Skip to main content

Evaluation of phenoxybenzamine in the CFA model of pain following gene expression studies and connectivity mapping

Abstract

Background

We have previously used the rat 4 day Complete Freund's Adjuvant (CFA) model to screen compounds with potential to reduce osteoarthritic pain. The aim of this study was to identify genes altered in this model of osteoarthritic pain and use this information to infer analgesic potential of compounds based on their own gene expression profiles using the Connectivity Map approach.

Results

Using microarrays, we identified differentially expressed genes in L4 and L5 dorsal root ganglia (DRG) from rats that had received intraplantar CFA for 4 days compared to matched, untreated control animals. Analysis of these data indicated that the two groups were distinguishable by differences in genes important in immune responses, nerve growth and regeneration. This list of differentially expressed genes defined a "CFA signature". We used the Connectivity Map approach to identify pharmacologic agents in the Broad Institute Build02 database that had gene expression signatures that were inversely related ('negatively connected') with our CFA signature. To test the predictive nature of the Connectivity Map methodology, we tested phenoxybenzamine (an alpha adrenergic receptor antagonist) - one of the most negatively connected compounds identified in this database - for analgesic activity in the CFA model. Our results indicate that at 10 mg/kg, phenoxybenzamine demonstrated analgesia comparable to that of Naproxen in this model.

Conclusion

Evaluation of phenoxybenzamine-induced analgesia in the current study lends support to the utility of the Connectivity Map approach for identifying compounds with analgesic properties in the CFA model.

Background

Several recent studies have characterized preclinical pain models with the goal of defining gene expression profiles related to different kinds of pain [1–9]. An understanding of gene alterations associated with pain is important as these may open up new pathways for targeting pain. While there are studies that describe gene expression profiles following nerve transection or nerve ligation models in animals [1–5, 9], reports defining gene expression in studies that utilize inflammatory models such as the Complete Freund's Adjuvant (CFA) model are scarce. The rat CFA model yields acute inflammation and pain as measured by tactile allodynia [10–12], changes in weight bearing, and paw pressure withdrawal thresholds [13]. We have used this model for screening potential compounds for osteoarthritic pain [11–13]. The non-steroidal anti-inflammatory drug (NSAID), naproxen [14], is active in the CFA model, reducing mechanical allodynia, weight bearing differentials and increasing the paw pressure withdrawal thresholds and is thus a clinically relevant positive control for this model.

The Connectivity Map was recently described by Lamb et al [15], as an approach to identify connections between diseases and drugs, based on their gene expression signatures. This resource consists of a large public database that includes hundreds of compound profiles termed the "reference gene signatures" obtained from cultured human cell lines treated with more than thirteen hundred pharmacologically active agents (Broad Build02 database) [15]. The database is attached to a pattern-matching tool. Gene signatures of interest from expression profiling studies can be compared to the Connectivity Map database, and subjected to a pattern matching algorithm that ranks similarities between the gene signature of interest and the reference signatures [15]. This can lead to identification of 'connections' between an investigator's gene expression signature (e.g. a disease tissue/model) and small molecule profiles in the database. Several recent studies have used this resource to identify novel agents for different indications, including hair growth and leukemia [16, 17].

Dorsal Root Ganglia (DRG) are known to be important sites for pain processing. In this study, we first generated gene expression profiles of the ipsilateral L4 and L5 dorsal root ganglia (DRG) extracted from rats injected with CFA in their left paws for 4 days and compared them to profiles of L4 and L5 DRGs from naïve rats. The gene signature of CFA-treated versus naïve rats was created and queried in the Connectivity Map database. Phenoxybenzamine - an alpha adrenergic receptor antagonist and several other agents showed significant inverse connectivity to the CFA signature. We thus reasoned that these agents could potentially reduce pain processing and have antinociceptive properties in the CFA model. Here we describe the data leading to the generation of this hypothesis and our subsequent experimental confirmation of the activity of phenoxybenzamine in this model of inflammatory pain.

Methods and materials

Animals

Adult Male Sprague Dawley rats (200-250 g) were used in all experiments. Rats were purchased from Harlan, (Indianapolis, IN) and maintained on a 12/12 hr light/dark cycle with food and water ad libitum. Rats were acclimated for a week before use in experiments. The Pfizer Institutional Animal Care and Use Committee reviewed and approved the animal use in these studies. The animal care and use program is fully accredited by the Association for Assessment and Accreditation of Laboratory Animal Care, International.

CFA-induced inflammation

Rats were anesthetized briefly with isoflurane (5% induction, then 2% maintenance) and their left foot swabbed with ethanol. 0.15 ml Complete Freund's adjuvant (CFA, SIGMA, St. Louis, MO) was injected subcutaneously into the plantar surface of the left hind paw of the rat [10]. The CFA injection immediately induces local inflammation, paw swelling and pain, which persists for at least 2 weeks post-injection. For behavior studies, rats were placed on the equipment and left to acclimate for 30 minutes. On day 0, baseline measurements were read and rats were injected with CFA thereafter. On day 3, post-CFA reads were taken and only rats that met criteria of hyperalgesia were placed on the study on day 4.

Mechanical allodynia and thermal hyperalgesia

To assess mechanical allodynia, rats were placed on an elevated wire mesh platform, and to confine their movement, a 15 × 22 × 25 cm plexiglass chamber was placed over each animal. Mechanical paw withdrawal thresholds (PWT) were measured by using a set of Semmes Weinstein monofilaments (Stoelting, Wood Dale, IL) using the Dixon up and down method [18]. Only rats that displayed a PWT of 8 g or less on day 3 (post-CFA) were placed on study. To assess thermal hyperalgesia rats were placed on glass plates with the source of heat applied from the bottom. On day 3 (post-CFA) rats that gave withdrawal latencies of 6 s or less were included in the experiments. Rats were then randomly assigned to either a vehicle group or drug group. On day 4, rats were treated with either the vehicle (saline), or drug and reads were taken 2 hrs after the treatment. All measurements were performed fully randomized and blinded and the reader did not know what treatment each rat received.

