Mast cells mediate substance P-induced bladder inflammation through an NK1 receptor-independent mechanism

Ricardo Saban1, Norma P. Gerard2,3, Marcia R. Saban1, Ngoc-Bich Nguyen1, Douglas J. DeBoer4, and Barry K. Wershil5

1 Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73190; 2 Pulmonary Division, Beth Israel Deaconess Medical Center, and 3 Ina Sue Pelmutter Laboratory, Children's Hospital, Harvard Medical School, Boston, Massachusetts 02215; 4 Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin 53706; and 5 Department of Pediatrics, State University of New York, Health Science Center at Brooklyn, Brooklyn, New York 11203-2098


    ABSTRACT
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

The role of neurokinin-1 receptors (NK1R) in the interaction between mast cells and substance P (SP) in bladder inflammation was determined. Mast cell-deficient KitW/KitW-v, congenic normal (+/+), and KitW/KitW-v mice that were reconstituted with bone marrow cells isolated from NK1R-/- mice were challenged by instillation of SP, antigen, or saline into the urinary bladder. Twenty-four hours after challenge, the bladders were prepared for morphological assessment and gene expression. SP-induced bladder inflammation was mast cell dependent and did not require NK1R expression on the mast cell. Cluster analysis identified functionally significant genes that were dependent on the presence of mast cells for their upregulation regardless of stimulus. Those include serine protein inhibitor 2.2, maspin, mitogen- and stress-activated protein kinase 2, and macrophage colony-stimulating factor 1. Our findings demonstrate that while mast cells are essential for both antigen- and SP-induced bladder inflammation, there are common genes and unique genes expressed in each type of inflammatory reaction. When combined with unique animal models, gene array analysis provides a useful approach for identifying and characterizing pathways involved in bladder inflammation.

transgenic/knockout; mast cells; inflammation; gene regulation; protein kinases/phosphatases


    INTRODUCTION
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

INFLAMMATION UNDERLIES ALL major bladder pathologies. Indeed, areas of bladder inflammation are found during chronic implantation of catheters (22, 88), as an integrative part of bladder responses to intravesical bacillus Calmette-Guerin (BCG) (10, 12) and capsaicin therapy (34), during radiation cystitis, and in cyclophosphamide-induced cystitis (86, 87), interstitial cystitis (28, 68), and bladder cancer (3-13).

Chronic inflammation in the bladder mucosa is typical in spinal cord-injured patients with a chronic indwelling bladder catheter (22, 88). Inflammation is also observed secondarily to intravesical treatments. Indeed, inflammation is part of the response to intravesical BCG instillation. However, some authors have reported that BCG inflammation obviously differs from nonspecific inflammation by its quality and subclinical duration (12), whereas others have qualified BCG-induced bladder inflammation as granulomatous (10). In addition, inflammation does not explain BCG immunotherapy for superficial bladder carcinoma; this complex response requires an immune response as well as a tumor response for its effects (43). Inflammation occurs also secondarily to intravesical capsaicin therapy for detrusor hyperreflexia (34), and part of this response is modulated by ethanol, used as a vehicle for capsaicin (23).

Inflammation seems to underlie bladder cancer. In this context, epidemiological studies indicate that the development of squamous cell carcinoma of the urinary bladder is closely associated with chronic inflammation of the urinary tract. Chronic inflammation was present in all cases; radiation-induced pseudocarcinomatous proliferations of the urinary bladder (3) and chronic subepithelial inflammation are also commonly present in intraepithelial neoplasms (13).

It seems that the very same cells and molecules that mediate bladder inflammation and the response to pathogens and trauma are integral parts of bladder cancer processes. Indeed, adhesion molecules known to have a role in inflammation are now being postulated to play a central role in the development and progression of bladder cancer (79). Another molecule that participates in bladder inflammation and cancer is IL-6. IL-6 is the primary cytokine elevated in the urine of interstitial cystitis patients (26). IL-6 functions as an autocrine growth factor for bladder carcinoma cells but not for normal urothelial cells, and it may be a factor accounting for the marked enhancement of inflammation-associated bladder carcinogenesis and tumor growth (60). A common pathway leading to a bladder response to inflammatory stimuli is NF-kappa B (89). A central role for NF-kappa B in bladder inflammation was proposed on the basis of the finding that this transcription factor stimulates the expression of enzymes whose products contribute to the pathogenesis of the inflammatory process, including the inducible form of nitric oxide synthase and inducible cyclooxygenase-2 (61). Abnormalities in the regulation of the NF-kappa B pathway are frequently seen in a variety of human malignancies including leukemias, lymphomas, and solid tumors (66). These abnormalities result in constitutively high levels of NF-kappa B in the nucleus of a variety of tumors including breast, ovarian, prostate, and colon cancers.

Neuroimmune mechanisms have been suggested to play an important role in the development of inflammatory reactions in the urinary bladder (40). One potential pathway that may mediate such neurogenic inflammation is the interaction between mast cells and the neurotransmitter substance P. The responses to substance P and other tachykinins such as neurokinin A, neurokinin B, and hemokinin B are mediated by three different G protein-coupled neurokinin receptors via changes in cytosolic calcium concentration (90). Neurokinin-1 (NK1) receptors (NK1Rs) are the predominant receptor subtype involved in inflammation and are expressed by endothelial cells, submucosal glands, and circulating leukocytes (85). In the urinary bladder, plasma extravasation induced by substance P seems to be modulated by NK1Rs, because it was inhibited by a specific receptor antagonist (33). More recently, we presented evidence of increased NK1R density during bladder inflammation (39). In addition, we have shown that in the urinary bladder, substance P receptors (NK1Rs) are downstream of NF-kappa B activation and that NK1R knockout mice failed to mount a bladder inflammatory response (89). Others have shown an increase in NK1R numbers, and distribution may underlie persistent pain such as that observed during chronic inflammation (1, 9, 55). In contrast, NK2Rs mediate detrusor muscle contraction in humans (63, 95), dogs (56, 71), hamsters (83), and mice (57), whereas NK3Rs have been found in the ganglia (35) and vascular smooth muscle (45), but they have yet to be identified in the urinary system (4).

Although there is considerable evidence that NK1Rs seem to play a critical role in the transmission of noxious stimuli (9, 47), NK1R antagonists were described to be of low efficacy in the clinical treatment of pain (41). The apparently low efficacy of NK1R antagonists in the treatment of pain in humans has been attributed to inadequate clinical trials (84). Alternatively, several dual antagonists of NK1Rs and NK2Rs have been developed (67, 77). With the exception of ZD-6021, which has been shown to dose dependently attenuate plasma extravasation in guinea pigs (6), little if any investigation of the therapeutic potential of dual antagonists has been done. Alternatively, several authors have used a combination of NK1, NK2, and NK3 antagonists to reduce visceral hyperalgesia (44).

Sensory nerves within the bladder contain high concentrations of substance P, which is a potent inducer of mast cell degranulation (93). In addition, mast cells are found in increased numbers in bladder biopsies obtained from patients with interstitial cystitis (42, 62), suggesting that mast cells potentially influence this condition. The release of mast cell mediators such as histamine, proteases, arachidonic acid metabolites, and TNF-alpha and other cytokines can potentially initiate and/or amplify inflammatory responses in the bladder (32) by stimulating sensory nerves to release substance P (11). Indeed, the inflammatory responses to mast cell tryptase are mediated through substance P release (21). This indirect evidence implies a link between substance P and mast cells in the pathogenesis of bladder inflammation, but a definitive demonstration of such a pathophysiological relation is lacking.

One area of controversy that continues to limit our understanding of substance P-mast cell interactions is the mechanism through which substance P induces mast cell degranulation. In other systems, tachykinins such as substance P interact with a family of receptors termed neurokinin receptors. The high-affinity receptor for substance P is NK1R. A straightforward speculation has been that substance P interacts with NK1Rs on mast cells. However, there is conflicting evidence in the literature as to whether mast cells express NK1Rs or whether substance P induces mast cell activation through an alternative, receptor-independent mechanism.

For example, evidence supporting mast cell expression of NK1Rs includes radiolabel binding assays suggestive of a single high-affinity binding site for substance P on freshly isolated rat peritoneal mast cells (PMCs) (59), the ability to inhibit substance P-induced degranulation of rat PMCs (58, 59) and a murine mast cell line (46) with specific NK1R antagonists, and the identification of NK1R mRNA by RT-PCR in freshly isolated PMCs (59). However, many of these findings using rat PMCs appear to depend on the strain of rat studied (58, 59). Similarly, the presence of NK1R mRNA was found in cultured RBL-2H3 cells, a rat mast cell line (18). By contrast, substantial evidence supports the concept that substance P induces mast cell degranulation by direct activation of certain G proteins (2, 53). Experiments using labeled substance P and patch-clamp techniques indicate that substance P translocates into mast cells and activates degranulation by a pertussis toxin- and GDPbeta S-sensitive mechanism (53). Our group and others have been investigating the role of substance P and NK1Rs in experimental cystitis. Our laboratory previously reported a mouse model of antigen-induced bladder inflammation characterized by histological evidence of mast cell degranulation, edema, and infiltration of the bladder by neutrophils (73). Using genetically engineered mice deficient in the expression of NK1Rs, we were able to directly evaluate the importance of NK1-substance P interactions in the development of cystitis. We reported that NK1R-/- mice exhibited no inflammation in response to antigen challenge, indicating that NK1Rs play a critical role in antigen-induced inflammation and suggesting that substance P is involved in the pathogenesis of this response (73).

