regulator
Zdbf2 -3,7348427202 1,50E-07 6,03E-06 zinc finger, DBF-type containing 2 Other other
Opn1mw -3,7263451513 8,04E-07 2,55E-05 opsin 1 (cone pigments), long-wave-sensitive Plasma Membrane G-protein coupled receptor
Gsg1l -3,7215249103 9,85E-10 8,07E-08 GSG1-like Plasma Membrane other
Abra -3,6979867699 9,51E-13 1,65E-10 actin binding Rho activating protein Cytoplasm other
Myom3 -3,6819161582 4,38E-08 2,07E-06 myomesin 3 Other other
Serpinb1c -3,6579176748 4,90E-11 5,20E-09 serine (or cysteine) peptidase inhibitor, clade B, member 1c Extracellular Space other
Foxg1 -3,648321574 1,21E-19 1,14E-16 forkhead box G1 Nucleus transcription regulator
Ifitm5 -3,6176801516 2,81E-09 2,03E-07 interferon induced transmembrane protein 5 Plasma Membrane other
9130024F11Rik -3,4900157355 2,34E-12 3,72E-10 RIKEN cDNA 9130024F11 gene Other other
Csrnp3 -3,4502519116 1,48E-06 4,37E-05 cysteine-serine-rich nuclear protein 3 Nucleus transcription regulator
Slc47a1 -3,4439945824 2,09E-06 5,96E-05 solute carrier family 47 (multidrug and toxin extrusion), member 1 Plasma Membrane transporter
2410137F16Rik -3,4411080614 6,33E-07 2,07E-05 Err:512 Err:512 Err:512
A930003A15Rik -3,4384931098 7,11E-08 3,21E-06 RIKEN cDNA A930003A15 gene Other other
Col11a2 -3,4328957502 9,67E-09 5,68E-07 collagen, type XI, alpha 2 Extracellular Space other
Cst6 -3,4209081684 2,33E-07 8,66E-06 cystatin E/M Extracellular Space other
Actc1 -3,400606916 1,73E-07 6,78E-06 actin, alpha, cardiac muscle 1 Cytoplasm enzyme
Atp1b4 -3,3529912076 2,08E-05 0,0004257257 ATPase, Na+/K+ transporting, beta 4 polypeptide Plasma Membrane transporter
Alpk2 -3,3411041845 9,43E-05 0,001527325 alpha-kinase 2 Nucleus kinase
Myh1 -3,324236394 1,55E-09 1,18E-07 myosin, heavy chain 1, skeletal muscle, adult Plasma Membrane enzyme
Hspb3 -3,3174390477 1,86E-08 9,63E-07 heat shock 27kDa protein 3 Cytoplasm other
Itgb1bp2 -3,3116206495 2,66E-09 1,93E-07 integrin beta 1 binding protein (melusin) 2 Other other
Casq2 -3,2920861485 1,53E-08 8,17E-07 calsequestrin 2 (cardiac muscle) Cytoplasm other
Dmp1 -3,2579947561 1,22E-09 9,51E-08 dentin matrix acidic phosphoprotein 1 Extracellular Space other
Plb1 -3,2423202635 4,00E-07 1,39E-05 phospholipase B1 Cytoplasm enzyme
Cacna2d2 -3,2410010861 1,16E-09 9,20E-08 calcium channel, voltage-dependent, alpha 2/delta subunit 2 Plasma Membrane ion channel
AU022793 -3,1985085807 2,72E-06 7,49E-05 expressed sequence AU022793 Other other
Mylpf -3,1964183009 5,32E-07 1,77E-05 myosin light chain, phosphorylatable, fast skeletal muscle Cytoplasm other
Xirp1 -3,193088545 6,65E-12 9,38E-10 xin actin binding repeat containing 1 Plasma Membrane other
Dok7 -3,1562169749 4,84E-09 3,08E-07 docking protein 7 Extracellular Space other
Hsd11b2 -3,1550207041 5,80E-10 5,06E-08 hydroxysteroid (11-beta) dehydrogenase 2 Cytoplasm enzyme
Fat3 -3,116929351 9,82E-07 3,04E-05 FAT atypical cadherin 3 Plasma Membrane other
Bex1 -3,0988525942 5,93E-07 1,96E-05 brain expressed gene 1 Other other
Siglec1 -3,0563222661 5,56E-36 1,52E-32 sialic acid binding Ig-like lectin 1, sialoadhesin Plasma Membrane other
Klhl30 -3,0426707735 3,66E-12 5,38E-10 kelch-like family member 30 Other other
Srpk3 -3,0365598419 8,83E-10 7,33E-08 SRSF protein kinase 3 Cytoplasm kinase
Nmrk2 -3,0365047079 9,23E-08 3,94E-06 nicotinamide riboside kinase 2 Plasma Membrane kinase
Hspb7 -3,0352239347 4,33E-12 6,23E-10 heat shock 27kDa protein family, member 7 (cardiovascular) Cytoplasm other
Hspb6 -3,0100384366 3,90E-11 4,38E-09 heat shock protein, alpha-crystallin-related, B6 Cytoplasm other
Myh3 -3,0054128778 3,96E-09 2,63E-07 myosin, heavy chain 3, skeletal muscle, embryonic Cytoplasm enzyme
Zim1 -2,9990022809 5,28E-07 1,77E-05 zinc finger, imprinted 1 Nucleus other
Fhl1 -2,9717168325 7,50E-12 1,05E-09 four and a half LIM domains 1 Cytoplasm other
Cryab -2,942079665 4,57E-11 4,93E-09 crystallin, alpha B Nucleus other
Col26a1 -2,9225992658 0,0001400874 0,0021186368 collagen, type XXVI, alpha 1 Extracellular Space other
Tpm2 -2,9010265451 3,06E-11 3,67E-09 tropomyosin 2, beta Cytoplasm other
Smpx -2,8905869001 3,29E-15 1,25E-12 small muscle protein, X-linked Cytoplasm other
Mybpc1 -2,8892754024 7,51E-07 2,41E-05 myosin binding protein C, slow type Cytoplasm other
Car3 -2,8836906344 3,67E-09 2,48E-07 carbonic anhydrase III Cytoplasm enzyme
Myl3 -2,879903468 1,51E-05 0,0003271839 myosin, light chain 3, alkali; ventricular, skeletal, slow Cytoplasm other
Acta1 -2,8584976657 2,23E-07 8,33E-06 actin, alpha 1, skeletal muscle Cytoplasm other
Adprhl1 -2,8395941876 3,39E-09 2,36E-07 ADP-ribosylhydrolase like 1 Other enzyme
Robo2 -2,838080162 1,25E-06 3,77E-05 roundabout guidance receptor 2 Plasma Membrane transmembrane receptor
Col9a2 -2,8365412794 9,14E-11 9,13E-09 collagen, type IX, alpha 2 Extracellular Space other
Frzb -2,8224077488 2,37E-07 8,77E-06 frizzled-related protein Extracellular Space other
Matn3 -2,8188908343 2,10E-05 0,0004291306 matrilin 3 Extracellular Space other
Vgll2 -2,7799837339 3,56E-09 2,42E-07 vestigial-like family member 2 Nucleus transcription regulator
Alpl -2,7744748405 3,78E-13 7,49E-11 alkaline phosphatase, liver/bone/kidney Plasma Membrane phosphatase
Cdo1 -2,7706361608 1,67E-08 8,76E-07 cysteine dioxygenase type 1 Cytoplasm enzyme
Mfsd7c -2,7620275638 6,69E-07 2,18E-05 feline leukemia virus subgroup C cellular receptor family, member 2 Plasma Membrane transporter