Drug administration

All drugs/vehicle were administered by oral gavage at 5 mg/kg/ml. Naproxen and phenoxybenzamine hydrochloride were purchased from SIGMA (St. Louis MO). Both were dissolved in saline, which was used as vehicle. All measurements were taken 2 hr post dosing. A group of naïve rats was also used in experiments, and these were not injected with CFA, and were not dosed.

Statistical analysis of behavioral data

This was done on the raw data using a one way ANOVA followed by the Student- Newman-Keul's Post-Hoc test.

RNA isolation and gene array profiling

A naive (control) group of rats (n = 6) and a group of rats injected with CFA (n = 6) as described above, were used for gene expression profiling experiments. On day 4, rats were euthanized and ipsilateral L4 and L5 dorsal root ganglia (DRG) were extracted, pooled for each rat, and immediately frozen in liquid nitrogen. DRG were stored at -80°C until used. RNA isolation and processing for gene arrays was done according to standard protocols by Genelogic (Boston, MA). Briefly, dorsal root ganglia tissues were homogenized in guanidium isothiocyanate with β-mercaptoethanol (GITC+BME), using the Omni TH-115 homogenizer. Trizol was added and mixed by repeated inversions. RNA aqueous phase was separated using Heavy Phase Lock Gel tubes. Chloroform and sample homogenate were added to the tube and shaken vigorously to remove protein contaminants, incubated on ice for 9-12 min, then centrifuged. The aqueous phase was cleaned using the Qiagen RNEasy Mini Kit (Cat # 74106). A bead cleanup was performed using Agencourt RNAClean magnetic beads (Cat # A29168) to further purify the RNA. RNA quality was evaluated by an Agilent Bioanalyzer. Only the RNA samples that passed the quality control measures proceeded for microarray. 12 high quality RNA samples each from an individual animal were prepared. 1 RNA sample from an animal in the control group had low yield and did not get processed further. The remaining 11 RNA samples were each run on a separate microarray. All 11 microarrays passed the quality control criteria and proceeded to downstream analysis. In total, 5 microarrays each from the 5 animals in the control group, and 6 microarrays each from the 6 animals in the CFA group were analyzed.

Rat whole genome 4X44K one color microarray processing

Sample preparation, hybridization, wash, scanning and feature extraction was performed according to the standard protocol, 'One-Color Microarray-Based Gene Expression Analysis v.5.7 (P/N G4140-90040)', by Agilent Technologies. One exception was that Triton was excluded from the wash buffer. Only the microarrays that passed the quality control criteria proceeded to downstream analysis.

Microarray data processing

The Agilent rat whole genome microarray signals were normalized with a program developed internally. Briefly, signal normalization against a common sample was performed using a smoothed piecewise linear curve, trained on the log intensities that differed by < 10% rank order between two samples. Local background subtraction was performed on each array prior to normalization. A computational naïve control pool was created from the averaged normalized signals of the individual naïve control arrays. Within the naïve control arrays, a median array was selected as the one least distant from the other naïve control arrays. All naïve control arrays were normalized against the median array prior to calculating the averaged normalized signals of the control pool. Finally each sample was normalized against the computational naïve control pool. Fold changes between samples were calculated, raising weak signals to a minimum value of 10 prior to comparison. Fold and p-values from student's t-test between groups was calculated.

Connectivity Map

The Broad's Connectivity Map algorithm was implemented in-house. All compound profiles were downloaded from Broad Build02 database [15]. A nonparametric, rank-based pattern matching strategy based on Kolmogorov-Smirnov statistics was devised and used in the calculation of the Connectivity Score and p-value as described in [15] and [19].

Gene Expression Pathway Analysis

Oligo probes that met the fold and p-value cutoff (arbitrarily set at 1.5 fold increase or decrease and p-value < = 0.05) were used for Functional Analysis using Ingenuity Pathway Analysis (Ingenuity IPA 8.0 (content version 2602); Ingenuity® Systems, http://www.ingenuity.com). Functional Analysis identified the biological functions and/or diseases that were significant to the dataset. Fischer's exact test was used to calculate a p-value determining the probability that each biological function and/or disease assigned to that dataset is due to chance alone. Canonical Pathway Analysis identified the pathways from the Ingenuity Pathways Analysis library of canonical pathways that were significant to the dataset. The significance of the association between the data set and the canonical pathway was measured in 2 ways: 1) A ratio of the number of genes from the dataset that map to the pathway divided by the total number of genes that map to the canonical pathway is displayed. 2) Fisher's exact test was used to calculate a p-value determining the probability that the association between the genes in the dataset and the canonical pathway is explained by chance alone.

Hierarchical Clustering

The transcript expression signal intensity was first mean centered and normalized across samples and then a complete linkage clustering method was applied on the correlation similarity measure for hierarchical clustering and heat map in Spotfire® DecisionSite.

Results

CFA-behavior studies

Injection of CFA significantly reduced mechanical paw withdrawal thresholds (PWT) by 62% when compared to naïve rats (P < 0.001, Figure 1A). Naproxen (10 mg/kg) significantly reversed mechanical allodynia back to about 73% (P < 0.01, Figure 1A) of naïve mechanical thresholds. Injection of CFA significantly induced thermal hyperalgesia as measured by a decrease of latencies to withdraw by about 76% when compared to naïve rats (P < 0.001, Figure 1B). Naproxen significantly reversed the thermal hyperalgesia back to about 86% of naïve controls (P < 0.001, Figure 1B). These effects are in close agreement with previously reported data [11, 12].

Figure 1
figure 1

Oral naproxen is effective in reducing nociceptive behavior in 4 day CFA-injected rats. Naproxen reduced A) mechanical thresholds and B) paw withdrawal latencies taken 2 hr post dose. n = 6-16 rats per group. For this and other figures, data is presented as mean +/- s.e.m. **p < 0.01 and ***p < 0.001, significant differences from vehicle (saline) - dosed rats.

Gene expression microarray studies

Gene expression in DRG was compared between arrays obtained from 4 day CFA-injected rats and from naïve rats. Genes that were up- or down- regulated by ≥ 1.5 fold (p ≤ 0.05), were used to generate a differential expression list for CFA-treated versus naive rat DRG. 235 oligo probes met these criteria. 178 oligo probes (representing 140 unique genes) have HUGO gene symbols and further analysis was focused on these genes. Hierarchical clustering indicated that these transcripts could separate the CFA samples from naïve samples, as shown in the heat map (Figure 2). We thus consider these transcripts as a molecular signature of the 4 day CFA model in the DRG tissue.