In a separate study, we investigated the role of mast cells in this antigen-induced model of bladder inflammation (72). For this purpose, we examined the responses in mast cell-deficient KitW/KitW-v, congenic normal (+/+), and KitW/KitW-v mice that were reconstituted with +/+ bone marrow cells (BMR-KitW/KitW-v). This procedure repopulates mast cells and other bone marrow-derived elements in KitW/KitW-v mice. Antigen-induced bladder inflammation occurred in +/+ and BMR-KitW/KitW-v mice but not in mast cell-deficient KitW/KitW-v mice. Gene expression was determined using mouse cDNA expression arrays and indicated that gene expression associated with the inflammatory response was dependent on the presence of mast cells, i.e., certain genes were upregulated in bladders isolated from antigen-challenged +/+ and BMR-KitW/KitW-v mice but were not altered in KitW/KitW-v mice. Taken together, these studies indicate an important role for mast cells and NK1Rs in this antigen-induced model of cystitis (72-73) but do not address the nature of the interaction of substance P and mast cells.

Thus to more precisely define the role of mast cells and NK1Rs in experimental bladder inflammation, we examined the bladder inflammatory reactions in +/+, mast cell-deficient KitW/KitW-v, and BMR-KitW/KitW-v mice. The controversy concerning the expression of NK1Rs on mast cells aside, this model would permit us to examine experimental cystitis in the absence of mast cells in a host that expresses NK1Rs and then compare the reactions in the same host that now has mast cells that do not, and cannot, express NK1Rs.


    METHODS
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

Reagents. Dinitrophenyl (DNP) was conjugated to ovalbumin (OVA) or human serum albumin (HSA; Sigma, St. Louis, MO) as previously described (52). Alum was purchased from Intergen (Purchase, NY) and used as an adjuvant. Substance P was purchased from Sigma. All reagents were prepared in pyrogen-free saline immediately before use.

Animals. All animal experimentation described here was performed in conformity with the "Guiding Principles for Research Involving Animals and Human Beings" (Oklahoma University Health Sciences Center Animal Care and Use Committee, protocol no. 00-109).

Three groups of 10- to 12-wk-old female mice were used in experiments. NK1R-/- and wild-type littermate control mice were generated and backcrossed to C57BL/6J. The colony at Oklahoma University Health Sciences Center was genotyped as described previously (14). Genetically mast cell-deficient WBB6F1-KitW/KitW-v and congenic normal WBB6F1 (+/+) mice were purchased from Jackson Laboratories (Bar Harbor, ME). Animals were maintained in facilities with air-filtered cages and allowed food and water ad libitum.

Bone marrow reconstitution of mast cell-deficient KitW/KitW-v mice. Mice with double mutations at the W loci have a variety of phenotypic abnormalites, including a profound deficiency in the numbers of tissue mast cells, macrocytic anemia, age-dependent changes in intraepithelial lymphocyte populations in the gastrointestinal tract, and other nonmyeloid abnormalites (92). Bone marrow transplantation repairs both the mast cell deficiency and the anemia in KitW/KitW-v mice (92). We utilized this approach to reconstitute KitW/KitW-v mice with bone marrow from NK1R-/- mice, and thus the mast cells that develop in the bladder would not be capable of expressing the NK1R.

Briefly, femoral and tibial bone marrow cells from NK1R-/- mice were harvested in DMEM. The cells were washed three times, resuspended in DMEM, and 2 × 107 bone marrow cells/mouse were injected intravenously into the mast cell-deficient KitW/KitW-v mice. Ten weeks later, the hematocrit was determined (to confirm repair of the anemia in the mice), and the mice were then used in the experiments. At the time of death, tissue was taken for histological examination to confirm the presence or absence of mast cells.

Antigen sensitization protocol. All mice in this study were sensitized intraperitoneally with 1 µg DNP4-HSA in 1 mg alum on days 0, 7, 14, and 21. In normal mice, this protocol induces sustained levels of IgE antibodies up to 56 days postsensitization (38), and mast cell-deficient KitW/KitW-v mice have been shown to develop normal IgE responses in similar active immunization protocols (30). One week after the last sensitization, an inflammatory reaction was elicited in the bladder (see Induction of experimental cystitis).

Induction of experimental cystitis. Cystitis was induced as described previously (69-70, 72-73). Briefly, sensitized KitW/KitW-v, +/+, and BMR-NK1R-/- mice were anesthetized (40 mg/kg ketamine and 2.5 mg/kg xylazine ip), transurethrally catheterized (Angiocath, Becton Dickinson, Sandy, UT), and drained of any urine present by the application of slight digital pressure to the lower abdomen. The urinary bladders were instilled with 150 µl of either pyrogen-free saline, substance P (10 µM), or DNP4-OVA (antigen; 1 µg/ml), infused at a slow rate to avoid trauma and vesicoureteral reflux. To ensure consistent contact of substances with the bladder, infusion was repeated twice within a 30-min interval, and a 1-ml Tb syringe maintained on the catheter retained the intravesical solution for at least 1 h. The catheter was then removed, and the mice were allowed to void normally. Twenty-four hours after challenge, the mice were killed with pentobarbital sodium (20 mg/kg ip) and bladders were removed.

The urinary bladders were randomly distributed into the following groups: 1) RNA extraction (n = 3), 2) replication of RNA extraction (n = 3), and 3) morphological analysis (n = 6). Unexpectedly, one-half of mice in the BMR-NK1R-/- group challenged with antigen died of anaphylaxis within 4-6 h. In this group, all of the bladders from the mice that survived for 24 h were used for cDNA expression arrays (n = 6 mice; 2 pools of RNA), and a morphological analysis was not performed.

Histological assessment of bladder inflammation. A cross section of bladder wall was fixed in formalin, dehydrated in graded alcohol and xylene, embedded in paraffin, and cut serially into four 5-µm sections (8 µm apart) stained with either hematoxylin and eosin or Giemsa. The urinary bladders were evaluated for the presence of edema, extent of inflammatory cell accumulation, and number of mast cells present. A semiquantitative score using defined criteria of inflammation severity was used to evaluate cystitis (69, 70, 72, 73). Slides were scanned using a Nikon digital camera (DXM1200) mounted on a Nikon microscope (Eclipse E600). Image analysis was performed using the MetaMorph Imaging System (Universal Imaging, West Chester, PA). The severity of lesions in the urinary bladder was graded as follows: 1+, mild (minimal infiltrate of neutrophils in the lamina propria and little or no interstitial edema); 2+, moderate (infiltration of neutrophils into the lamina propria and moderate interstitial edema); and 3+, severe (diffuse infiltration of a large number of neutrophils in the lamina propria and severe interstitial edema) (73). Identification of mast cells and quantification of their degree of degranulation was performed in Giemsa-stained sections. The extent of degranulation is presented as the percentage of mast cells per cross section that exhibited morphological evidence of degranulation.

Sample preparation for cDNA expression arrays. cDNA expression arrays were performed as previously described (69, 72). Briefly, three bladders from each group were pooled and homogenized in Ultraspec RNA solution (Biotecx Laboratories, Houston, TX) for isolation and purification of total RNA (we had previously determined that the pooling of 3 bladders is necessary to provide enough RNA for gene array analysis). RNA was DNAse treated according to the manufacturer's instructions (Clontech Laboratories, Palo Alto, CA), and the quality of 10 µg RNA was evaluated by denaturing formaldehyde-agarose gel electrophoresis. This procedure was replicated using an additional three bladders per each experimental group. Therefore, two pools of RNA were generated per experimental group, and two separate hybridizations were performed per group.

cDNA probes prepared from each of the experimental groups were hybridized simultaneously to membranes containing Atlas Mouse 1.2 Arrays (78531, Clontech; for a complete list of genes present in this array, see http://www.clontech.com/atlas/genelists/index.html). Briefly, 5 µg of DNAse-treated RNA were reverse transcribed to cDNA and labeled with [alpha -32P]dATP according to the manufacturer's protocol (Clontech). The radioactively labeled complex cDNA probes were hybridized overnight to mouse cDNA expression arrays (Clontech) using ExpressHyb hybridization solution with continuous agitation at 68°C. After high- and low-stringency washes, the hybridized membranes were exposed overnight (at room temperature) to an ST Cyclone PhosphorImaging screen. Quantification was performed using OptiQuant image-analysis software (Packard BioScience, Downers Grove, IL). Data from each detectable spot were expressed as digital light units (DLUs). The results were placed in an Excel (Microsoft) spreadsheet, and the background DLUs were subtracted. Within each membrane, expression was calculated as the percentage of beta -actin, as previously described (72). PhosphorImages of each array were first imported into ATLASIMAGE software (ver. 1.5; Clontech) for densitometric measurement of gene- and array-specific parameters such as intensity and global background levels.