Crhr2 -2,7363362326 1,20E-11 1,54E-09 corticotropin releasing hormone receptor 2 Plasma Membrane G-protein coupled receptor
Myadml2 -2,7242624015 4,46E-06 0,0001135398 myeloid-associated differentiation marker-like 2 Cytoplasm other
Pax2 -2,7196002549 9,94E-06 0,0002282677 paired box 2 Nucleus transcription regulator
Zic2 -2,7156986445 1,17E-05 0,0002632318 Zic family member 2 Nucleus transcription regulator
S100b -2,7099906055 1,76E-14 5,13E-12 S100 calcium binding protein B Cytoplasm other
Synpo2l -2,7014418753 3,57E-09 2,42E-07 synaptopodin 2-like Cytoplasm other
Cox6a2 -2,6939364839 4,57E-07 1,54E-05 cytochrome c oxidase subunit VIa polypeptide 2 Cytoplasm enzyme
Gm6524 -2,6924107256 5,56E-05 0,0009709226 katanin p60 (ATPase-containing) subunit A1 pseudogene Other other
Ccrl1 -2,6765792744 2,47E-06 6,85E-05 Err:512 Err:512 Err:512
Col22a1 -2,6747279869 3,90E-10 3,47E-08 collagen, type XXII, alpha 1 Extracellular Space other
Cav3 -2,673548764 1,73E-07 6,78E-06 caveolin 3 Plasma Membrane enzyme
Slc38a3 -2,6541805856 6,41E-05 0,0010921343 solute carrier family 38, member 3 Plasma Membrane transporter
Tmem8c -2,6532666696 8,29E-06 0,0001932668 transmembrane protein 8C Plasma Membrane other
Klhl41 -2,6423475394 2,57E-07 9,39E-06 kelch-like family member 41 Cytoplasm other
Des -2,6197695334 8,19E-12 1,11E-09 desmin Cytoplasm other
Ldb3 -2,6193858755 7,34E-06 0,0001736884 LIM domain binding 3 Cytoplasm transporter
Sbk2 -2,6067752909 8,68E-05 0,0014207532 SH3 domain binding kinase family, member 2 Other other
Popdc2 -2,5892730802 2,98E-06 8,02E-05 popeye domain containing 2 Other other
Snca -2,5888071583 4,55E-06 0,0001152365 synuclein, alpha (non A4 component of amyloid precursor) Cytoplasm other
Ogn -2,586436671 3,77E-10 3,37E-08 osteoglycin Extracellular Space growth factor
Lmod2 -2,5746115711 7,58E-07 2,43E-05 leiomodin 2 (cardiac) Other other
Lepr -2,5681443994 1,03E-08 5,96E-07 leptin receptor Plasma Membrane transmembrane receptor
Lrrc30 -2,5645341012 0,0001039464 0,0016555181 leucine rich repeat containing 30 Other other
Tuba8 -2,5635544579 0,0003196724 0,0042133313 tubulin, alpha 8 Cytoplasm other
Tceal5 -2,5576013151 5,74E-05 0,0009956384 transcription elongation factor A (SII)-like 5 Other other
Myot -2,538095094 1,07E-05 0,0002426224 myotilin Cytoplasm other
Ndnf -2,5371561256 6,13E-10 5,28E-08 neuron-derived neurotrophic factor Extracellular Space other
Ch25h -2,5314620058 3,17E-12 4,77E-10 cholesterol 25-hydroxylase Cytoplasm enzyme
Lrtm1 -2,5314227234 0,0003041813 0,004044222 leucine-rich repeats and transmembrane domains 1 Other other
Yipf7 -2,5205191962 3,17E-06 8,47E-05 Yip1 domain family, member 7 Other other
Rsad2 -2,5201701105 3,91E-08 1,88E-06 radical S-adenosyl methionine domain containing 2 Cytoplasm enzyme
Myl1 -2,5084514213 8,20E-05 0,0013563441 myosin, light chain 1, alkali; skeletal, fast Cytoplasm other
Gm10767 -2,5079943644 1,62E-05 0,0003446054 predicted gene 10767 Other other
Col9a3 -2,5032744148 0,0002073095 0,002966738 collagen, type IX, alpha 3 Extracellular Space other
Pdlim3 -2,5026600666 9,22E-09 5,50E-07 PDZ and LIM domain 3 Plasma Membrane other
Tnnc2 -2,5022510521 2,88E-05 0,0005689415 troponin C type 2 (fast) Cytoplasm other
Myom2 -2,4811407132 6,30E-07 2,07E-05 myomesin 2 Cytoplasm other
Ccl4 -2,4732545233 1,56E-09 1,18E-07 chemokine (C-C motif) ligand 4 Extracellular Space cytokine
Fgfr4 -2,4677672576 5,43E-05 0,000952702 fibroblast growth factor receptor 4 Plasma Membrane kinase
Hand2 -2,4662081456 5,90E-05 0,0010189343 heart and neural crest derivatives expressed 2 Nucleus transcription regulator
Ppargc1a -2,4611712117 2,96E-05 0,0005800445 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha Nucleus transcription regulator
Asb12 -2,4553872143 9,25E-05 0,001503012 ankyrin repeat and SOCS box containing 12 Nucleus transcription regulator
Klhl40 -2,4539973384 1,87E-08 9,68E-07 kelch-like family member 40 Other other
Hspa1l -2,4484962306 7,22E-10 6,10E-08 heat shock 70kDa protein 1-like Cytoplasm other
Srl -2,437036897 2,82E-06 7,69E-05 sarcalumenin Cytoplasm other
Fndc5 -2,4340347436 6,89E-06 0,0001644017 fibronectin type III domain containing 5 Other other
Tnnt3 -2,4339473603 4,26E-05 0,0007861961 troponin T type 3 (skeletal, fast) Cytoplasm other
Greb1 -2,4265030143 1,19E-08 6,75E-07 growth regulation by estrogen in breast cancer 1 Cytoplasm other
I830012O16Rik -2,419437095 0,0001805492 0,0026305582 Err:512 Err:512 Err:512
Sox11 -2,4165935852 3,33E-09 2,34E-07 SRY (sex determining region Y)-box 11 Nucleus transcription regulator
Nrcam -2,4099940272 1,07E-05 0,0002426224 neuronal cell adhesion molecule Plasma Membrane other
Foxl1 -2,4084233801 5,02E-06 0,0001246328 forkhead box L1 Nucleus transcription regulator
Foxc1 -2,3807345005 1,25E-19 1,14E-16 forkhead box C1 Nucleus transcription regulator
Tuba4a -2,3679859152 4,07E-07 1,41E-05 tubulin, alpha 4a Cytoplasm other
Tcap -2,3626526316 7,01E-07 2,27E-05 titin-cap Cytoplasm other
B430306N03Rik -2,3511775625 4,46E-07 1,51E-05 RIKEN cDNA B430306N03 gene Other other
Cap2 -2,3510931688 7,43E-08 3,31E-06 CAP, adenylate cyclase-associated protein, 2 (yeast) Plasma Membrane other
Ucp3 -2,3457431207 3,11E-05 0,0006065499 uncoupling protein 3 (mitochondrial, proton carrier) Cytoplasm transporter
Dmrta2 -2,3386869716 1,16E-06 3,53E-05 DMRT-like family A2 Nucleus transcription regulator
Fgfr3 -2,337098522 2,07E-11 2,60E-09 fibroblast growth factor receptor 3 Plasma Membrane kinase