Figure 2
figure 2

Heat map of transcripts differentially expressed in the DRG between naïve and CFA injected rats. The branch length in the dendrogram represents the relation between the samples, the shorter the branch, the higher the similarity between the samples. Up-regulated genes are shown in yellow and down-regulated genes are in blue.

Overall, it appears that genes that mediate inflammation are suppressed, while genes that mediate neuron growth/survival are enhanced in this 4 day CFA gene signature. Ingenuity® Functional and Pathway Analysis (IPA) was conducted on the genes decreased by CFA treatment in the DRG to help understand the function and canonical pathways perturbed. IPA is a web-based application that enables the visualization, discovery and analysis of molecular functions and pathways within gene expression profiles. One function identified that was significantly affected (p-value < 0.01) is suppression of a group of genes that have a role in immune response or inflammatory disorders (Table 1). Also noted is that leucine rich repeat interacting protein 1 (Lrrfip1), a gene that represses TNF alpha expression, is increased in the DRG from CFA-treated rats compared to that from naïve rats. This data suggests that the immune and inflammatory response may play a role in regulating pain processing in the CFA treated DRG. The eicosanoid signaling pathway was also significantly affected in this model (p-value = 4E-3), with multiple pathway components down-regulated in the DRG tissue of the CFA-treated group 4 days after CFA treatment (Figure 3). These include the phospholipase A2 group IVA (PLA2g4a), prostaglandin-endoperoxide synthase 1 (Ptgs1), and rat arachidonate 12/15-lipoxygenase. In addition, several genes that might modulate the release of eicosanoids, such as BCL6 [20], BTK [21] and CNR2 [22], were also reduced 4 days post-CFA injection.

Table 1 Immune and inflammatory response genes down-regulated in the DRG by intraplantar CFA
Figure 3
figure 3

The eicosanoid signaling pathway from Ingenuity Pathway Analysis software. Only part of the pathway is shown. The highlighted genes (in yellow) were down-regulated in the 4 day CFA model of inflammatory pain. PLA2: phospholipase A2 group IVA; PTGS: prostaglandin-endoperoxide synthase 1; ALOX12, ALOX15: rat arachidonate 12/15-lipoxygenase.

Another group of genes suppressed by CFA encode proteins that may contribute to T cell function. Among these genes, IL21R [23], IL2RB [24], RHOH [25], CNR2 [26], IKZF1 [27], BCL6 [28], PTGS1 [29], CASP8 [30], and CD52 [31] were all down-regulated. In addition, several genes related to T-cell function were increased (Table 2) in the CFA treated rat DRGs. Examples include IL22RA2 [32]; CD7 [33]; Lgals2 (also called galectin-2) [34], and IL2RG [35].

Table 2 Genes up-regulated in the DRG by intraplantar CFA

Genes that were up-regulated in the DRG of CFA treated rats, include some that have been previously shown to play a role in pain and neuronal growth, differentiation and prevention of neuronal death. These include neuromedin U, heptocyte growth factor, steroid-5-alpha-reductase (SRD5A2), SLC30A3, PC4, SFRS1 interacting protein 1 (PSIP1), PER2, ZHX2 and ENC1. The functions of these are discussed in the discussion section.

Connectivity Map Results

Using the list of differentially expressed genes between the CFA-treated and naive rats as a DRG CFA signature, we evaluated an internal version of the Broad's Connectivity Map to identify compounds that display inverse connectivity with this CFA signature. This approach yielded five compounds that met the criteria with a p-val < 0.005 and a connectivity score < -0.5 (Figure 4). Based on their negative connectivity with the CFA signature, we hypothesized that these compounds could potentially reduce pain in the CFA model. To test this further, we selected one of the most strongly inferred compounds, phenoxybenzamine, for follow up experiments in the 4 day CFA model to examine its antinociceptive potential.

Figure 4
figure 4

Compounds identified following connectivity mapping of the CFA signature.

Phenoxybenzamine in the 4 day CFA assay

Phenoxybenzamine was evaluated in the 4 day CFA model using mechanical allodynia as an outcome measurement and with naproxen as a positive control. When compared to the vehicle treated rats, naproxen (10 mg/kg) reversed mechanical allodynia by 93% (P < 0.01, Figure 5). Phenoxybenzamine, at 10 mg/kg was almost as efficacious as naproxen and reversed mechanical allodynia by 82% (P < 0.05, Figure 5).

Figure 5
figure 5

Effect of oral phenoxybenzamine administered to rats on day 4 of the CFA model of inflammatory pain. Mechanical thresholds were read 2 hours post-dosing. n = 8-15 per group. *p < 0.05 and **p < 0.01, significant differences from vehicle (saline) - dosed rats.

Discussion

In this study, we first conducted gene expression microarray experiments to examine transcriptional profiles in the ipsilateral dorsal root ganglia (DRG) of adult rats in a 4 day CFA model of inflammatory pain and generated a rat CFA model gene signature. Our second aim was to identify compounds that were negatively connected to this CFA gene signature using the Connectivity Map approach. Our third objective was to test the hypothesis that compounds identified in this way could reduce CFA-induced pain in this 4 day model. Reports on transcriptional profiling in inflammatory pain models are scarce. Yang et. al. examined gene expression changes in the inflamed tissues and the corresponding DRGs in a carrageenan model using a mini-array containing 100 cytokines, chemokines and related receptors [8]. Parkitna et. al. conducted transcriptional profiling of 4000 genes in the lumbar section of rat spinal cord of a 3-day and a 14-day CFA model [4]. The authors found a dramatic shift in the regulation of secretory vesicle trafficking in the spinal cord in the CFA model. The authors did not profile the L4-L5 DRG. However, they selected 4 transcripts that were changed in the spinal cord and tested them in the L4-5 DRG with qPCR. Consistent with our results, none of the 4 transcripts were significantly changed in the CFA DRG comparing to the naïve rats. To the best of our knowledge, there are no published DRG whole genome microarray studies following 4 days CFA inflammation in rats. We used the CFA model since commonly used NSAIDs such as naproxen that are used to treat OA pain are effective in this model and it is thus frequently used pre-clinically to identify new therapies for OA pain [11, 12].