Background subtraction. Background represented by empty rows and columns was averaged. This background intensity was deducted from the intensity of each spot. In addition, the intensity of negative controls M13 mp18 (+) strand DNA, lambda -DNA, and pUC18 DNA were taken as unspecific hybridization and averaged. This unspecific hybridization was deducted from the intensity of each spot. Only signals with 3 SD above background were used. This was done to avoid ratios either mathematically impossible or astronomically high. The values for all genes with background-subtracted intensities >1 were then exported to an Excel spreadsheet to generate a sorted data matrix for all arrays within each experiment. At this point, data were filtered to eliminate genes detected fewer than two times in each condition and replicate array values normalized by the sum-of-intensities method. Total intensities from the two conditions were then scaled, and mean intensities were calculated for scatterplot visualization of the fold-changes. The signal intensities were normalized to the mean signal intensity of all the spots on the array. Duplicate spots of each cDNA were averaged. To compare two arrays (2 conditions), expression was calculated as the percentage of beta -actin, as previously described (72).

Experimental criteria. To determine which genes were dependent on the presence of tissue mast cells but independent of NK1Rs, we set the following arbitrary criteria: 1) the gene should be upregulated at least threefold in response to antigen or substance P challenge in +/+ tissues; 2) the same gene should not be upregulated in the absence of mast cells (KitW/KitW-v mice); and 3) upregulation should be demonstrated in BMR-NK1R-/- mice.

Cluster analysis using self-organizing maps. Genes fulfilling the above criteria were identified on the basis of a Euclidean distance metric, after the elimination of genes that failed to vary in expression level within an experiment by a factor of 3 SE and an absolute value of 100% of beta -actin and normalization within experiments to a mean of 0 and an SD of 1.

Cluster analysis was performed using self-organizing maps (SOMs), as described by Tamayo and co-workers (80). SOMs are an unsupervised network learning algorithm that has been successfully used for the analysis and organization of large data files (27, 80, 82). We applied SOMs to analyze the time course of gene regulation during LPS-induced cystitis (69), and others have shown that SOMs are an excellent tool for the analysis and visualization of gene expression profiles (27, 80, 82). SOMs are a type of mathematical cluster analysis that is particularly well suited to recognizing and classifying features in complex, multidimensional data (80). The method has been implemented in GeneCluster software, which performs the analytical calculations and provides easy data visualization (http://www-genome.wi.mit.edu/cancer/software/software.html). This focuses attention on the "shape" of expression patterns rather than on absolute levels of expression. The advantage of this approach is that large data sets can be clustered much more quickly than by using hierarchical clustering because a lower number of clusters are assigned.

The technique employed here comprises essentially different ways of clustering points in multidimensional space. They can be directly applied to gene expression by regarding the quantitative expression levels of n genes in k samples as defining n points in k-dimensional space (80). The nodes are mapped into k-dimensional space, initially at random, and then iteratively adjusted. Each interaction involves randomly selecting a data point P and moving the nodes in the direction of P. The closest node NP is moved the most, whereas other nodes are moved by smaller amounts depending on their distance from NP in the initial geometry. In this fashion, neighboring points in the initial geometry tend to be mapped to nearby points in k-dimensional space. The process continues for 20,000-50,000 iterations. SOMs impose structure on the data, with neighboring nodes tending to define related clusters (80).

An SOM has a set of nodes with a simple topology (e.g., 2-dimensional grid) and a distance function d(N1,N2) on the nodes (80). Nodes are iteratively mapped into k-dimensional "gene expression" space (in which the ith coordinate represents the expression level in the ith sample). The position of node N at iteration i is denoted fi(N). The initial mapping, f0, is random. On subsequent iterations, a data point P is selected and the node NP that maps nearest to P is identified. The mapping of nodes is then adjusted by moving points toward P by the formula (69, 80)
f<SUB>i+1</SUB>(N)=f<SUB>i</SUB>(<IT>N</IT>)<IT>+&tgr;</IT>[<IT>d</IT>(<IT>N, N<SUB>P</SUB></IT>)<IT>, i</IT>][<IT>P − f</IT><SUB>i</SUB> (<IT>N</IT>)]
The learning rate tau  decreases with distance of node N from NP and with iteration number i. The point P used at each iteration is determined by random ordering of the n data points generated once and recycled as needed. The function tau  is defined by tau (x, i) = 0.02T/(T +100i) for × 5 rho (i) and tau  (x, i) = 0 otherwise, where radius rho (i) decreases linearly with i [rho (0) = 3] and eventually becomes 0 (80).

Data preprocessing. Using GeneCluster, SOMs were constructed by choosing a 6 × 4 grid that generated 24 clusters. It was also our concern to present as few clusters as possible that would still give a clear picture of substance P- and antigen-induced gene expression. Increasing the number of clusters by increasing the grid did not gave us any additional correlation between genes. A variation filter was used to eliminate genes that did not change significantly across samples.

Array reproducibility. To determine the reproducibility of the arrays, we performed a regression analysis using an Excel spreadsheet. This test compared the results obtained with two pools of RNA isolated from sensitized wild-type mice that were challenged with saline and hybridized separately. The same analysis was performed using RNA isolated from sensitized wild-type mice that were challenged with antigen and substance P.

Venn diagrams. For representations of the numbers of genes up- or downregulated in each group as well as genes common to two or more groups, we used Venn diagram analysis performed by GeneSpring software [Silicon Genetics, Redwood City, CA (http://www.silicongenetics.com/cgi/SiG.cgi/index.smf.)] using raw data and filtering genes that were upregulated at least threefold in a comparison of substance P-, antigen-, and saline-challenged bladders isolated from +/+ , KitW/KitW-v, and BMR-NK1R-/- mice. Another set of analyses was performed by comparing genes upregulated or downregulated in antigen- and saline-challenged groups. Finally, we determined genes that satisfied all three conditions in response to either antigen or substance P.

Statistical analysis. For each group, 1,200 genes were analyzed by 2 different hybridizations using 2 different pools of RNA isolated from sensitized KitW/KitW-v, +/+, and BMR-NK1R-/- mice that were challenged with saline, antigen, or substance P. Therefore, a total of 21,600 data points were analyzed. SOMs were constructed by choosing a 6 × 4 grid that generated 24 clusters. As this type of analysis assumes that the data can be divided into a certain number of clusters and that they are well separated, it was also our concern to present the fewest clusters possible that would still give a clear picture of antigen- and substance P-induced gene expression. Increasing the number of clusters by increasing the grid did not produce any additional correlation between genes. GeneCluster software provides a list of genes present in each cluster and a centroid. However, to permit comparisons, in each cluster, gene expression was averaged and the SE was calculated. In each cluster, comparisons between gene regulation in response to antigen or substance P challenge were obtained by the ratio of gene expression in relation to the corresponding saline group. Significant differences were determined using an unpaired Student's t-test. The statistical analysis of histological data was performed using Wilcoxon's rank-sum test. Results are expressed as means ± SE. The n values reported refer to the number of animals used for each experiment. In all cases, a value of P < 0.05 was considered to represent a significant difference.


    RESULTS
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

Morphological assessment of substance P-induced cystitis. We recently reported that mast cells were critical to the inflammatory response and gene expression associated with antigen-induced cystitis (72). In that study, we validated the approach of repopulating bladder mast cells in mast cell-deficient KitW/KitW-v mice by transplantation of bone marrow from normal +/+ mice (BMR-KitW/KitW-v).

Using a similar approach, we now examined the histological changes and requirement for NK1R expression on mast cells during substance P-induced cystitis. For this study, normal +/+, mast cell-deficient KitW/KitW-v, and BMR-NK1R-/- mice were used. In parallel, we also examined the response to antigen-induced inflammation for two reasons. We wished to examine whether the response was similar in this model (BMR-NK1R-/-) as was previously found using BMR-+/+ mice and compare the genes expressed during substance P- and antigen-induced bladder inflammation.

We found that either mode of experimental inflammation resulted in significant edema and neutrophil infiltration in normal +/+ mice compared with respective, saline-challenged normal +/+ mice (Table 1). Neither the substance P- nor antigen-induced inflammatory responses were associated with remarkable edema or neutrophil accumulation in the bladders of mast cell-deficient KitW/KitW-v mice (Table 1). However, elicitation of substance P-induced cystitis in BMR-NK1R-/- mice resulted in edema and neutrophil infiltration similar to that seen in the substance P-challenged +/+ mice (Table 1). In addition, we found morphologically identifiable mast cells in the bladders of BMR-NK1R-/- mice (Table 1). Unexpectedly, 50% of the sensitized BMR-NK1R-/- mice died within 4-6 h after intravesical challenge with antigen. Therefore, the bladders of the mice that survived for at least 24 h were used for gene array analysis, and no histological evaluation was performed (Table 1).