Mapt -2,3312427067 2,08E-08 1,06E-06 microtubule-associated protein tau Plasma Membrane other
Fgfr2 -2,3214291501 2,02E-05 0,0004189066 fibroblast growth factor receptor 2 Plasma Membrane kinase
Hhatl -2,3115195047 2,06E-05 0,0004242945 hedgehog acyltransferase-like Cytoplasm enzyme
Jsrp1 -2,3077005594 7,35E-08 3,30E-06 junctional sarcoplasmic reticulum protein 1 Cytoplasm other
Ppm1e -2,302246505 7,68E-07 2,45E-05 protein phosphatase, Mg2+/Mn2+ dependent, 1E Nucleus phosphatase
Flnc -2,2961734732 1,55E-07 6,21E-06 filamin C, gamma Cytoplasm other
Smad9 -2,295442355 7,21E-06 0,0001712293 SMAD family member 9 Nucleus transcription regulator
Alpk3 -2,2863909276 7,03E-07 2,27E-05 alpha-kinase 3 Nucleus kinase
Npr3 -2,2817383405 5,37E-07 1,79E-05 natriuretic peptide receptor 3 Plasma Membrane G-protein coupled receptor
Fras1 -2,2791232259 1,29E-07 5,32E-06 Fraser extracellular matrix complex subunit 1 Extracellular Space other
Cmpk2 -2,2787940934 6,99E-06 0,0001666607 cytidine monophosphate (UMP-CMP) kinase 2, mitochondrial Cytoplasm kinase
Rbp7 -2,2760499231 1,43E-10 1,38E-08 retinol binding protein 7, cellular Cytoplasm other
Popdc3 -2,2702398464 1,86E-07 7,13E-06 popeye domain containing 3 Other other
Dusp26 -2,2627736738 1,97E-05 0,000409563 dual specificity phosphatase 26 (putative) Cytoplasm enzyme
Slc28a2 -2,2616673797 3,16E-06 8,47E-05 solute carrier family 28 (concentrative nucleoside transporter), member 2 Plasma Membrane transporter
Smyd1 -2,2578202344 1,29E-05 0,0002872314 SET and MYND domain containing 1 Nucleus transcription regulator
Tbx1 -2,2535657152 3,52E-09 2,42E-07 T-box 1 Nucleus transcription regulator
Tnni2 -2,2509555214 0,000114204 0,0017876709 troponin I type 2 (skeletal, fast) Cytoplasm enzyme
Ccl3 -2,249366721 9,28E-05 0,0015063479 chemokine (C-C motif) ligand 3-like 3 Extracellular Space cytokine
Slc16a4 -2,2473788712 7,91E-05 0,0013143003 solute carrier family 16, member 4 Plasma Membrane transporter
3425401B19Rik -2,2453941809 7,12E-06 0,0001693054 chromosome 10 open reading frame 71 Other other
Lrtm2 -2,2371297621 0,0002659964 0,0036354616 leucine-rich repeats and transmembrane domains 2 Other other
Sult1a1 -2,2368861465 3,44E-06 9,05E-05 sulfotransferase family 1A, phenol-preferring, member 1 Cytoplasm enzyme
Nrap -2,225366834 3,52E-06 9,21E-05 nebulin-related anchoring protein Cytoplasm other
Cacna1s -2,2160757853 1,08E-05 0,0002451586 calcium channel, voltage-dependent, L type, alpha 1S subunit Plasma Membrane ion channel
Mum1l1 -2,2134976366 0,0001791721 0,0026160655 melanoma associated antigen (mutated) 1-like 1 Cytoplasm other
Hk3 -2,2134069389 2,77E-12 4,31E-10 hexokinase 3 (white cell) Cytoplasm kinase
Camk2b -2,2094963558 6,55E-09 3,98E-07 calcium/calmodulin-dependent protein kinase II beta Cytoplasm kinase
Lamc3 -2,2081224163 5,95E-05 0,001026687 laminin, gamma 3 Extracellular Space other
Wnt10b -2,205958028 2,75E-06 7,53E-05 wingless-type MMTV integration site family, member 10B Extracellular Space other
Fam107a -2,2041088438 0,0002586932 0,003553395 family with sequence similarity 107, member A Nucleus other
2310002L09Rik -2,1941690085 5,30E-05 0,0009337987 RIKEN cDNA 2310002L09 gene Cytoplasm other
Meis1 -2,1933570384 1,42E-08 7,72E-07 Meis homeobox 1 Nucleus transcription regulator
Trdn -2,1922612482 4,81E-06 0,0001206056 triadin Cytoplasm other
Mlip -2,1872375725 3,41E-06 8,99E-05 muscular LMNA-interacting protein Nucleus other
Sh3bgr -2,1830333327 0,0002489727 0,0034336646 SH3-binding domain glutamic acid-rich protein Cytoplasm other
Prkag3 -2,1819393959 0,0001064001 0,0016828441 protein kinase, AMP-activated, gamma 3 non-catalytic subunit Cytoplasm other
Cacng1 -2,168663866 1,05E-06 3,25E-05 calcium channel, voltage-dependent, gamma subunit 1 Plasma Membrane ion channel
Sypl2 -2,1675629138 0,0003457368 0,0044919522 synaptophysin-like 2 Other other
Hspb1 -2,1595598549 4,85E-08 2,26E-06 heat shock 27kDa protein 1 Cytoplasm other
Dusp27 -2,1338945037 4,49E-09 2,91E-07 dual specificity phosphatase 27 (putative) Other phosphatase
Notum -2,1194953012 6,47E-06 0,0001560839 notum pectinacetylesterase homolog (Drosophila) Extracellular Space other
Pdk4 -2,119339262 9,60E-08 4,04E-06 pyruvate dehydrogenase kinase, isozyme 4 Cytoplasm kinase
Myo18b -2,1193011763 1,98E-06 5,66E-05 myosin XVIIIB Cytoplasm other
Trim72 -2,1151432273 1,68E-07 6,67E-06 tripartite motif containing 72, E3 ubiquitin protein ligase Cytoplasm enzyme
1500017E21Rik -2,1114567749 0,0002225197 0,0031481818 RIKEN cDNA 1500017E21 gene Other other
Cnih2 -2,0990123672 9,24E-06 0,000213856 cornichon family AMPA receptor auxiliary protein 2 Extracellular Space other
Mustn1 -2,0928115222 1,30E-06 3,91E-05 musculoskeletal, embryonic nuclear protein 1 Nucleus other
Rbm20 -2,0924172418 1,29E-05 0,0002871618 RNA binding motif protein 20 Nucleus other
Casq1 -2,090786079 0,0003888509 0,0049258049 calsequestrin 1 (fast-twitch, skeletal muscle) Cytoplasm other
H19 -2,0874720503 7,92E-13 1,39E-10 H19, imprinted maternally expressed transcript (non-protein coding) Cytoplasm other
Tlr7 -2,0715018314 8,33E-08 3,64E-06 toll-like receptor 7 Plasma Membrane transmembrane receptor
Kcnc3 -2,069589796 1,37E-06 4,09E-05 potassium channel, voltage gated Shaw related subfamily C, member 3 Plasma Membrane ion channel
Twist1 -2,0665237438 1,16E-16 5,49E-14 twist family bHLH transcription factor 1 Nucleus transcription regulator
Galnt3 -2,0600685111 4,79E-05 0,0008613647 polypeptide N-acetylgalactosaminyltransferase 3 Cytoplasm enzyme