By clustering differentially expressed genes by function or related pathways, we identified a number of genes that are implicated in the modulation of the immune system. Surprisingly, we found that the expression of these genes is decreased in the CFA-treated rats, suggesting suppression of the immune system at day 4 post-CFA treatment in this model system. These include genes in the eicosanoid signaling pathway, including phospholipase A2 group IVA (PLA2g4a), prostaglandin-endoperoxide synthase 1 (Ptgs1), and rat arachidonate 12/15-lipoxygenase. In addition, several genes such as BCL6, BTK and CNR2 that may modulate release of eicosanoids [20–22] are also reduced in the ipsilateral DRGs four days after CFA treatment. We speculate that these genes are early players that initiate inflammation and pain, and their signals are carried forth by other pathways that maintain chronic pain/inflammation with time.

Another group of genes down-regulated by CFA treatment encode proteins that regulate T cell function. These include IL21 receptor subtype II, IL2 receptor beta, RHOH (Ras homolog gene family member H), cannabinoid receptor 2, IKZF1, CASP8, and CD52 [23–31]. It is also noteworthy that IL22RA2, a soluble receptor for IL22 that antagonizes IL22 which is primarily produced by activated T cells [32], CD7 which promotes T cell apoptosis [33], Lgals2 (galectin-2), a proapoptotic effector of activated T-cells [34], and IL2 receptor gamma [35] are all increased in the CFA group compared to the naïve group. This may represent a mechanism of communication by the DRG that modulates T cell activity during inflammation or pain processing. In this regard, T cells have been recently suggested to play a role in neuropathic pain [36, 37]. In a model where DRG inflammation was induced by administration of epidural zymosan in incomplete Freund's Adjuvant [7], an increase of CCL2 (MCP-1) among other cytokines was observed, while IL-2 and IL-12 were decreased on day 3 post inflammation. Similarly, in a peripheral inflammation model induced by hind paw injection of carrageenan, both the Scya2 (the CCL2 mRNA) and the gene product MCP-1 (CCL2) were up-regulated following nociceptive stimuli [8]. In that study, Scya2 mRNA was increased about 2-fold in the ipsilateral versus contralateral rat DRG and lasted up to 72 hour post carrageenan. Scya2 mRNA was localized to a subpopulation of vanilloid receptor 1 (TRPV1) containing neurons. Stimulation by a TRPV1 agonist, resiniferatoxin increased expression of Scya2 mRNA. These results are very similar to our findings where we found increased expression of the CCL2 gene in the DRG following CFA injection. CCL2 has also been reported to be up-regulated in the DRG in models of neuropathic pain ([38] and [39] for review). Thus, CCL2 seems to be a common signal elicited in different preclinical pain models.

Other genes that are induced in the DRG of CFA treated rats, include some that encode proteins either known to play a role in pain, promote neuronal growth/differentiation, or prevent neuronal death. One that stands out is neuromedin U (NMU) which has been shown to have an emerging physiological role in nociception upon binding to the NMU receptor 2 [40]. Mice deficient in NMUR2 displayed reduced thermal nociceptive responses in the hot plate test, decreased thermal hyperalgesia following capsaicin injection and reduced the late phase response in the formalin test [41]. In other studies, NMU inhibited inflammation-mediated memory impairment and neuronal cell-death in rodents [42]. Ketterer et. al. (2009) have shown recently that NMU may signal via the hepatocyte growth factor (HGF) c-Met pathway. In our study, HGF is also increased in the DRG following CFA treatment. HGF is a pleiotropic cytokine which partly functions to promote neuronal survival and growth [43]. HGF cooperates with nerve growth factor (NGF) to enhance axonal outgrowth from cultured dorsal root ganglion (DRG) neurons. HGF also enhances the neurotrophic activities of NGF in vivo where Met receptor signaling is required for the survival of a proportion of DRG neurons [44].

Another gene which is up-regulated in the DRG is the steroid-5-alpha-reductase (SRD5A2) which is a key enzyme in the conversion of several Δ4-3keto steroids, such as testosterone, progesterone, aldosterone and corticosterone, into their respective reductase derivatives. Morphine has been shown to increase SRD5A2 level and SRD5A2 inhibitor finasteride potentiates the antinociceptive effect of morphine, prevents the development of morphine tolerance in rats, suggesting that SRD5A2 plays a role in pain perception [45]. SLC30A3 which is also increased in the current study is responsible for transport of zinc into synaptic vesicles. It may have a role in neuropathic pain [46].

We also observed a number of genes whose expression in DRG is modulated by CFA, though their roles in nociception have not been established, they may have indirect linkages to pain plasticity. Two genes involved in regulating circadian rhythm are modulated in the DRG of CFA treated rats versus the naïve rats. Pain perception is influenced by the circadian rhythm in humans and in animals [47]. Expression of PER2 is increased, whereas expression of NPAS2 is decreased. Per/Cry form a heterodimer that interacts with the NPAS2/BMAL1 heterodimer to inhibit the transcription of Per and Cry [48]. SFRS1 interacting protein 1 (PSIP1, or Lens epithelium-derived growth factor (LEDGF)) is up-regulated in response to stress and enhances the survival of neurons in the retina and optic nerve [49]. We speculate that the increased expression of PSIP1 we observed may support neuronal growth in the DRG. ZHX2, a transcriptional regulator of neural progenitor cells, is also up-regulated in the DRG of CFA treated rats. Blocking ZHX2 function causes neuronal differentiation, whereas overexpression of ZHX2 or its ephrin-B1 intracellular domain disrupts the normal differentiation of cortical neural progenitor cells [50]. ENC1 (or NRP/B), also a regulator of neuronal differentiation is increased in the DRG of CFA treated rats. Overexpression of NRP/B significantly induced neurite outgrowth in PC12 cells, whereas inhibition of NRP/B by antibodies or siRNA inhibited neurite outgrowth and suppressed the NGF-induced outgrowth of neurites [50, 51]. In summary, we have found a number of genes which regulate neuronal growth in the CFA model, suggesting a neuronal growth component to this peripheral inflammation model.