                              
View this table:
[in this window]
[in a new window]
 
Table 1.   Morphological assessment of extent of inflammation and mast cell numbers in experimental cystitis

Reproducibility of array hybridization. We previously presented evidence of the reproducibility of gene array methodology for the analysis of bladder inflammatory genes (69, 72) and verified the results using RNase protection assays (69). In the present work, we determined the reproducibility of our hybridization technique by performing regression analysis from values obtained with two different pools of RNA. Figure 1A represents regression analysis of RNA isolated from the bladder of sensitized +/+ mice that were challenged with saline (Fig. 1A). Similar analysis was performed using RNA isolated from the bladder of +/+ mice that were challenged with either substance P (Fig. 1B) or antigen (Fig. 1C). The calculated correlation coefficients for +/+ mice challenged with saline, antigen, or substance P were 0.9838, 0.9628, and 0.9718, respectively.


View larger version (15K):
[in this window]
[in a new window]
 
Fig. 1.   A-C: reproducibility of gene arrays. Regression analysis between 2 pools of RNA (1 and 2) isolated from sensitized normal (+/+) mice that were challenged with saline (A), substance P (SP; B), or antigen (C) was performed. cDNA probes were prepared from DNAse-treated RNAs, and 2 separate hybridizations to Atlas Mouse cDNA Expression Arrays were performed. Background counts were subtracted, and gene expression was calculated as percentage of beta -actin.

Role of mast cell NK1R expression on gene expression during experimental cystitis. Figure 2 is a Venn diagram showing the genes that were upregulated at least threefold in each animal group 24 h after substance P or antigen challenge (Fig. 2, A and B, respectively). The diagram indicates genes commonly upregulated between two or more groups. Interestingly, the genes expressed by mast cell-deficient KitW/KitW-v mice as a result of antigen challenge differed fundamentally from those observed with substance P challenge. In the mast cell-deficient KitW/KitW-v mice, only 13 genes were uniquely expressed after antigen challenge compared with 128 genes upregulated in response to substance P. In addition, the Venn diagram also highlights genes that were dependent on tissue mast cells for their upregulation but independent of the presence of NK1R on the mast cells. Thirty-two genes in the group of antigen-challenged mice and 21 genes in the group challenged with substance P fulfilled our criteria of being upregulated at least 3-fold in tissues isolated from +/+ and BMR-NK1R-/- but not in tissues isolated from mast cell-deficient KitW/KitW-v mice. In the presence of mast cells, only four genes were always upregulated in either inflammatory response (antigen or substance P). Those genes are represented by cluster 1 (Fig. 3A; Table 2) and include serine protease inhibitor (Spi) 2.2 (M64086), maspin (U54705), mitogen- and stress-activated protein kinase 2 (MSK2; AF074714), and macrophage colony-stimulating factor 1 (CSF-1 or M-CSF; X05010).


View larger version (15K):
[in this window]
[in a new window]
 
Fig. 2.   Cluster analysis of genes upregulated during SP- or antigen (Ag)-induced cystitis and the influence of the presence of mast cells and mast cells lacking neurokinin-1 receptors (NK1Rs). The ratio of gene expression obtained in bladders isolated from sensitized +/+, mast cell-deficient KitW/KitW-v (Kit), and Kit mice reconstituted with bone marrow mast cells from NK1R-/- mice (BMR-NK1-/-) during SP (A)- or antigen (B)-induced cystitis was determined using GeneSpring software. Venn diagrams were generated using raw data and filtering genes that were upregulated at least 3-fold compared with respective saline-challenged mice. All mice were sensitized and killed 24 h after intravesical challenge. Yellow, congenic +/+ mice; blue, Kit mice; and red, BMR-NK1-/- mice.



View larger version (41K):
[in this window]
[in a new window]
 
Fig. 3.   A-E: clusters 1-5. Kit, +/+, and BMR-NK1R-/- mice were challenged with either Ag, SP, or saline. Twenty-four hours later, the urinary bladders of 3 mice were isolated, homogenized, and processed for isolation of RNA. The RNA was reverse transcribed to cDNA, labeled with [alpha -32P]dATP, and hybridized to membranes containing Atlas Mouse cDNA Expression Arrays. Radioactivity was quantified using a PhosphorImager and interpreted by OptiQuant image-analysis software. Gene expression was normalized to beta -actin expression (72) and analyzed by GeneGluster software using self-organizing maps (SOMs) as described before (69, 72, 80). For each group, 1,200 genes were analyzed by 2 different hybridizations using 2 different pools of RNA isolated from sensitized Kit, +/+, and BMR-NK1R-/- mice that were challenged with saline, Ag, or SP. Therefore, a total of 21,600 data points were analyzed. SOMs were constructed by choosing a 6 × 4 grid that generated 24 clusters, and 5 selected clusters are represented. The gene composition of cluster 1 is found in Table 2, and those for clusters 2-5 are presented in Table 3.


                              
View this table:
[in this window]
[in a new window]
 
Table 2.   Cluster 1: Mast cell-dependent genes commonly upregulated by antigen and substance P

The 21 mast cell-dependent genes that were induced by substance P were further sorted into 2 clusters (clusters 2 and 3; Fig. 3, B and C, respectively, and Table 3). Cluster 2 contained highly expressed genes that included fos-B, prostaglandin I2 (prostacyclin) synthase, Fas l receptor, ACHE, and voltage-gated sodium channel. Cluster 3 contained genes expressed at lower levels but that still fulfilled the established criteria and included tubulin beta -4, P-selectin glycoprotein ligand 1, calcium-activated potassium channel K (VCA)-beta , and RAB17 (RAS oncogene family).

                              
View this table:
[in this window]
[in a new window]
 
Table 3.   Mast cell-dependent genes upregulated by substance P and antigen

The 32 mast cell-dependent genes that were upregulated during antigen challenge were sorted into 2 clusters (clusters 4 and 5; Fig. 3, D and E, respectively, and Table 3). Cluster 4 contained highly expressed genes, including TNF receptor-1, interferon-gamma receptor, dihydropyridine-sensitive calcium channel, GLYCAM-1, interferon regulatory factor 1, IL-5Ralpha , HBGF-8, and transcription factors. Cluster 5 contained genes expressed at lower levels, including rab2 ras-related protein, retinoic X receptor, fibroblast growth factor 3 precursor, Met protooncogene, and SHC-transforming protein. Table 3 presents all genes contained in clusters 2-5.

For the sake of completeness, we also have presented genes that had their downregulation dependent on the presence of mast cells. Mast cell-dependent genes that were downregulated in response to antigen or substance P are presented in Table 4, whereas genes that were downregulated specifically by antigen are in Table 5, and those specifically downregulated by substance P are in Table 6.

                              
View this table:
[in this window]
[in a new window]
 
Table 4.   Mast cell-dependent genes commonly downregulated by antigen and substance P


                              
View this table:
[in this window]
[in a new window]
 
Table 5.   Mast cell-dependent genes downregulated by antigen


                              
View this table:
[in this window]
[in a new window]
 
Table 6.   Mast cell-dependent genes downregulated by substance P

Figure 4 depicts a proposed mechanism by which mast cell and sensory nerves amplify a proinflammatory stimulus such as antigen stimulation or substance P administration. Activation of sensory nerves leads to release of substance P. Substance P induces mast cell degranulation (11) and gene upregulation independently of NK1Rs on mast cells. In addition, mast cell degranulation leads to activation of sensory nerves. Mast cell mediators such as tryptase, acting on protease-activated receptors, and prostaglandin D2, acting on prostaglandin receptors, promote further release of substance P (21). The amplification of substance P release by mast cells seems to be essential because mast cell-deficient mice presented a reduced inflammatory response compared with wild-type mice. Finally, NK1Rs present on blood vessels, endothelial cells, and smooth muscle cells play an absolutely mandatory role in inflammation because NK1R-/- mice do not mount inflammation in response to substance P and antigen stimulation despite an increased number of mast cells (73).


View larger version (52K):
[in this window]
[in a new window]
 
Fig. 4.   Sensory nerve-mast cell communication in cystitis. Activation of sensory nerves leads to release of SP, which stimulates mast cells to release their granules and to modulate gene upregulation independently of NK1Rs on mast cells. Mast cell mediators such as PGD2, histamine, and tryptase activate sensory nerves to further release SP. PGD2 activates prostaglandin receptors (PG), whereas tryptase responses are modulated by protease-activated receptors (PAR) present on bladder sensory nerves (21). The amplification of SP release by mast cells seems to be essential because mast cell-deficient mice presented a reduced inflammatory response compared with wild-type mice. Finally, NK1Rs present on blood vessels, endothelial cells, and smooth muscle cells play an absolutely mandatory role in inflammation because NK1R-/- mice do not mount inflammation to SP and antigen stimulation despite an increased number of mast cells (73).