Aldoart2 -2,0562388537 0,000286733 0,0038534318 aldolase 1 A, retrogene 2 Other enzyme
Bves -2,0526262248 1,58E-05 0,0003381397 blood vessel epicardial substance Plasma Membrane other
Myf6 -2,0416618308 0,0002429648 0,0033727 myogenic factor 6 (herculin) Nucleus transcription regulator
Sgms2 -2,0410252914 2,31E-05 0,0004673089 sphingomyelin synthase 2 Plasma Membrane enzyme
Mrc1 -2,0389149214 2,65E-15 1,04E-12 mannose receptor, C type 1 Plasma Membrane transmembrane receptor
Slc8a3 -2,0361408029 1,48E-06 4,37E-05 solute carrier family 8 (sodium/calcium exchanger), member 3 Plasma Membrane transporter
Mx1 -2,0329632492 4,78E-05 0,0008596134 MX dynamin-like GTPase 1 Nucleus enzyme
Dlx5 -2,0136073507 9,59E-05 0,0015485895 distal-less homeobox 5 Nucleus transcription regulator
Cd180 -1,9999888987 3,23E-09 2,28E-07 CD180 molecule Plasma Membrane other
Hspb2 -1,9991448351 2,17E-06 6,09E-05 heat shock 27kDa protein 2 Cytoplasm other
Penk -1,9920735886 5,76E-05 0,0009976139 proenkephalin Extracellular Space other
Phospho1 -1,9748386545 3,91E-05 0,0007370669 phosphatase, orphan 1 Extracellular Space enzyme
Colq -1,971439149 2,18E-05 0,0004419249 collagen-like tail subunit (single strand of homotrimer) of asymmetric acetylcholinesterase Extracellular Space other
Myom1 -1,9644311431 8,37E-05 0,0013771314 myomesin 1 Cytoplasm other
Eef1a2 -1,9639243981 6,39E-06 0,0001548096 eukaryotic translation elongation factor 1 alpha 2 Cytoplasm translation regulator
Ovol1 -1,9629718595 3,25E-05 0,0006310341 ovo-like zinc finger 1 Nucleus transcription regulator
Lrrc2 -1,9627188118 3,67E-06 9,57E-05 leucine rich repeat containing 2 Other other
Ccl12 -1,9604166943 4,35E-09 2,83E-07 chemokine (C-C motif) ligand 2 Extracellular Space cytokine
Otud1 -1,9582092997 1,43E-08 7,74E-07 OTU deubiquitinase 1 Other peptidase
Lonrf3 -1,9558398156 6,45E-09 3,96E-07 LON peptidase N-terminal domain and ring finger 3 Other other
Bai1 -1,9557246538 0,0001009963 0,0016160593 Err:512 Err:512 Err:512
Hoxc9 -1,9556608584 1,01E-12 1,72E-10 homeobox C9 Nucleus transcription regulator
Arpp21 -1,945185646 0,0002332773 0,0032665983 cAMP-regulated phosphoprotein, 21kDa Cytoplasm other
Obscn -1,939029162 0,0002063891 0,0029597579 obscurin, cytoskeletal calmodulin and titin-interacting RhoGEF Cytoplasm kinase
Trem2 -1,9364460628 4,42E-08 2,09E-06 triggering receptor expressed on myeloid cells 2 Plasma Membrane transmembrane receptor
Tpm1 -1,9333159873 6,35E-05 0,0010835893 tropomyosin 1, alpha Plasma Membrane other
Mb -1,9272409814 4,29E-10 3,79E-08 myoglobin Cytoplasm transporter
Coro6 -1,923052355 9,30E-05 0,0015066949 coronin 6 Extracellular Space other
Satb2 -1,9221588552 0,0001134213 0,0017774529 SATB homeobox 2 Nucleus transcription regulator
Dlgap3 -1,9213737133 0,0003417198 0,0044482103 discs, large (Drosophila) homolog-associated protein 3 Cytoplasm other
Ptn -1,9153143395 0,0001669623 0,0024714031 pleiotrophin Extracellular Space growth factor
Bmp5 -1,9053999219 3,70E-05 0,0007001959 bone morphogenetic protein 5 Extracellular Space growth factor
Ttn -1,9013482409 0,0003645852 0,0046703094 titin Cytoplasm kinase
Art1 -1,901283303 1,60E-05 0,0003407107 ADP-ribosyltransferase 1 Plasma Membrane enzyme
Sybu -1,9008965981 7,80E-06 0,0001829949 syntabulin (syntaxin-interacting) Other other
Tex15 -1,9003784172 1,57E-05 0,0003379675 testis expressed 15 Extracellular Space other
Wnt5a 1,9069405946 1,25E-07 5,19E-06 wingless-type MMTV integration site family, member 5A Extracellular Space cytokine
Ero1l 1,9109747334 1,72E-11 2,18E-09 endoplasmic reticulum oxidoreductase alpha Cytoplasm enzyme
Cyp7b1 1,9131280746 6,13E-05 0,0010537647 cytochrome P450, family 7, subfamily B, polypeptide 1 Cytoplasm enzyme
Timp1 1,9143910707 1,11E-09 8,91E-08 TIMP metallopeptidase inhibitor 1 Extracellular Space cytokine
Bhlhe22 1,9200774466 0,0002433196 0,0033727 basic helix-loop-helix family, member e22 Nucleus transcription regulator
Clca5 1,9222526464 0,0001289967 0,0019800765 Err:512 Err:512 Err:512
Nos2 1,9344905204 9,69E-06 0,0002235862 nitric oxide synthase 2, inducible Cytoplasm enzyme
Sdc1 1,9347650433 1,90E-12 3,13E-10 syndecan 1 Plasma Membrane enzyme
Ccl11 1,9356854854 1,01E-05 0,0002318802 chemokine (C-C motif) ligand 11 Extracellular Space cytokine
Sfrp2 1,9375704316 0,0001053902 0,0016707332 secreted frizzled-related protein 2 Plasma Membrane transmembrane receptor
Adora2b 1,9377193184 1,43E-06 4,25E-05 adenosine A2b receptor Plasma Membrane G-protein coupled receptor
C1rb 1,948311191 6,64E-06 0,0001593925 complement component 1, r subcomponent Extracellular Space peptidase
Cadm3 1,9548688009 1,65E-06 4,81E-05 cell adhesion molecule 3 Plasma Membrane other
Gcnt4 1,9591464224 5,60E-05 0,0009745424 glucosaminyl (N-acetyl) transferase 4, core 2 Cytoplasm enzyme
AA467197 1,95975912 0,0001344618 0,0020530929 chromosome 15 open reading frame 48 Nucleus other
Adamts5 1,9655031303 1,70E-12 2,84E-10 ADAM metallopeptidase with thrombospondin type 1 motif, 5 Extracellular Space peptidase
Il6 1,9697078203 3,32E-05 0,0006424258 interleukin 6 Extracellular Space cytokine
Acp5 1,9720844805 1,73E-05 0,0003658452 acid phosphatase 5, tartrate resistant Cytoplasm phosphatase
Plac8 1,9726542288 1,03E-06 3,18E-05 placenta-specific 8 Nucleus other
Hic1 1,9776663709 2,86E-10 2,64E-08 hypermethylated in cancer 1 Nucleus transcription regulator