Using the Connectivity Map approach, we identified 5 compounds in the Broad database that were negatively connected with the CFA signature. These compounds belong to different chemical classes and their structural properties are different. They also bind to different targets. Fenoprofen, an NSAID, was among them. Another compound we identified with this approach was pyrvinium, a non-competitive androgen receptor inhibitor [52]. Intrathecal administration of testosterone, an androgen derived from DHEA, has been shown to cause analgesia in neuropathic rats [53]. Difenidol, also identified by Connectivity Mapping is used for treatment of vertigo. The precise molecular target of this drug is not known, although recently it was found to be a ligand for muscarinic receptors: M1, M3 and M4 [54], and may thus play a role in processing of pain stimuli [55].

Phenoxybenzamine, a non-selective α-blocker [56] was also identified as having negative connectivity with the CFA pain signature. Previous studies have reported efficacy of phenoxybenzamine in patients with various pain conditions. In one study, 40 patients with causalgia were treated successfully for their pain with phenoxybenzamine [57]. In a case report, 3 out of 4 patients got relief from complex regional pain syndrome type I with phenoxybenzamine [58]. In the current study, we examined effects of phenoxybenzamine in the 4 Day CFA model to determine whether the Connectivity Map approach could identify compounds that had analgesic potential in this model. Our data confirm the antinociceptive properties of phenoxybenzamine; this effect may be due to a direct blockade of adrenergic receptors that may have become supersensitive to catecholamines or perhaps due to possible sympathetic sprouting, as a result of increased expression of nerve growth regulating genes. Sympathetic sprouting in the DRG has been shown to occur in a study that examined gene expression profiles at 3 days post zymosan in incomplete Freund's adjuvant [7]. Furthermore, antinociception has been reported for phenoxybenzamine in the rat hot plate test [59], the mouse tail flick test [60] and mouse writhing tests [61]. Although we do not yet understand the basis for the diminished efficacy of phenoxybenzamine at the higher dose tested here (20 mg/kg), it is possibly due to excessive antagonism of the adrenergic receptors in addition to other mechanisms, such as its agonist-antagonistic properties [60]. Regardless, our data suggest that the CFA pain model may have broader utility for testing compounds that modulate pain involving the sympathetic nervous system.

Conclusion

We have established a 4 day CFA signature in the DRG that we subjected to a Connectivity Map approach and identified a compound already known to have a role in the treatment of pain, such as fenoprofen. Another of these compounds, phenoxybenzamine, has been reported in the literature to treat complex regional pain syndrome, and our studies confirm the analgesic properties of this compound in the 4 day CFA model. We conclude that use of the Connectivity Map approach to accompany gene expression microarray and behavior studies, represents a potentially fruitful way to identify novel pain therapies.

References

  1. Vega-Avelaira D, Geranton S, Fitzgerald M: Differential regulation of immune responses and macrophage/neuron interactions in the dorsal root ganglion in young and adult rats following nerve injury. Molecular Pain 2009, 5: 70. 10.1186/1744-8069-5-70

    Article  PubMed Central  PubMed  Google Scholar 

  2. Takeuchi H, Kawaguchi S, Mizuno S, Kirita T, Takebayashi T, Shimozawa K, Torigoe T, Sato N, Yamashita T: Gene Expression Profile of Dorsal Root Ganglion in a Lumbar Radiculopathy Model. Spine 2008, 33: 2483–2488. 10.1097/BRS.0b013e318184acc3

    Article  PubMed  Google Scholar 

  3. Costigan M, Befort K, Karchewski L, Griffin R, D'Urso D, Allchorne A, Sitarski J, Mannion J, Pratt R, Woolf C: Replicate high-density rat genome oligonucleotide microarrays reveal hundreds of regulated genes in the dorsal root ganglion after peripheral nerve injury. BMC Neuroscience 2002, 3: 16. 10.1186/1471-2202-3-16

    Article  PubMed Central  PubMed  Google Scholar 

  4. Rodriguez Parkitna JKM, Kaminska-Chowaniec D, Obara I, Mika J, Przewlocka B, Przewlocki R: Comparison of gene expression profiles in neuropathic and inflammatory pain. J Physiol Pharmacol 2006, 57: 14.

    Google Scholar 

  5. Rabert D, Xiao Y, Yiangou Y, Kreder D, Sangameswaran L, Segal MR, Hunt CA, Birch R, Anand P: Plasticity of gene expression in injured human dorsal root ganglia revealed by GeneChip oligonucleotide microarrays. Journal of Clinical Neuroscience 2004, 11: 289–299. 10.1016/j.jocn.2003.05.008

    Article  CAS  PubMed  Google Scholar 

  6. Li X, Sahbaie P, Zheng M, Ritchie J, Peltz G, Mogil J, Clark JD: Expression genetics identifies spinal mechanisms supporting formalin late phase behaviors. Molecular Pain 2010, 6: 11. 10.1186/1744-8069-6-11

    Article  PubMed Central  PubMed  Google Scholar 

  7. Xie WR, Deng H, Li H, Bowen TL, Strong JA, Zhang JM: Robust increase of cutaneous sensitivity, cytokine production and sympathetic sprouting in rats with localized inflammatory irritation of the spinal ganglia. Neuroscience 2006, 142: 809–822. 10.1016/j.neuroscience.2006.06.045

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Yang HY, Mitchell K, Keller JM, Iadarola MJ: Peripheral inflammation increases Scya2 expression in sensory ganglia and cytokine and endothelial related gene expression in inflamed tissue. J Neurochem 2007, 103: 1628–1643. 10.1111/j.1471-4159.2007.04874.x

    Article  CAS  PubMed  Google Scholar 

  9. Hammer P, Banck MS, Amberg R, Wang C, Petznick G, Luo S, Khrebtukova I, Schroth GP, Beyerlein P, Beutler AS: mRNA-seq with agnostic splice site discovery for nervous system transcriptomics tested in chronic pain. Genome Res 2010, 20: 847–860. 10.1101/gr.101204.109

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  10. Stein CMM, Herz A: Unilateral inflammation of the hindpaw in rats as a model of prolonged noxious stimulation: alterations in behavior and nociceptive thresholds. Pharmacol Biochem Behav 1988, 31: 155–151. 10.1016/0091-3057(88)90372-3

    Article  Google Scholar 

  11. Ahn K, Johnson DS, Mileni M, Beidler D, Long JZ, McKinney MK, Weerapana E, Sadagopan N, Liimatta M, Smith SE, Lazerwith S, Stiff C, Kamtekar S, Bhattacharya K, Zhang Y, Swaney S, Van Becelaere K, Stevens RC, Cravatt BF: Discovery and characterization of a highly selective FAAH inhibitor that reduces inflammatory pain. Chemistry & Biology 2009, 16: 411–420.