    DISCUSSION
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

Several lines of indirect evidence suggest that substance P-mast cell interactions are involved in the development of interstitial cystitis. We now provide direct evidence that mast cells play a critical role in the pathogenesis of substance P-induced cystitis in mice. In the absence of mast cells, no demonstrable tissue edema or neutrophil infiltration was noted. However, these findings do not absolutely prove that mast cells are the cells responsible for this effect. For example, some other defect resulting from W mutations could theoretically account for the difference seen between +/+ and mast cell-deficient KitW/KitW-v mice.

To further investigate the role of mast cells in this reaction and whether mast cell-substance P interactions occurred via a mechanism utilizing the NK1R, we reconstituted mast cell-deficient KitW/KitW-v mice with bone marrow cells derived from NK1R-deficient mice. This unique animal model permitted us to examine the reaction in a tissue milieu where only the mast cell was deficient in NK1R expression. We found that inflammation induced by substance P was similar in BMR-NK1R-/- compared with normal +/+ mice. This finding provides definitive proof that mast cells are essential for substance P-induced bladder inflammation and that NK1Rs on mast cells are not required.

At this time, we do not know the mechanism involved in mast cell activation by substance P in the bladder. Clearly, NK1Rs are not involved; however, NK2R may play a role. In guinea pigs, an NK2R-dependent mechanism has been suggested in the activation of lung mast cells by substance P (51). Alternatively, a receptor-independent mechanism may be involved (52). In either case, it is important to emphasize that our results pertain to murine mast cells in the bladder, and given the known heterogeneity that exists in mast cell populations, these results should not be generalized to other anatomic sites or species.

We observed that 50% of sensitized mice bearing NK1R-/- mast cells died as a consequence of intravesical antigen challenge. In more than six years of research challenging sensitized mice with intravesical antigen administration, this was the first time that we lost mice to anaphylaxis. Our previous work indicates that this outcome was not observed when mast cells from wild-type mice were used to reconstitute the mast cell-deficient mice (72). However, NK1R-/- mice have at least three times more mast cells in the urinary bladder (73), and the same is valid in the stomach and esophagus (Saban R, unpublished observations). It is not clear why the absence of substance P receptors would lead to an increase in tissue mast cell numbers (72). Together with the present findings, our data strongly indicate that mast cells from NK1R-/- mice present an overwhelming response to antigen stimulation. Future studies are necessary to fully explain the differences between wild-type and NK1R-/- mast cells.

We next focused our attention on the genes expressed during experimental bladder inflammation. For this purpose, we examined gene expression in antigen- and substance P-induced cystitis, both of which we have now characterized as mast cell-dependent forms of inflammation (72). We found that there were more mast cell-dependent genes expressed during substance P responses than in antigen-induced bladder inflammation; i.e., only 13 genes were upregulated in KitW/KitW-v mice during antigen-induced reactions compared with 128 genes upregulated during responses to substance P. This is not entirely unexpected because relatively few cell types express IgE receptors compared with multiple immune and nonimmune cells that express receptors capable of interacting with substance P.

Although our results indicate that an NK1R on the mast cell is not necessary for the communication between sensory nerves and this immune cell, others have presented indirect evidence that activation of PMNs by substance P requires NK1Rs. In this instance, substance P primes PMNs exposed to recombinant IL-8 and suggested that substance P-priming effects are receptor mediated (24). In addition, substance P activates PMNs to release cytokines such as TNF-alpha and interferon-gamma , and this effect is potentiated by bacterial LPS (70). However, further studies using adequate animal models such as the one described in the present work are necessary to definitively access the role of neurokinin receptors on inflammatory cells.

In normal +/+ or BMR-NK1R-/- mice, only four mast cell-dependent genes were upregulated by either substance P or antigen. Those included Spi 2.2, maspin, MSK2, and M-CSF (or CSF-1). Interestingly, all four genes modulate inflammatory responses.

M-CSF (or CSF-1) acts to regulate the development and function of cells of the macrophage lineage (19). Macrophages are involved in inflammatory responses and, on treatment with M-CSF, displayed greater secretion of IL-6 (5). It was suggested that IL-6 occupies a central role in the CSF-1-regulated macrophage response to infection (36). In addition, it was suggested that the interplay between IL-6 and M-CSF switches monocyte differentiation to macrophages rather than dendritic cells and that IL-6 is an essential factor in the molecular control of antigen-presenting cell development (17).

Spi 2.2 is a member of the serpin gene family (74). In terms of physiological regulation of rat Spi 2.1 and 2.2 mRNAs, Spi 2.1 is dependent on growth hormone (GH) for maximal mRNA content (48, 94), whereas Spi 2.2 mRNA is not GH dependent (74). Spi 2 not only acts as a basal promoter element but also mediates transcriptional activation by GH and IL-6 (50). Spi 2 is known to be associated with proinflammatory processes. In the liver, Spi 2 genes have been correlated with the acute-phase response (8). These effects are mediated by the products of stimulated monocytes and macrophages in combination with glucocorticoids (7). In rat hepatocytes, turpentine induced an acute-phase response and an increase in Spi 2.2 expression, which was mediated primarily by IL-6 (7). The role of IL-6 on Spi 2.2 upregulation in the bladder should be further pursued because this cytokine is consistently elevated in patients with cystitis (26).

Maspin, another serine protease inhibitor related to the serpin family, is a secreted protein encoded by a class II tumor suppressor gene, whose downregulation is associated with the development of breast and prostate cancers (54). Maspin is associated with tumorigenesis, inflammation, and protection from autolysis by granule proteinases (75). Maspin interacts with the p53 tumor-suppressor pathway and functions as an inhibitor of angiogenesis in vitro and in vivo (96).

MSK2 is a novel protein kinase that is activated in vitro and in vivo by either MAPK/ERKs or SAPK2/p38. MSK1 (and/or MSK2) mediates activation of the transcription factors calcium/cAMP response element binding protein and activating transcription factor 1 by either growth factors or stress signals (20). Others have shown that substance P activates MAPKs (15) and, at nanomolar concentrations, induces the human astrocytoma cell line U373 MG to produce IL-6 by a p38 MAPK-dependent pathway (29).

The result of upregulation of proteases such as Spi 2.2 and maspin, SAPK, and CSF-1 by either substance P- or antigen-induced responses indicates a common mast cell-dependent pathway involved in acute bladder inflammation. These results suggest that activation of mast cells initiates activation of monocytes and/or macrophages and their resulting products such as IL-6 may modulate bladder inflammation.

There are several available methods for the analysis of gene microarray data, based on different mathematical assumptions and algorithms. Each of the analysis methods has advantages and limitations (65, 76).

An increasingly common approach involves using gene expression behavior observed over multiple experiments to first cluster genes together into groups, either by manually examining the data (16) or by using statistical methods such as SOMs (80), K-means clustering, or hierarchical clustering (25, 78). K-means clustering is a completely unstructured approach, which proceeds in an entirely local fashion and produces an unorganized collection of clusters that is not conducive to interpretation (80). Several recent papers employed hierarchical clustering algorithms to organize genes into a phylogenetic tree, reflecting similarity in expression patterns. However, the interpretation of these clusters is left to the observer.

We preferred to use SOMs because they are potent tools for identifying clusters involved in biologically related pathways and mechanisms (25, 69, 80, 82). The basic assumption underlying this unsupervised analysis is that genes with similar expression behavior (for example, increasing and decreasing together under similar circumstances) are likely to be related functionally. Although not logically rigorous, the utility of the cluster approach has been demonstrated, as genes already known to be related do, in fact, tend to cluster together based on their experimentally determined expression patterns (69). The validity of this approach has been demonstrated for many genes in Saccharomyces cerevisiae, a simple organism for which the entire genomic sequence and the functional roles of ~60% of the genes are known (25, 91). One limitation of SOMs is that genes negatively associated are not clustered.

SOMs can be performed by using Cluster software (http://rana.lbl.gov/) developed by Michael Eisen's laboratory or GeneSpring (http://www.silicongenetics.com/cgi/SiG.cgi/index.smf.), which permit the calculation of SOMs, K-means, and principal component analysis on the same set of data. The reason for the use of SOMs with GeneCluster software developed by Tamayo et al. (80) is that it performs the analytical calculations and provides easy data visualization. The approach is made more systematic and statistically sound by calculating the probability that the observed functional distribution of differentially expressed genes could have happened by chance. The application of statistical rigor is essential to avoid overly subjective interpretations of the results based on the predispositions, prior knowledge, and interests of the individual researcher (49). The power of cluster analysis was enhanced in the present work by the use of an excellent biological paradigm and a stringent criterion for cluster selection.