Il18rap 1,9880470687 1,49E-05 0,0003247781 interleukin 18 receptor accessory protein Plasma Membrane transmembrane receptor
Prss46 2,005016082 0,0001525426 0,0022883055 protease, serine, 46 Other peptidase
Csgalnact1 2,0068328917 2,07E-12 3,33E-10 chondroitin sulfate N-acetylgalactosaminyltransferase 1 Cytoplasm enzyme
Phlda2 2,0120954522 0,000118738 0,0018480707 pleckstrin homology-like domain, family A, member 2 Cytoplasm other
Barx2 2,0139643819 1,83E-06 5,25E-05 BARX homeobox 2 Nucleus transcription regulator
Kctd11 2,0201232426 8,08E-12 1,11E-09 potassium channel tetramerization domain containing 11 Cytoplasm other
Hilpda 2,0227794241 7,35E-08 3,30E-06 hypoxia inducible lipid droplet-associated Cytoplasm other
Klhdc8a 2,0291635706 7,64E-06 0,0001801151 kelch domain containing 8A Other other
Crabp2 2,0410006948 3,54E-05 0,0006761489 cellular retinoic acid binding protein 2 Cytoplasm transporter
Medag 2,0449718306 1,99E-12 3,24E-10 mesenteric estrogen-dependent adipogenesis Cytoplasm other
Napsa 2,0508081665 6,69E-08 3,03E-06 napsin A aspartic peptidase Extracellular Space peptidase
Col23a1 2,0746154765 6,01E-09 3,72E-07 collagen, type XXIII, alpha 1 Plasma Membrane other
Wnt2b 2,0775644591 1,78E-05 0,0003737783 wingless-type MMTV integration site family, member 2B Extracellular Space other
Lgi3 2,0831858983 0,0002010135 0,002897856 leucine-rich repeat LGI family, member 3 Extracellular Space other
Il33 2,0846464546 5,04E-11 5,30E-09 interleukin 33 Extracellular Space cytokine
H2-Ab1 2,0874682966 1,75E-09 1,30E-07 major histocompatibility complex, class II, DQ beta 1 Plasma Membrane other
4930502E18Rik 2,0875906062 5,50E-05 0,0009614585 RIKEN cDNA 4930502E18 gene Other other
Osr1 2,1140930409 1,84E-08 9,56E-07 odd-skipped related transciption factor 1 Nucleus other
Serping1 2,1162586167 1,60E-13 3,43E-11 serpin peptidase inhibitor, clade G (C1 inhibitor), member 1 Extracellular Space other
P2ry10 2,1176600139 6,16E-05 0,0010554986 purinergic receptor P2Y, G-protein coupled, 10 Plasma Membrane G-protein coupled receptor
Ddit4 2,1207273552 1,04E-15 4,32E-13 DNA-damage-inducible transcript 4 Cytoplasm other
Tmeff2 2,1238497575 0,0002865921 0,0038534318 transmembrane protein with EGF-like and two follistatin-like domains 2 Cytoplasm other
Pthlh 2,1259957502 3,81E-05 0,0007191955 parathyroid hormone-like hormone Extracellular Space other
Pla1a 2,1282975021 3,15E-12 4,77E-10 phospholipase A1 member A Extracellular Space enzyme
Cwc22 2,1311284838 0,0002890768 0,0038735159 CWC22 spliceosome-associated protein Nucleus other
Adamts4 2,1319106258 9,89E-12 1,30E-09 ADAM metallopeptidase with thrombospondin type 1 motif, 4 Extracellular Space peptidase
Ocstamp 2,1337076217 0,0002859088 0,0038499195 osteoclast stimulatory transmembrane protein Other other
Avpr1a 2,1350587993 3,05E-08 1,49E-06 arginine vasopressin receptor 1A Plasma Membrane G-protein coupled receptor
Sphk1 2,1375776274 5,04E-10 4,42E-08 sphingosine kinase 1 Cytoplasm kinase
Alox12 2,1477034586 7,02E-05 0,0011790923 arachidonate 12-lipoxygenase Cytoplasm enzyme
Cd74 2,154265386 8,23E-10 6,87E-08 CD74 molecule, major histocompatibility complex, class II invariant chain Plasma Membrane transmembrane receptor
Ier3 2,1564131609 6,99E-10 5,94E-08 immediate early response 3 Cytoplasm other
Niacr1 2,1610174589 4,17E-06 0,0001071082 Err:512 Err:512 Err:512
Galnt16 2,1633322133 1,33E-11 1,70E-09 polypeptide N-acetylgalactosaminyltransferase 16 Cytoplasm enzyme
Fam83f 2,1634644566 9,66E-05 0,0015573137 family with sequence similarity 83, member F Other other
Phyhipl 2,1669207089 0,0003526029 0,0045526963 phytanoyl-CoA 2-hydroxylase interacting protein-like Cytoplasm other
H2-Aa 2,1697429797 3,79E-09 2,53E-07 major histocompatibility complex, class II, DQ alpha 1 Plasma Membrane transmembrane receptor
Il1rl1 2,175512643 4,51E-06 0,0001146525 interleukin 1 receptor-like 1 Plasma Membrane transmembrane receptor
Dpt 2,1800125464 2,29E-13 4,74E-11 dermatopontin Extracellular Space other
Kcnj15 2,1806732045 1,37E-08 7,48E-07 potassium channel, inwardly rectifying subfamily J, member 15 Plasma Membrane ion channel
Rnd1 2,1819676606 9,57E-09 5,64E-07 Rho family GTPase 1 Cytoplasm enzyme
Gpr114 2,1896462967 1,94E-07 7,41E-06 Err:512 Err:512 Err:512
Ccbp2 2,1939046346 2,20E-07 8,27E-06 Err:512 Err:512 Err:512
Elfn1 2,1994673663 4,19E-05 0,0007762324 extracellular leucine-rich repeat and fibronectin type III domain containing 1 Plasma Membrane other
Cxadr 2,2008285797 0,0003329927 0,0043511681 coxsackie virus and adenovirus receptor Plasma Membrane transmembrane receptor
Mcpt4 2,2062544846 0,0001693429 0,0024965309 mast cell protease 4 Other peptidase
Stac2 2,2123660385 9,24E-09 5,50E-07 SH3 and cysteine rich domain 2 Other other
Cxcr7 2,2164068787 4,59E-14 1,16E-11 Err:512 Err:512 Err:512
Foxd1 2,2171416872 2,09E-05 0,0004272157 forkhead box D1 Nucleus transcription regulator
Cd209f 2,2324306028 0,0001401481 0,0021186368 CD209f antigen Other other
Crabp1 2,2350538061 0,0003778686 0,0048134271 cellular retinoic acid binding protein 1 Cytoplasm transporter
Rtn4rl2 2,2365868607 9,06E-08 3,91E-06 reticulon 4 receptor-like 2 Plasma Membrane other
Slc39a14 2,238458836 2,66E-14 7,14E-12 solute carrier family 39 (zinc transporter), member 14 Plasma Membrane transporter
Ifnlr1 2,2427740237 1,94E-05 0,000403681 interferon, lambda receptor 1 Plasma Membrane transmembrane receptor
5730416F02Rik 2,2538012286 5,43E-06 0,0001330703 capping protein (actin filament), gelsolin-like pseudogene Other other