    Article  CAS  Google Scholar 

  12. Johnson DS, Ahn K, Kesten S, Lazerwith SE, Song Y, Morris M, Fay L, Gregory T, Stiff C, Dunbar JB Jr, Liimatta M, Beidler D, Smith S, Nomanbhoy TK, Cravatt BF: Benzothiophene piperazine and piperidine urea inhibitors of fatty acid amide hydrolase (FAAH). Bioorganic & Medicinal Chemistry Letters 2009, 19: 2865–2869.

    Article  CAS  Google Scholar 

  13. Cortes-Burgos LA, Zweifel BS, Settle SL, Pufahl RA, Anderson GD, Hardy MM, Weir DE, Hu G, Happa FA, Stewart Z, Muthian S, Graneto MJ, Masferrer JL: CJ-13610, an orally active inhibitor of 5-lipoxygenase is efficacious in preclinical models of pain. European Journal of Pharmacology 2009, 617: 59–67. 10.1016/j.ejphar.2009.06.058

    Article  CAS  PubMed  Google Scholar 

  14. Fendrick AM, Greenberg B: A review of the benefits and risks of nonsteroidal anti-inflammatory drugs in the management of mild-to-moderate osteoarthritis. Osteopathic Medicine and Primary Care 2009, 3: 1. 10.1186/1750-4732-3-1

    Article  PubMed Central  PubMed  Google Scholar 

  15. Lamb J, Crawford ED, Peck D, Modell JW, Blat IC, Wrobel MJ, Lerner J, Brunet J-P, Subramanian A, Ross KN, Reich M, Hieronymus H, Wei G, Armstrong SA, Haggarty SJ, Clemons PA, Wei R, Carr SA, Lander ES, Golub TR: The Connectivity Map: Using gene-expression signatures to connect small molecules, genes, and disease. Science 2006, 313: 1929–1935. 10.1126/science.1132939

    Article  CAS  PubMed  Google Scholar 

  16. Ishimatsu-Tsuji Y, Soma T, Kishimoto J: Identification of novel hair-growth inducers by means of connectivity mapping. FASEB J 2009. fj.09–145292

    Google Scholar 

  17. Hassane DC, Guzman ML, Corbett C, Li X, Abboud R, Young F, Liesveld JL, Carroll M, Jordan CT: Discovery of agents that eradicate leukemia stem cells using an in silico screen of public gene expression data. Blood 2008, 111: 5654–5662. 10.1182/blood-2007-11-126003

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Dixon WJ: Efficient analysis of experimental observations. Annual Review of Pharmacology and Toxicology 1980, 20: 441–462. 10.1146/annurev.pa.20.040180.002301

    Article  CAS  PubMed  Google Scholar 

  19. Lamb J, Ramaswamy S, Ford HL, Contreras B, Martinez RV, Kittrell FS, Zahnow CA, Patterson N, Golub TR, Ewen ME: A mechanism of cyclin D1 action encoded in the patterns of gene expression in human cancer. Cell 2003, 114: 323–334. 10.1016/S0092-8674(03)00570-1

    Article  CAS  PubMed  Google Scholar 

  20. Ravaux L, Denoyelle C, Monne C, Limon I, Raymondjean M, El Hadri K: Inhibition of interleukin-1{beta}-induced group IIA secretory phospholipase A2 expression by peroxisome proliferator-activated receptors (PPARs) in rat vascular smooth muscle cells: cooperation between PPAR{beta} and the proto-oncogene BCL-6. Mol Cell Biol 2007, 27: 8374–8387. 10.1128/MCB.00623-07

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Kawakami Y, Kitaura J, Satterthwaite AB, Kato RM, Asai K, Hartman SE, Maeda-Yamamoto M, Lowell CA, Rawlings DJ, Witte ON, Kawakami T: Redundant and opposing functions of two tyrosine kinases, Btk and Lyn, in mast cell activation. J Immunol 2000, 165: 1210–1219.

    Article  CAS  PubMed  Google Scholar 

  22. Maccarrone M, Battista N, Centonze D: The endocannabinoid pathway in Huntington's disease: A comparison with other neurodegenerative diseases. Progress in Neurobiology 2007, 81: 349–379. 10.1016/j.pneurobio.2006.11.006

    Article  CAS  PubMed  Google Scholar 

  23. Monteleone G, Pallone F, Macdonald TT: Interleukin-21 (IL-21)-mediated pathways in T cell-mediated disease. Cytokine & Growth Factor Reviews 2009, 20: 185–191.

    Article  CAS  Google Scholar 

  24. Liu DV, Maier LM, Hafler DA, Wittrup KD: Engineered interleukin-2 antagonists for the inhibition of regulatory T cells. Journal of Immunotherapy 2009, 32: 887–894. 10.1097/CJI.0b013e3181b528da

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Suzuki HOH: The atypical small GTPase RhoH: a novel role in T cell development. Nihon Rinsho Meneki Gakkai Kaishi 2008, 31: 37–46.