To fairly interpret gene cluster analysis, we must be cognizant of a growing body of evidence of mechanisms that control the rate of synthesis and half-life of proteins (31) and that gene and protein changes can be dissociated (31, 37). Future proteomic correlation must determine how directly mRNA changes reflect translated protein levels and the physiological consequence of these proteins.

In conclusion, our results confirm a mandatory role of mast cells in substance P-induced bladder inflammation in mice and suggest that mast cells mediate inflammation and gene regulation in the absence of NK1Rs on its cell surface. The cDNA array experimental approach provides a global profile of gene expression changes in bladder tissue after stimulation with antigen or substance P. SOMs identified functionally significant gene clusters. These gene expression responses may represent a balance between the cytoprotective and inflammatory processes that accompany bladder response to injury. Gene cluster analysis techniques can be applied in the future to begin to understand clinically relevant issues, such as how and why the transition from acute to chronic inflammation occurs only under selected circumstances and which therapeutic strategies can be used to selectively target genes expressed in bladder inflammation. However, cluster analysis and gene array profiling will have a strong impact whenever unique animal models such as KitW/KitW-v, BMR, and BMR-NK1R-/- are included, which allow establishment of biological paradigms to be tested.


    ACKNOWLEDGEMENTS

This work was supported by National Institute of Diabetes and Digestive and Kidney Diseases Grants DK-55828-01 (R. Saban), DK-46819 (B. K. Wershil), and DK-33506 (B. K. Wershil) and National Heart, Lung, and Blood Institute Grant HL-41587 (N. P. Gerard).


    FOOTNOTES

Address for reprint requests and other correspondence: R. Saban, Dept. of Physiology, College of Medicine, Univ. of Oklahoma Health Sciences Center (OUHSC), 940 SL Young Blvd., Rm. 605, Oklahoma City, OK 73104-0505 (E-mail: ricardo-saban{at}ouhsc.edu).

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

May 22, 2002;10.1152/ajprenal.00096.2002

Received 12 March 2002; accepted in final form 9 May 2002.


    REFERENCES
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

1.   Abbadie, C, Brown JL, Mantyh PW, and Basbaum AI. Spinal cord substance P receptor immunoreactivity increases in both inflammatory and nerve injury models of persistent pain. Neuroscience 70: 201-209, 1996[ISI][Medline].

2.   Aridor, M, Traub LM, and Sagi-Eisenberg R. Exocytosis in mast cells by basic secretagogues: evidence for direct activation of GTP-binding proteins. J Cell Biol 111: 909-917, 1990[Abstract].

3.   Baker, PM, and Young RH. Radiation-induced pseudocarcinomatous proliferations of the urinary bladder: a report of 4 cases. Human Pathol 31: 678-683, 2000[ISI][Medline].

4.   Beaujouan, JC, Saffroy M, Torrens Y, and Glowinski J. Potency and selectivity of the tachykinin NK3 receptor antagonist SR 142801. Eur J Pharmacol 319: 307-316, 1997[ISI][Medline].

5.   Bernier, T, Tschernig T, Pabst R, Macke O, Steinmueller C, and Emmendorffer A. Effects of macrophage-CSF on pulmonary-macrophage repopulation after bone marrow transplantation. J Leukoc Biol 70: 39-45, 2001[Abstract/Free Full Text].

6.   Bernstein, PR, Aharony D, Albert JS, Andisik D, Barthlow HG, Bialecki R, Davenport T, Dedinas RF, Dembofsky BT, Koether G, Kosmider BJ, Kirkland K, Ohnmacht CJ, Potts W, Rumsey WL, Shen L, Shenvi A, Sherwood S, Stollman D, and Russell K. Discovery of novel, orally active dual NK1/NK2 antagonists. Bioorg Med Chem Lett 11: 2769-2773, 2001[ISI][Medline].

7.   Berry, SA, Bergad PL, and Bundy MV. Expression of growth hormone-responsive serpin messenger RNAs in perinatal rat liver. Am J Physiol Endocrinol Metab 264: E973-E980, 1993[Abstract/Free Full Text].

8.   Berry, SA, Bergad PL, Stolz AM, Towle HC, and Schwarzenberg SJ. Regulation of Spi 2.1 and 2.2 gene expression after turpentine inflammation: discordant responses to IL-6. Am J Physiol Cell Physiol 276: C1374-C1382, 1999[Abstract/Free Full Text].

9.   Bester, H, De Felipe C, and Hunt SP. The NK1 receptor is essential for the full expression of noxious inhibitory controls in the mouse. J Neurosci 21: 1039-1046, 2001[Abstract/Free Full Text].

10.   Bhan, R, Pisharodi LR, Gudlaugsson E, and Bedrossian C. Cytological, histological, and clinical correlations in intravesical bacillus Calmette-Guerin immunotherapy. Ann Diagn Pathol 2: 55-60, 1998[Medline].

11.   Bjorling, DE, Saban MR, and Saban R. Neurogenic inflammation of the guinea pig bladder. Mediators Inflamm 3: 189-197, 1994[ISI].

12.   Bohle, A. BCG's mechanism of action---increasing our understanding. For the EBIN Group. Eur Urol 37, Suppl1: 1-8, 2000.

13.   Boone, CW, and Kelloff GJ. Endpoint markers for clinical trials of chemopreventive agents derived from the properties of epithelial precancer (intraepithelial neoplasia) measured by computer-assisted image analysis. Cancer Surv 32: 133-147, 1998[ISI][Medline].

14.   Bozic, CR, Lu B, Hopken UE, Gerard C, and Gerard NP. Neurogenic amplification of immune complex inflammation. Science 273: 1722-1725, 1996[Abstract/Free Full Text].

15.   Castagliuolo, I, Valenick L, Liu J, and Pothoulakis C. Epidermal growth factor receptor transactivation mediates substance P-induced mitogenic responses in U-373 MG cells. J Biol Chem 275: 26545-26550, 2000[Abstract/Free Full Text].

16.   Cho, RJ, Campbell MJ, Winzeler EA, Steinmetz L, Conway A, Wodicka L, Wolfsberg TG, Gabrielian AE, Landsman D, Lockhart DJ, and Davis RW. A genome-wide transcriptional analysis of the mitotic cell cycle. Mol Cell 2: 65-73, 1998[ISI][Medline].

17.   Chomarat, P, Banchereau J, Davoust J, and Palucka AK. IL-6 switches the differentiation of monocytes from dendritic cells to macrophages. Nat Immun 1: 510-514, 2000[ISI].

18.   Cooke, HJ, Fox P, Alferes L, Fox CC, and Wolfe SA, Jr. Presence of NK1 receptors on a mucosal-like mast cell line, RBL-2H3 cells. Can J Physiol Pharmacol 76: 188-193, 1998[ISI][Medline].

19.   Csar, XF, Wilson NJ, McMahon KA, Marks DC, Beecroft TL, Ward AC, Whitty GA, Kanangasundarum V, and Hamilton JA. Proteomic analysis of macrophage differentiation. p46/52(Shc) Tyrosine phosphorylation is required for CSF-1-mediated macrophage differentiation. J Biol Chem 276: 26211-26217, 2001[Abstract/Free Full Text].

20.   Deak, M, Clifton AD, Lucocq LM, and Alessi DR. Mitogen- and stress-activated protein kinase-1 (MSK1) is directly activated by MAPK and SAPK2/p38, and may mediate activation of CREB. EMBO J 3: 4426-4441, 1988.

21.   De Garavilla, L, Vergnolle N, Young SH, Ennes H, Steinhoff M, Ossovskaya VS, D'Andrea MR, Mayer EA, Wallace JL, Hollenberg MD, Andrade-Gordon P, and Bunnett NW. Agonists of proteinase-activated receptor 1 induce plasma extravasation by a neurogenic mechanism. Br J Pharmacol 133: 975-987, 2001[Abstract/Free Full Text].

22.   Delnay, KM, Stonehill WH, Goldman H, Jukkola AF, and Dmochowski RR. Bladder histological changes associated with chronic indwelling urinary catheter. J Urol 161: 1106-1108, 1999[ISI][Medline].

23.   De Seze, M, Wiart L, Joseph PA, Dosque JP, Mazaux JM, and Barat M. Capsaicin and neurogenic detrusor hyperreflexia: a double-blind placebo-controlled study in 20 patients with spinal cord lesions. Neurourol Urodyn 17: 513-523, 1998[ISI][Medline].

24.   Dianzani, C, Lombardi G, Collino M, Ferrara C, Cassone MC, and Fantozzi R. Priming effects of substance P on calcium changes evoked by interleukin-8 in human neutrophils. J Leuk Biol 69: 1013-1018, 2001[Abstract/Free Full Text].

25.   Eisen, MB, Spellman PT, Brown PO, and Botstein D. Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA 95: 14863-14868, 1998[Abstract/Free Full Text].