Trpm6 2,2576937598 2,03E-07 7,73E-06 transient receptor potential cation channel, subfamily M, member 6 Plasma Membrane kinase
Gfra1 2,2583619818 1,82E-06 5,23E-05 GDNF family receptor alpha 1 Plasma Membrane transmembrane receptor
Egln3 2,2602272203 4,82E-15 1,69E-12 egl-9 family hypoxia-inducible factor 3 Cytoplasm enzyme
S100a9 2,2610770768 0,0002366314 0,0033067965 S100 calcium binding protein A9 Cytoplasm other
Fbln2 2,2616569353 3,57E-10 3,21E-08 fibulin 2 Extracellular Space other
Tnfsf11 2,2622207658 0,0001685651 0,0024892914 tumor necrosis factor (ligand) superfamily, member 11 Extracellular Space cytokine
S1pr3 2,2680648897 8,22E-08 3,62E-06 sphingosine-1-phosphate receptor 3 Plasma Membrane G-protein coupled receptor
Acsbg1 2,2720632096 2,08E-05 0,0004259564 acyl-CoA synthetase bubblegum family member 1 Cytoplasm enzyme
Kcne3 2,2822848304 7,11E-11 7,32E-09 potassium channel, voltage gated subfamily E regulatory beta subunit 3 Plasma Membrane ion channel
Lmx1a 2,2862950666 8,74E-05 0,0014292832 LIM homeobox transcription factor 1, alpha Nucleus transcription regulator
Sfrp1 2,2866459321 0,0003828327 0,0048630771 secreted frizzled-related protein 1 Plasma Membrane transmembrane receptor
Aqp2 2,2948733594 3,68E-05 0,0006990255 aquaporin 2 (collecting duct) Plasma Membrane transporter
1810033B17Rik 2,3037509066 1,76E-05 0,0003719818 Err:512 Err:512 Err:512
Tmem178 2,3083875095 8,85E-06 0,0002051263 transmembrane protein 178A Other other
Figf 2,3246159117 3,70E-09 2,48E-07 c-fos induced growth factor (vascular endothelial growth factor D) Extracellular Space growth factor
Slc6a2 2,3273864485 5,24E-05 0,0009261293 solute carrier family 6 (neurotransmitter transporter), member 2 Plasma Membrane transporter
Gpr123 2,333408054 2,32E-07 8,66E-06 Err:512 Err:512 Err:512
Ces2g 2,3378235095 2,07E-05 0,0004242945 carboxylesterase 2G Other enzyme
Treml4 2,3561846468 7,19E-05 0,0012049698 triggering receptor expressed on myeloid cells-like 4 Other other
Doc2b 2,3562635343 0,0001438927 0,0021704484 double C2-like domains, beta Cytoplasm transporter
Lbp 2,3580344403 5,08E-13 9,65E-11 lipopolysaccharide binding protein Plasma Membrane transporter
Ifi205 2,3607474961 5,29E-14 1,32E-11 interferon, gamma-inducible protein 16 Nucleus transcription regulator
Rgs9 2,3608171109 6,48E-05 0,0011014854 regulator of G-protein signaling 9 Cytoplasm enzyme
Arsi 2,3712686248 2,16E-08 1,09E-06 arylsulfatase family, member I Extracellular Space enzyme
Ciita 2,3737724012 6,36E-07 2,08E-05 class II, major histocompatibility complex, transactivator Nucleus transcription regulator
Dusp4 2,3793883457 2,36E-06 6,57E-05 dual specificity phosphatase 4 Nucleus phosphatase
Rorb 2,3808978402 0,0002953095 0,0039377477 RAR-related orphan receptor B Nucleus ligand-dependent nuclear receptor
Sbsn 2,383926086 1,80E-05 0,0003776836 suprabasin Cytoplasm other
Cdh1 2,3927691811 3,51E-08 1,69E-06 cadherin 1, type 1 Plasma Membrane other
Fgr 2,3959977219 1,73E-17 9,84E-15 FGR proto-oncogene, Src family tyrosine kinase Nucleus kinase
Kcnip1 2,4014742171 1,95E-05 0,0004058051 Kv channel interacting protein 1 Plasma Membrane ion channel
Ak4 2,4018466994 5,33E-08 2,46E-06 adenylate kinase 4 Cytoplasm kinase
A630023A22Rik 2,414937431 7,63E-05 0,0012728828 RIKEN cDNA A630023A22 gene Other other
Has1 2,4174245023 1,62E-08 8,57E-07 hyaluronan synthase 1 Plasma Membrane enzyme
Sdk1 2,4247205936 1,24E-08 6,91E-07 sidekick cell adhesion molecule 1 Plasma Membrane other
Gjb5 2,429127176 6,04E-06 0,0001468549 gap junction protein, beta 5, 31.1kDa Plasma Membrane transporter
5730559C18Rik 2,4349327649 5,17E-05 0,0009198264 chromosome 1 open reading frame 106 Other other
Adm 2,4377964729 3,10E-09 2,21E-07 adrenomedullin Extracellular Space other
Hmga2 2,4451415929 1,35E-05 0,0002978128 high mobility group AT-hook 2 Nucleus enzyme
Itgax 2,4490586308 3,26E-16 1,49E-13 integrin, alpha X (complement component 3 receptor 4 subunit) Plasma Membrane transmembrane receptor
Itln1 2,454578666 3,52E-07 1,24E-05 intelectin 1 (galactofuranose binding) Plasma Membrane other
Ifitm1 2,4549025353 1,49E-13 3,24E-11 interferon induced transmembrane protein 1 Other other
Sh2d5 2,4579661128 5,17E-13 9,68E-11 SH2 domain containing 5 Plasma Membrane other
Ndufa4l2 2,4612733009 4,04E-12 5,88E-10 NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 4-like 2 Other enzyme
Ffar2 2,4722092141 0,0002263316 0,0031856406 free fatty acid receptor 2 Plasma Membrane G-protein coupled receptor
Scd1 2,488851454 3,66E-07 1,28E-05 stearoyl-CoA desaturase (delta-9-desaturase) Cytoplasm enzyme
Mmp9 2,499142569 4,69E-05 0,0008452829 matrix metallopeptidase 9 Extracellular Space peptidase
Cxcl1 2,5153765405 3,67E-05 0,0006967073 chemokine (C-X-C motif) ligand 2 Extracellular Space cytokine
Nrn1 2,5205155569 2,96E-06 8,01E-05 neuritin 1 Cytoplasm other
Inhbb 2,5230108142 5,62E-13 1,04E-10 inhibin, beta B Extracellular Space growth factor
Col28a1 2,5311824591 9,55E-07 2,97E-05 collagen, type XXVIII, alpha 1 Extracellular Space other
Dnmt3l 2,532511137 0,0002695057 0,0036687639 DNA (cytosine-5-)-methyltransferase 3-like Nucleus transcription regulator
Fcrla 2,5352892135 0,0001400172 0,0021186368 Fc receptor-like A Plasma Membrane other
Cxcl14 2,5386683962 1,33E-09 1,04E-07 chemokine (C-X-C motif) ligand 14 Extracellular Space cytokine
Pi16 2,5454176851 9,68E-08 4,06E-06 peptidase inhibitor 16 Extracellular Space other