    Article  CAS  PubMed  Google Scholar 

  26. Maresz K, Pryce G, Ponomarev ED, Marsicano G, Croxford JL, Shriver LP, Ledent C, Cheng X, Carrier EJ, Mann MK, Giovannoni G, Pertwee RG, Yamamura T, Buckley NE, Hillard CJ, Lutz B, Baker D, Dittel BN: Direct suppression of CNS autoimmune inflammation via the cannabinoid receptor CB1 on neurons and CB2 on autoreactive T cells. Nat Med 2007, 13: 492–497. 10.1038/nm1561

    Article  CAS  PubMed  Google Scholar 

  27. Ronni T, Payne KJ, Ho S, Bradley MN, Dorsam G, Dovat S: Human ikaros function in activated T cells is regulated by coordinated expression of its largest isoforms. Journal of Biological Chemistry 2007, 282: 2538–2547. 10.1074/jbc.M605627200

    Article  CAS  PubMed  Google Scholar 

  28. Crotty S, Johnston RJ, Schoenberger SP: Effectors and memories: Bcl-6 and Blimp-1 in T and B lymphocyte differentiation. Nat Immunol 11: 114–120. 10.1038/ni.1837

  29. Rocca B, Spain LM, Puré E, Langenbach R, Patrono C, FitzGerald GA: Distinct roles of prostaglandin H synthases 1 and 2 in T-cell development. The Journal of Clinical Investigation 1999, 103: 1469–1477. 10.1172/JCI6400

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Algeciras-Schimnich A, Vlahakis SR, Villasis-Keever A, Gomez T, Heppelmann CJ, Bou G, Paya CV: CCR5 mediates Fas- and caspase-8 dependent apoptosis of both uninfected and HIV infected primary human CD4 T cells. AIDS 2002, 16: 1467–1478. 10.1097/00002030-200207260-00003

    Article  CAS  PubMed  Google Scholar 

  31. Watanabe T, Masuyama J-i, Sohma Y, Inazawa H, Horie K, Kojima K, Uemura Y, Aoki Y, Kaga S, Minota S, Tanaka T, Yamaguchi Y, Kobayashi T, Serizawa I: CD52 is a novel costimulatory molecule for induction of CD4+ regulatory T cells. Clinical Immunology 2006, 120: 247–259. 10.1016/j.clim.2006.05.006

    Article  CAS  PubMed  Google Scholar 

  32. Wolk KWE, Witte K, Warszawska K, Sabat R: Biology of interleukin-22. Semin Immunopathol 2010, 32: 17–31. 10.1007/s00281-009-0188-x

    Article  CAS  PubMed  Google Scholar 

  33. Fukumori T, Takenaka Y, Yoshii T, Kim H-RC, Hogan V, Inohara H, Kagawa S, Raz A: CD29 and CD7 mediate galectin-3-induced type II T-cell apoptosis. Cancer Res 2003, 63: 8302–8311.

    CAS  PubMed  Google Scholar 

  34. Sturm A, Lensch M, Andre S, Kaltner H, Wiedenmann B, Rosewicz S, Dignass AU, Gabius H-J: Human galectin-2: novel inducer of T cell apoptosis with distinct profile of caspase activation. J Immunol 2004, 173: 3825–3837.

    Article  CAS  PubMed  Google Scholar 

  35. Tsujino S, Di Santo JP, Takaoka A, McKernan TL, Noguchi S, Taya C, Yonekawa H, Saito T, Taniguchi T, Fujii H: Differential requirement of the cytoplasmic subregions of γc chain in T cell development and function. Proceedings of the National Academy of Sciences of the United States of America 2000, 97: 10514–10519. 10.1073/pnas.180063297

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  36. Thacker MA, Clark AK, Marchand F, McMahon SB: Pathophysiology of peripheral neuropathic pain: immune cells and molecules. Anesth Analg 2007, 105: 838–847. 10.1213/01.ane.0000275190.42912.37

    Article  PubMed  Google Scholar 

  37. Costigan M, Moss A, Latremoliere A, Johnston C, Verma-Gandhu M, Herbert TA, Barrett L, Brenner GJ, Vardeh D, Woolf CJ, Fitzgerald M: T-Cell infiltration and signaling in the adult dorsal spinal cord is a major contributor to neuropathic pain-like hypersensitivity. J Neurosci 2009, 29: 14415–14422. 10.1523/JNEUROSCI.4569-09.2009

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  38. Gao Y-J, Ji R-R: Chemokines, neuronal-glial interactions, and central processing of neuropathic pain. Pharmacology & Therapeutics 2010, 126: 56–68.

    Article  CAS  Google Scholar 

  39. White FA, Feldman P, Miller RJ: Chemokine signaling and the management of neuropathic pain. Molecular Interventions 2009, 9: 188–195. 10.1124/mi.9.4.7

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Mitchell JD, Maguire JJ, Davenport AP: Emerging pharmacology and physiology of neuromedin U and the structurally related peptide neuromedin S. British Journal of Pharmacology 2009, 158: 87–103. 10.1111/j.1476-5381.2009.00252.x

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  41. Torresa Richard, Crolla SusanD, Vercollonea Jeffrey, Reinhardta Joel, Griffithsa Jennifer, Zabskia Stephanie, Andersona KeithD, Adamsa NielsC, Gowena Lori, Sleemana MarkW, Valenzuelaa DavidM, Wieganda StanleyJ, Yancopoulosa GeorgeD, Murphya AndrewJ: Mice genetically deficient in neuromedin U receptor 2, but not neuromedin U receptor 1, have impaired nociceptive responses. Pain 2007, 130: 267–278. 10.1016/j.pain.2007.01.036

    Article  Google Scholar 

  42. Takashi Iwaia YI, Kodania Reiye, Oka Jun-Ichiro: Neuromedin U inhibits inflammation-mediated memory impairment and neuronal cell-death in rodents. Neuroscience Research 2008, 61: 113–119. 10.1016/j.neures.2008.01.018

    Article  Google Scholar 

  43. Maina Flavio, Hilton MarkC, Andres Rosa, Wyatt Sean, Klein Rüdiger, Davies AlunM: Multiple roles for hepatocyte growth factor in sympathetic neuron development. Neuron 1998, 20: 835–846. 10.1016/S0896-6273(00)80466-3

    Article  CAS  PubMed  Google Scholar 

  44. Maina FHM, Ponzetto C, Davies AM, Klein R: Met receptor signaling is required for sensory nerve development and HGF promotes axonal growth and survival of sensory neurons. Genes Dev 1997, 11: 3341–3350. 10.1101/gad.11.24.3341

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  45. Verdi J, Ahmadiani A: Finasteride, a 5[alpha]-reductase inhibitor, potentiates antinociceptive effects of morphine, prevents the development of morphine tolerance and attenuates abstinence behavior in the rat. Hormones and Behavior 2007, 51: 605–610. 10.1016/j.yhbeh.2007.02.008

    Article  CAS  PubMed  Google Scholar 

  46. Jo SM DG, Schrøder HD, Suh SW: Depletion of vesicular zinc in dorsal horn of spinal cord causes increased neuropathic pain in mice. Biometals 2008: 151–158.