26.   Erickson, DR, Belchis DA, and Dabbs DJ. Inflammatory cell types and clinical features of interstitial cystitis. J Urol 158: 790-793, 1997[ISI][Medline].

27.   Everitt, BS, Landau S, and Leese M. Cluster Analysis (4th ed.). New York: Oxford University Press, 2001.

28.   Fall, M, Johansson SL, and Aldenborg F. Chronic interstitial cystitis: a heterogeneous syndrome. J Urol 137: 35-38, 1987[ISI][Medline].

29.   Fiebich, BL, Schleicher S, Butcher RD, Craig A, and Lieb K. The neuropeptide substance P activates p38 mitogen-activated protein kinase resulting in IL-6 expression independently from NF-kappa B. J Immunol 165: 5606-5611, 2000[Abstract/Free Full Text].

30.   Furuta, GT, Schmidt-Choudhury A, Wang MY, Wang ZS, Lu L, Furlano RI, and Wershil BK. Mast cell-dependent tumor necrosis factor alpha production participates in allergic gastric inflammation in mice. Gastroenterology 113: 1560-1569, 1997[ISI][Medline].

31.   Futcher, B, Latter GI, Monardo P, McLaughlin CS, and Garrels JI. A sampling of the yeast proteome. Mol Cell Biol 19: 7357-7368, 1999[Abstract/Free Full Text].

32.   Galli, SJ. New concepts about the mast cell. N Engl J Med 328: 257-265, 1993[Free Full Text].

33.   Gao, GC, and Wei ET. Inhibition of substance P-induced vascular leakage in rat by N-acetyl-neurotensin-(8-13). Regul Pept 58: 117-121, 1995[ISI][Medline].

34.   Giannantoni, A, Di Stasi S, Stephen RL, Navarra P, Scivoltetto G, Mearini E, and Porena M. Intravesical capsaicin versus resinferatoxin in patients with detrusor hyperreflexia: A prospective randomized study. J Urol 167: 1710-1714, 2002[ISI][Medline].

35.   Grady, EF, Baluk P, Bohm S, Gamp PD, Wong H, Payan DG, Ansel J, Portbury AL, Furness JB, McDonald DM, and Bunnett NW. Characterization of antisera specific to NK1, NK2, and NK3 neurokinin receptors and their utilization to localize receptors in the rat gastrointestinal tract. J Neurosci 16: 6975-6986, 1996[Abstract/Free Full Text].

36.   Guleria, I, and Pollard JW. Aberrant macrophage and neutrophil population dynamics and impaired Th1 response to Listeria monocytogenes in colony-stimulating factor 1-deficient mice. Infect Immun 69: 1795-1807, 2001[Abstract/Free Full Text].

37.   Gygi, S, Rochon Y, Franza BR, and Aebersold R. Correlation between protein and mRNA abundance in yeast. Mol Cell Biol 19: 1720-1730, 1999[Abstract/Free Full Text].

38.   Haak-Frendscho, M, Saban R, Shields R, and Jardieu P. Blockade of histamine release and tissue contraction by antibodies to IgE in an established IgE response. Immunology 94: 115-121, 1998[ISI][Medline].

39.   Hammond, TG, Saban R, Bost KL, Harris HW, Jr, Kaysen JH, Goda FO, Navar GL, Bjorling DE, Saban M, and Zeidel ML. Inhibition of endosomal fusion during bladder inflammation is substance P dependent. Am J Physiol Renal Physiol 278: F440-F450, 2000[Abstract/Free Full Text].

40.   Hanno, PM, Landis JR, Matthews-Cook Y, Kusek J, and Nyberg L, Jr. The diagnosis of interstitial cystitis revisited:lessons learned from the National Institutes of Health Interstitial Cystitis Database study. J Urol 161: 553-557, 1999[ISI][Medline].

41.   Hill, R. NK1 (substance P) receptor antagonists---why are they not analgesic in humans? Trends Pharmacol Sci 21: 244-246, 2000[ISI][Medline].

42.   Hohenfellner, M, Nunes L, Schmidt RA, Lampela A, Thuroff JW, and Tanagho EA. Interstitial cystitis: increased sympathetic innervation and related neuropeptide synthesis. J Urol 147: 587-591, 1992[ISI][Medline].

43.   Jackson, AM, Ivshina AV, Senko O, Kuznetsova A, Sundan A, O'Donnell MA, Clinton S, Alexandroff AB, Selby PJ, James K, and Kuznetsov VA. Prognosis of intravesical bacillus Calmette-Guerin therapy for superficial bladder cancer by immunological urinary measurements: statistically weighted syndrome analysis. J Urol 159: 1054-1063, 1998[ISI][Medline].

44.   Kamp, EH, Beck DR, and Gebhart GF. Combinations of neurokinin receptor antagonists reduce visceral hyperalgesia. J Pharmacol Exp Ther 299: 105-113, 2001[Abstract/Free Full Text].

45.   Karlsson, JA, Souness J, Webber S, Pollock K, and Raeburn D. Anti-inflammatory effects of the novel phosphodiesterase IV inhibitor RP 73401. Int Arch Allergy Immunol 107: 5-6, 1995.

46.   Krumins, SA, and Broomfield CA. Evidence of NK1 and NK2 tachykinin receptors and their involvement in histamine release in a murine mast cell line. Neuropeptides 21: 65-72, 1992[ISI][Medline].

47.   Laird, JM, Roza C, De Felipe C, Hunt SP, and Cervero F. Role of central and peripheral tachykinin NK1 receptors in capsaicin-induced pain and hyperalgesia. Pain 90: 97-103, 2001[ISI][Medline].

48.   Le Cam, A, Pages G, Auberger P, Le Cam G, Leopold P, Benarous R, and Glaichenhaus N. Study of a growth hormone regulated protein secreted by rat hepatocytes: cDNA cloning,anti-protease activity and regulation of its synthesis by various hormones. EMBO J 6: 1225-1132, 1987[Abstract].

49.   Lee, MLT, Kuo FC, Whitmore GA, and Sklar J. Importance of replication in microarray gene expression studies: statistical methods and evidence from repetitive cDNA hybridizations. PNAS 97: 9834-9839, 2000[Abstract/Free Full Text].

50.   Leverrier, S, Cinato E, Paul C, Derancourt J, Bemark M, Leanderson T, and Legraverend C. Purification and cloning of type A/B hnRNP proteins involved in transcriptional activation from the Rat spi 2 gene GAGA box. Biol Chem 381: 1031-1040, 2000[ISI][Medline].

51.   Lilly, CM, Hall AE, Rodger IW, Kobzik L, Haley KJ, and Drazen JM. Substance P-induced histamine release in tracheally perfused guinea pig lungs. J Appl Physiol 78: 1234-1241, 1995[Abstract/Free Full Text].

52.   Little, SR, and Eisen AN. Preparation of immunogenic 2, 4-dinitrophenyl and 2, 4, 6-trinitrophenyl proteins. Methods Immunol Immunochem 1: 128-133, 1967.

53.   Lorenz, D, Wiesner B, Zipper J, Winkler A, Krause E, Beyermann M, Lindau M, and Bienert M. Mechanism of peptide-induced mast cell degranulation. Translocation and patch-clamp studies. J Gen Physiol 112: 577-591, 1998[Abstract/Free Full Text].

54.   Maass, N, Hojo T, Zhang M, Sager R, Jonat W, and Nagasaki K. Maspin---a novel protease inhibitor with tumor-suppressing activity in breast cancer. Acta Oncologica 39: 931-934, 2000[Medline].

55.   Marchand, JE, Sant GR, and Kream RM. Increased expression of substance P receptor-encoding mRNA in bladder biopsies from patients with interstitial cystitis. Br J Urol 81: 224-228, 1998[ISI][Medline].

56.   Mussap, CJ, Stamatakos C, and Burcher E. Radioligand binding, autoradiographic and functional studies demonstrate tachykinin NK-2 receptors in dog urinary bladder. J Pharmacol Exp Ther 279: 423-434, 1996[Abstract].

57.   Nsa Allogho, S, Nguyen-Le XK, Gobeil F, Pheng LH, and Regoli D. Neurokinin receptors (NK1, NK2) in the mouse: a pharmacological study. Can J Physiol Pharmacol 75: 552-557, 1997[ISI][Medline].

58.   Ogawa, K, Nabe T, Yamamura H, and Kohno S. Nanomolar concentrations of neuropeptides induce histamine release from peritoneal mast cells of a substrain of Wistar rats. Eur J Pharmacol 18: 285-291, 1999.

59.   Okada, T, Hirayama Y, Kishi S, Miyayasu K, Hiroi J, and Fuji T. Functional neurokinin NK-1 receptor expression in rat peritoneal mast cells. Inflamm Res 48: 274-279, 1999[ISI][Medline].

60.   Okamoto, M, Hattori K, and Oyasu R. Interleukin-6 functions as an autocrine growth factor in human bladder carcinoma cell lines in vitro. Int J Cancer 72: 149-154, 1997[ISI][Medline].