C4b 2,5543273887 3,52E-09 2,42E-07 complement component 4B (Chido blood group) Extracellular Space other
Gzmc 2,5867195956 2,23E-06 6,24E-05 granzyme C Cytoplasm peptidase
Car9 2,5882215762 5,94E-09 3,70E-07 carbonic anhydrase IX Nucleus enzyme
H2-Eb1 2,5893619923 4,28E-11 4,72E-09 major histocompatibility complex, class II, DR beta 5 Plasma Membrane transmembrane receptor
Cpa3 2,5922247079 0,0003499471 0,004527105 carboxypeptidase A3 (mast cell) Extracellular Space peptidase
Rhov 2,597158136 3,97E-05 0,0007442687 ras homolog family member V Plasma Membrane enzyme
Smoc1 2,6092766592 1,13E-17 7,01E-15 SPARC related modular calcium binding 1 Extracellular Space other
Cd244 2,6106259629 2,48E-09 1,82E-07 CD244 molecule, natural killer cell receptor 2B4 Plasma Membrane transmembrane receptor
Serpina3h 2,6260865461 9,48E-16 4,05E-13 serine (or cysteine) peptidase inhibitor, clade A, member 3H Extracellular Space other
Dpp6 2,6272698949 3,28E-06 8,69E-05 dipeptidyl-peptidase 6 Plasma Membrane other
Tmem95 2,6306048459 3,26E-06 8,64E-05 transmembrane protein 95 Other other
Rgs16 2,6415039443 7,07E-14 1,61E-11 regulator of G-protein signaling 16 Cytoplasm other
Mmp12 2,6613871376 8,47E-12 1,14E-09 matrix metallopeptidase 12 Extracellular Space peptidase
Ttyh1 2,6663102685 1,17E-08 6,67E-07 tweety family member 1 Plasma Membrane ion channel
Tmem125 2,6845927384 0,0002089896 0,0029845376 transmembrane protein 125 Other other
Pcsk5 2,6881810914 2,88E-12 4,42E-10 proprotein convertase subtilisin/kexin type 5 Extracellular Space peptidase
Slc2a1 2,7010769556 2,95E-13 5,94E-11 solute carrier family 2 (facilitated glucose transporter), member 1 Plasma Membrane transporter
Frmd5 2,7042557706 7,70E-05 0,0012828347 FERM domain containing 5 Other other
Col5a3 2,7069948839 9,13E-22 9,61E-19 collagen, type V, alpha 3 Extracellular Space other
Dmkn 2,7111406883 0,0003443895 0,0044787007 dermokine Extracellular Space other
Lrrc15 2,7171280297 3,40E-12 5,06E-10 leucine rich repeat containing 15 Plasma Membrane other
C3 2,7269008192 2,87E-10 2,64E-08 complement component 3 Extracellular Space peptidase
Nt5e 2,7329472131 7,62E-12 1,05E-09 5'-nucleotidase, ecto (CD73) Plasma Membrane phosphatase
Serpind1 2,7626239864 9,29E-07 2,91E-05 serpin peptidase inhibitor, clade D (heparin cofactor), member 1 Extracellular Space other
Unc13a 2,7726850312 1,69E-09 1,27E-07 unc-13 homolog A (C. elegans) Plasma Membrane other
Tpsb2 2,7806510063 8,40E-08 3,65E-06 tryptase alpha/beta 1 Extracellular Space peptidase
Inhba 2,7953383322 3,18E-08 1,54E-06 inhibin, beta A Extracellular Space growth factor
C4a 2,7987311043 2,61E-10 2,44E-08 complement component 4B (Chido blood group) Extracellular Space other
Slc2a3 2,8106978677 2,17E-13 4,56E-11 solute carrier family 2 (facilitated glucose transporter), member 3 Plasma Membrane transporter
Wt1 2,82046663 0,0001696783 0,002498782 Wilms tumor 1 Nucleus transcription regulator
1300002K09Rik 2,8342745076 7,10E-07 2,29E-05 Err:512 Err:512 Err:512
Vat1l 2,8423187963 6,74E-05 0,0011386776 vesicle amine transport 1-like Other enzyme
Il1b 2,842564699 3,03E-24 3,76E-21 interleukin 1, beta Extracellular Space cytokine
Gjb3 2,8506647858 2,13E-06 6,01E-05 gap junction protein, beta 3, 31kDa Plasma Membrane transporter
Sfrp4 2,8734310411 5,56E-14 1,35E-11 secreted frizzled-related protein 4 Plasma Membrane transmembrane receptor
Osbp2 2,8942675794 8,23E-11 8,28E-09 oxysterol binding protein 2 Cytoplasm other
Serpina3i 2,8967330407 3,59E-11 4,13E-09 serine (or cysteine) peptidase inhibitor, clade A, member 3G Other other
Ccbe1 2,8995915956 1,80E-14 5,13E-12 collagen and calcium binding EGF domains 1 Extracellular Space other
Dnase1l3 2,9140956117 4,71E-11 5,03E-09 deoxyribonuclease I-like 3 Nucleus enzyme
Prg4 2,9206520954 1,48E-13 3,24E-11 proteoglycan 4 (megakaryocyte stimulating factor, articular superficial zone protein) Extracellular Space other
Serpine1 2,9431123294 2,65E-13 5,41E-11 serpin peptidase inhibitor, clade E (nexin, plasminogen activator inhibitor type 1), member 1 Extracellular Space other
Nfasc 2,9553860813 4,40E-11 4,81E-09 neurofascin Plasma Membrane other
Tnfsf8 2,9580980252 7,42E-08 3,31E-06 tumor necrosis factor (ligand) superfamily, member 8 Plasma Membrane cytokine
Adra2a 2,9630892368 3,96E-36 1,36E-32 adrenoceptor alpha 2A Plasma Membrane G-protein coupled receptor
Syt5 2,9678626987 1,03E-09 8,32E-08 synaptotagmin V Cytoplasm transporter
Erv3 2,9762539953 1,57E-05 0,0003379675 endogenous retroviral sequence 3 Other other
Lgi2 2,9802806685 7,82E-07 2,49E-05 leucine-rich repeat LGI family, member 2 Extracellular Space other
Adcy5 2,9897455481 1,35E-15 5,42E-13 adenylate cyclase 5 Plasma Membrane enzyme
Lcn2 3,0056929099 4,88E-09 3,09E-07 lipocalin 2 Extracellular Space transporter
Syt17 3,0120742606 1,31E-06 3,93E-05 synaptotagmin XVII Plasma Membrane other
Efemp1 3,0132171131 2,56E-07 9,39E-06 EGF containing fibulin-like extracellular matrix protein 1 Extracellular Space enzyme
Fam5c 3,0227690304 1,82E-07 7,05E-06 Err:512 Err:512 Err:512
Sorcs1 3,0453556883 1,60E-05 0,0003409679 sortilin-related VPS10 domain containing receptor 1 Plasma Membrane transporter
Adamts15 3,054035878 7,61E-15 2,54E-12 ADAM metallopeptidase with thrombospondin type 1 motif, 15 Extracellular Space peptidase
Clec2e 3,0763578096 6,84E-06 0,0001635351 C-type lectin domain family 2, member h Plasma Membrane transmembrane receptor