  47. Bruguerolle B, Labrecque G: Rhythmic pattern in pain and their chronotherapy. Advanced Drug Delivery Reviews 2007, 59: 883–895. 10.1016/j.addr.2006.06.001

    Article  CAS  PubMed  Google Scholar 

  48. Kitanishi K, Igarashi J, Hayasaka K, Hikage N, Saiful I, Yamauchi S, Uchida T, Ishimori K, Shimizu T: Heme-binding characteristics of the isolated PAS-A domain of mouse Per2, a transcriptional regulatory factor associated with circadian rhythms. Biochemistry 2008, 47: 6157–6168. 10.1021/bi7023892

    Article  CAS  PubMed  Google Scholar 

  49. Raz-Prag D, Zeng Y, Sieving PA, Bush RA: Photoreceptor protection by adeno-associated virus-mediated LEDGF expression in the RCS rat model of retinal degeneration: probing the mechanism. Invest Ophthalmol Vis Sci 2009, 50: 3897–3906. 10.1167/iovs.08-3153

    Article  PubMed Central  PubMed  Google Scholar 

  50. Wu C, Qiu R, Wang J, Zhang H, Murai K, Lu Q: ZHX2 interacts with ephrin-B and regulates neural progenitor maintenance in the developing cerebral cortex. J Neurosci 2009, 29: 7404–7412. 10.1523/JNEUROSCI.5841-08.2009

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. Kim T-A, Jiang S, Seng S, Cha K, Avraham HK, Avraham S: The BTB domain of the nuclear matrix protein NRP/B is required for neurite outgrowth. J Cell Sci 2005, 118: 5537–5548. 10.1242/jcs.02643

    Article  CAS  PubMed  Google Scholar 

  52. Jones JeremyO, B EC, Huang Yong, Feau Clementine, Kiplin Guy R, Yamamoto KeithR, Byron Hann, Marc DiamondabdI: Non-competitive androgen receptor inhibition in vitro and in vivo. Proc Natl Acad Sci USA 2009, 106: 7233–7238. 10.1073/pnas.0807282106

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  53. Kibaly C, Meyer L, Patte-Mensah C, Mensah-Nyagan AG: Biochemical and functional evidence for the control of pain mechanisms by dehydroepiandrosterone endogenously synthesized in the spinal cord. FASEB J 2008, 22: 93–104. 10.1096/fj.07-8930com

    Article  CAS  PubMed  Google Scholar 

  54. Varoli L, Andreani A, Burnelli S, Granaiola M, Leoni A, Locatelli A, Morigi R, Rambaldi M, Bedini A, Fazio N, Spampinato S: Diphenidol-related diamines as novel muscarinic M4 receptor antagonists. Bioorganic & Medicinal Chemistry Letters 2008, 18: 2972–2976.

    Article  CAS  Google Scholar 

  55. AM T: Muscarinic acetylcholine receptors: new potential therapeutic targets in antinociception and in cancer therapy. Recent Pat CNS Drug Discov 2008, 3: 94–103. 10.2174/157488908784534621

    Article  Google Scholar 

  56. Frang H, Cockcroft V, Karskela T, Scheinin M, Marjamäki A: Phenoxybenzamine binding reveals the helical orientation of the third transmembrane domain of adrenergic receptors. Journal of Biological Chemistry 2001, 276: 31279–31284. 10.1074/jbc.M104167200

    Article  CAS  PubMed  Google Scholar 

  57. Ghostine SY, Comair YG, Turner DM, Kassell NF, Azar CG: Phenoxybenzamine in the treatment of causalgia. Journal of Neurosurgery 1984, 60: 1263–1268. 10.3171/jns.1984.60.6.1263

    Article  CAS  PubMed  Google Scholar 

  58. Mario AI Jr, Grigory K: Treatment of Complex Regional Pain Syndrome Type I with oral phenoxybenzamine: rationale and case reports. Pain Practice 2008, 8: 125–132. 10.1111/j.1533-2500.2007.00170.x

    Article  Google Scholar 

  59. Cicero TJ, Meyer ER, Smithloff BR: Alpha adrenergic blocking agents: antinociceptive activity and enhancement of morphine-induced analgesia. Journal of Pharmacology and Experimental Therapeutics 1974, 189: 72–82.

    CAS  PubMed  Google Scholar 

  60. Spiehler VRRL: Agonist--antagonist properties of phenoxybenzamine in antinociception and opiate dependence tests. Eur J Pharmacol 1979, 55: 389–395. 10.1016/0014-2999(79)90113-4

    Article  CAS  PubMed  Google Scholar 

  61. Hendershot LC, Forsaith J: Antagonism of the frequency of phenylquinone-induced writhing in the mouse by weak analgesic and nonalgesics. Journal of Pharmacology and Experimental Therapeutics 1959, 125: 237–240.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Pfizer colleagues from the Computational Sciences Center of Emphasis (particularly Dmitri Bichko and Simon Xi), and we thank Eric Welsch and Richard Head for building some of the tools used in this manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Farzana Karim.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

MC conceived of the study, analyzed the gene expression microarray data, generated data from connectivity mapping and drafted the manuscript. SS performed part of the behavior studies. AT participated in design of the study. MB participated in design of the study and editing the manuscript. FK conceived of the study, designed and coordinated the study, performed part of the behavior studies, and drafted the manuscript. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Chang, M., Smith, S., Thorpe, A. et al. Evaluation of phenoxybenzamine in the CFA model of pain following gene expression studies and connectivity mapping. Mol Pain 6, 56 (2010). https://doi.org/10.1186/1744-8069-6-56

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1744-8069-6-56

Keywords