61.   Pahl, HL. Activators and target genes of Rel/NF-kappa B transcription factors. Oncogene 18: 6853-6866, 1999[ISI][Medline].

62.   Pang, X, Marchand J, Sant GR, and Theoharides TC. Increased number of SP-positive fibres in interstitial cystitis. Br J Urol 75: 744-750, 1995[ISI][Medline].

63.   Patacchini, R, Giuliani S, Turini A, Navarra G, and Maggi CA. Effect of nepadutant at tachykinin NK(2) receptors in human intestine and urinary bladder. Eur J Pharmacol 398: 389-397, 2000[ISI][Medline].

64.   Peeker, R, Enerback L, Fall M, and Aldenborg F. Recruitment, distribution and phenotypes of mast cells in interstitial cystitis. J Urol 163: 1009-1015, 2000[ISI][Medline].

65.   Quackenbush, J. Computational analysis of microarray data. Nat Rev Genet 2: 418-427, 2001[ISI][Medline].

66.   Rayet, B, and Gelinas C. Aberrant rel/NF-kappa B genes and activity in human cancer. Oncogene 18: 6938-6947, 1999[ISI][Medline].

67.   Reichard, GA, Ball ZT, Aslanian R, Anthes JC, Shih NY, and Piwinski JJ. The design and synthesis of novel NK1/NK2 dual antagonists. Bioorg Med Chem Lett 10: 2329-2332, 2000[ISI][Medline].

68.   Rosamilia, A, Cann L, Dwyer P, Scurry J, and Rogers P. Bladder microvasculature in women with interstitial cystitis. J Urol 161: 1865-1870, 1999[ISI][Medline].

69.   Saban, MR, Hellmich H, Nguyen NB, Winston J, Hammond TG, and Saban R. Time course of LPS-induced gene-expression in a mouse model of genitourinary inflammation. Physiol Genom 5: 147-160, 2001[Abstract/Free Full Text].

70.   Saban, MR, Saban R, Hammond TG, Haak-Frendscho M, Steinberg H, Tengowski MW, and Bjorling DE. LPS-sensory peptide communication in experimental cystitis. Am J Physiol Renal Physiol 282: F202-F210, 2002[Abstract/Free Full Text].

71.   Saban, R, Nielsen KT, Rhodes PR, Christensen MM, and Bruskewitz RC. Effects of mucosa removal and an enkephalinase inhibitor on the contractions of dog isolated bladder strips. J Urol 147: 750-755, 1992[ISI][Medline].

72.   Saban, R, Saban MR, Nguyen NB, Hammond TG, and Wershil BK. Mast cell regulation of inflammation and gene-expression during antigen-induced bladder inflammation in the mouse. Physiol Genom 7: 35-44, 2001[Medline].

73.   Saban, R, Saban MR, Nguyen NB, Lu B, Gerard C, Gerard NP, and Hammond TG. Neurokinin-1 (NK-1) receptor is required in antigen-induced cystitis. Am J Pathol 156: 775-780, 2000[Abstract/Free Full Text].

74.   Schwarzenberg, SJ, Yoon JB, Seelig S, Potter CJ, and Berry SA. Discoordinate hormonal and ontogenetic regulation of four rat serpin genes. Am J Physiol Cell Physiol 262: C1144-C1148, 1992[Abstract/Free Full Text].

75.   Scott, FL, Eyre HJ, Lioumi M, Ragoussis J, Irving JA, Sutherland GA, and Bird PI. Human ovalbumin serpin evolution: phylogenic analysis, gene organization, and identification of new PI8-related genes suggest that two inter-chromosomal and several intra-chromosomal duplications generated the gene clusters at 18q21-q23 and 6p25. Genomics 62: 490-499, 1999[ISI][Medline].

76.   Sherlock, G. Analysis of large-scale gene expression data. Curr Opin Immunol 12: 201-205, 2000[ISI][Medline].

77.   Shih, NY, Albanese M, Anthes JC, Carruthers NI, Grice CA, Lin L, Mangiaracina P, Reichard GA, Schwerdt J, Seidl V, Wong SC, and Piwinski JJ. Synthesis and structure-activity relationships of oxime neurokinin antagonists: discovery of potent arylamides. Bioorg Med Chem Lett 12: 141-145, 2002[ISI][Medline].

78.   Spellman, PT, Sherlock G, Zhang MQ, Iyer VR, Anders K, Eisen MB, Brown PO, Botstein D, and Futcher B. Comprehensive identification of cell cycle-regulated genes of the yeast Saccharomyces cerevisiae by microarray hybridization. Mol Biol Cell 9: 3273-3297, 1998[Abstract/Free Full Text].

79.   Syrigos, KN, Harrington KJ, and Pignatelli M. Role of adhesion molecules in bladder cancer: an important part of the jigsaw. Urology 53: 428-434, 1999[ISI][Medline].

80.   Tamayo, P, Slonim D, Mesirov J, Zhu Q, Kitareewan S, Dmitrovsky E, Lander ES, and Golub TR. Interpreting patterns of gene expression with self-organizing maps: methods and application to hematopoietic differentiation. PNAS 96: 2907-2912, 1999[Abstract/Free Full Text].

81.   Theoharides, TC, Pang X, Letourneau R, and Sant GR. Interstitial cystitis: a neuroimmunoendocrine disorder. Ann NY Acad Sci 840: 619-634, 1998[Abstract/Free Full Text].

82.   Toronen, P, Kolehmainen M, Wong G, and Castren E. Analysis of gene expression data using self-organizing maps. FEBS Lett 451: 142-146, 1999[ISI][Medline].

83.   Tramontana, M, Patacchini R, Lecci A, Giuliani S, and Maggi CA. Tachykinin NK2 receptors in the hamster urinary bladder: in vitro and in vivo characterization. Naunyn-Schmiedebergs Arch Pharmacol 358: 293-300, 1998[ISI][Medline].

84.   Urban, LA, and Fox AJ. NK1 receptor antagonists---are they really without effect in the pain clinic? Trends Pharmacol Sci 21: 462-464, 2000[ISI][Medline].

85.   Van Giersbergen, PLM, and Buck SH. Characterization of tachykinin receptors. In: The Tachykinin Receptors, edited by Buck SH.. Totowa, NJ: Humana, 1994, p. 39-68.

86.   Vizzard, MA. Alterations in neuropeptide expression in lumbosacral bladder pathways following chronic cystitis. J Chem Neuroanat 21: 125-138, 2001[ISI][Medline].

87.   Vizzard, MA. Up-regulation of pituitary adenylate cyclase-activating polypeptide in urinary bladder pathways after chronic cystitis. J Comp Neurol 420: 335-348, 2000[ISI][Medline].

88.   Wall, BM, Dmochowski RR, Malecha M, Mangold T, Bobal MA, and Cooke CR. Inducible nitric oxide synthase in the bladder of spinal cord injured patients with a chronic indwelling urinary catheter. J Urol 165: 1457-1461, 2001[ISI][Medline].

89.   Wang, XC, Saban R, Kaysen JH, Saban MR, Allen PL, Benes EN, and Hammond TG. Nuclear factor kappa B mediates lipopolysaccharide-induced inflammation in the urinary bladder. J Urol 163: 993-998, 2000[ISI][Medline].

90.   Watson, S, and Arkinstall S. The G Protein-Linked Receptor, edited by Watson S, and Arkinstall S.. New York: Academic, 1994, p. 261-271.

91.   Wen, X, Fuhrman S, Michaels GS, Carr DB, Smith S, Barker JL, and Somogyi R. Large-scale temporal gene expression mapping of central nervous system development. PNAS 95: 334-339, 1998[Abstract/Free Full Text].

92.   Wershil, B. Mast cell-deficient mice and intestinal biology. Am J Physiol Gastrointest Liver Physiol 278: G343-G348, 2000[Abstract/Free Full Text].

93.   Yano, H, Wershil BK, Arizono N, and Galli SJ. Substance P-induced augmentation of cutaneous vascular permeability and granulocyte infiltration in mice is mast cell dependent. J Clin Invest 84: 1276-1286, 1989[ISI][Medline].

94.   Yoon, JB, Towle HC, and Seelig S. Growth hormone induces two mRNA species of the serine protease inhibitor gene family in rat liver. J Biol Chem 262: 4284-4289, 1987[Abstract/Free Full Text].

95.   Zeng, XP, and Burcher E. Tachykinin NK-2 receptors in child urinary bladder. J Urol 155: 1104-1107, 1996[ISI][Medline].

96.   Zhang, M, Volpert O, Shi YH, and Bouck N. Maspin is an angiogenesis inhibitor. Nat Med 6: 196-199, 2000[ISI][Medline].


Am J Physiol Renal Fluid Electrolyte Physiol 283(4):F616-F629
0363-6127/02 $5.00 Copyright © 2002 the American Physiological Society