Chl1 3,0846062317 5,81E-07 1,92E-05 cell adhesion molecule L1-like Plasma Membrane other
Mmrn1 3,1101689322 0,0001043427 0,0016560467 multimerin 1 Extracellular Space other
Gpr35 3,1187264919 3,52E-19 2,84E-16 G protein-coupled receptor 35 Plasma Membrane G-protein coupled receptor
Rarres2 3,1472762591 1,10E-09 8,87E-08 retinoic acid receptor responder (tazarotene induced) 2 Plasma Membrane transmembrane receptor
Pgf 3,1505506792 8,07E-17 4,09E-14 placental growth factor Extracellular Space growth factor
Serpina3f 3,1553756415 5,61E-14 1,35E-11 serine (or cysteine) peptidase inhibitor, clade A, member 3G Other other
Il1r2 3,1558180965 9,76E-15 3,11E-12 interleukin 1 receptor, type II Plasma Membrane transmembrane receptor
Il13ra2 3,2087123722 4,27E-05 0,0007861961 interleukin 13 receptor, alpha 2 Plasma Membrane transmembrane receptor
Nxph4 3,2124295511 1,20E-08 6,76E-07 neurexophilin 4 Extracellular Space other
Slit1 3,2215218659 8,08E-08 3,56E-06 slit guidance ligand 1 Extracellular Space other
Col10a1 3,2552996895 8,78E-08 3,80E-06 collagen, type X, alpha 1 Extracellular Space other
Grem1 3,3069988414 4,81E-09 3,07E-07 gremlin 1, DAN family BMP antagonist Extracellular Space other
Rpl21 3,3191584943 0,0003215949 0,0042264554 ribosomal protein L21 Cytoplasm other
Ly6k 3,3302102629 1,32E-05 0,0002925648 lymphocyte antigen 6 complex, locus K Nucleus other
Pcsk9 3,3436628717 1,11E-05 0,0002497527 proprotein convertase subtilisin/kexin type 9 Extracellular Space peptidase
Dbx2 3,3741247116 8,16E-10 6,85E-08 developing brain homeobox 2 Nucleus transcription regulator
B3galt5 3,42887914 3,17E-06 8,47E-05 UDP-Gal:betaGlcNAc beta 1,3-galactosyltransferase, polypeptide 5 Cytoplasm enzyme
Il11 3,446479515 1,36E-08 7,48E-07 interleukin 11 Extracellular Space cytokine
Htr1b 3,4700924697 2,52E-14 6,90E-12 5-hydroxytryptamine (serotonin) receptor 1B, G protein-coupled Plasma Membrane G-protein coupled receptor
Cxcl13 3,5547993869 3,92E-05 0,0007372354 chemokine (C-X-C motif) ligand 13 Extracellular Space cytokine
9330182L06Rik 3,5991544866 3,76E-06 9,79E-05 KIAA1324-like Other other
Cd207 3,6985009787 6,80E-11 7,05E-09 CD207 molecule, langerin Plasma Membrane other
Serpina3n 3,6993293723 1,12E-13 2,51E-11 serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 3 Extracellular Space other
Tmem132e 3,7067364259 9,24E-08 3,94E-06 transmembrane protein 132E Other other
Serpina3m 3,7222266038 5,48E-18 3,57E-15 serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 3 Extracellular Space other
Kcnmb1 3,7715525937 2,30E-08 1,15E-06 potassium channel subfamily M regulatory beta subunit 1 Plasma Membrane ion channel
Gpr141 3,8983535425 1,01E-10 9,98E-09 G protein-coupled receptor 141 Plasma Membrane G-protein coupled receptor
Arg1 3,924911517 2,76E-08 1,37E-06 arginase 1 Cytoplasm enzyme
Tpsab1 3,958781996 9,04E-15 2,94E-12 tryptase alpha/beta 1 Nucleus peptidase
Ereg 3,9936044165 9,00E-07 2,82E-05 epiregulin Extracellular Space growth factor
Mmp13 4,0251327052 3,95E-15 1,42E-12 matrix metallopeptidase 13 Extracellular Space peptidase
Tnfrsf9 4,1000432435 1,10E-24 1,67E-21 tumor necrosis factor receptor superfamily, member 9 Plasma Membrane transmembrane receptor
Slc7a11 4,1230641662 1,29E-17 7,68E-15 solute carrier family 7 (anionic amino acid transporter light chain, xc- system), member 11 Plasma Membrane transporter
Akr1c18 4,1339602735 1,07E-11 1,40E-09 aldo-keto reductase family 1, member C3 Cytoplasm enzyme
Mgarp 4,2158539106 3,91E-11 4,38E-09 mitochondria-localized glutamic acid-rich protein Cytoplasm other
Serpina3k 4,2584780951 6,90E-13 1,26E-10 serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 3 Extracellular Space other
Ccl20 4,3256578414 1,88E-10 1,80E-08 chemokine (C-C motif) ligand 20 Extracellular Space cytokine
Cfi 4,5899335826 1,17E-09 9,20E-08 complement factor I Extracellular Space peptidase
Reg3g 4,6641211698 1,46E-12 2,47E-10 regenerating islet-derived 3 gamma Extracellular Space other
Krt19 4,7792418952 1,21E-05 0,0002719027 keratin 19, type I Cytoplasm other
Ptprn 4,8246859963 6,13E-22 6,98E-19 protein tyrosine phosphatase, receptor type, N Plasma Membrane phosphatase
A2m 4,9366443654 1,29E-07 5,33E-06 alpha-2-macroglobulin Extracellular Space transporter
Saa3 4,9381905541 6,34E-08 2,88E-06 serum amyloid A 3 Extracellular Space other
Gzme 5,281856145 6,57E-14 1,52E-11 granzyme H (cathepsin G-like 2, protein h-CCPX) Cytoplasm peptidase
Mmp3 5,4476607139 4,84E-28 8,28E-25 matrix metallopeptidase 3 Extracellular Space peptidase
Prokr2 5,9033135539 2,25E-14 6,29E-12 prokineticin receptor 2 Plasma Membrane G-protein coupled receptor
Fgf23 6,2232739132 4,42E-14 1,14E-11 fibroblast growth factor 23 Extracellular Space growth factor
Mcpt2 6,8573049814 4,93E-30 1,12E-26 mast cell protease 2 Extracellular Space peptidase
Gzmd 7,2483935421 7,17E-13 1,29E-10 granzyme H (cathepsin G-like 2, protein h-CCPX) Cytoplasm peptidase
Cldn10 7,6368083658 5,34E-09 3,35E-07 claudin 10 Plasma Membrane other
Mmp10 7,6454322902 2,07E-24 2,84E-21 matrix metallopeptidase 10 Extracellular Space peptidase
Gm9992 7,648664919 4,61E-08 2,16E-06 unc-93 homolog A (C. elegans) Plasma Membrane other
Mcpt8 8,1171694198 5,31E-12 7,57E-10 mast cell protease 8 Cytoplasm other
Reg1 10,7468584634 8,53E-19 6,49E-16 regenerating islet-derived 1 alpha Extracellular Space growth factor
Mcpt1 11,2538222733 2,86E-47 3,92E-43 mast cell protease 1 Other peptidase

 


Share this article / Teilen Sie diesen Artikel