Skip to main content
Erschienen in: BMC Gastroenterology 1/2003

Open Access 01.12.2003 | Research article

Differential gene expression in the murine gastric fundus lacking interstitial cells of Cajal

verfasst von: Yataro Daigo, Ichiro Takayama, Bruce AJ Ponder, Carlos Caldas, Sean M Ward, Kenton M Sanders, Masayuki A Fujino

Erschienen in: BMC Gastroenterology | Ausgabe 1/2003

Abstract

Background

The muscle layers of murine gastric fundus have no interstitial cells of Cajal at the level of the myenteric plexus and only possess intramuscular interstitial cells and this tissue does not generate electric slow waves. The absence of intramuscular interstitial cells in W/W V mutants provides a unique opportunity to study the molecular changes that are associated with the loss of these intercalating cells.

Method

The gene expression profile of the gastric fundus of wild type and W/W V mice was assayed by murine microarray analysis displaying a total of 8734 elements. Queried genes from the microarray analysis were confirmed by semi-quantitative reverse transcription-polymerase chain reaction.

Results

Twenty-one genes were differentially expressed in wild type and W/W V mice. Eleven transcripts had 2.0–2.5 fold higher mRNA expression in W/W V gastric fundus when compared to wild type tissues. Ten transcripts had 2.1–3.9 fold lower expression in W/W V mutants in comparison with wild type animals. None of these genes have ever been implicated in any bowel motility function.

Conclusions

These data provides evidence that several important genes have significantly changed in the murine fundus of W/W V mutants that lack intramuscular interstitial cells of Cajal and have reduced enteric motor neurotransmission.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-230X-3-14) contains supplementary material, which is available to authorized users.
Yataro Daigo, Ichiro Takayama contributed equally to this work.

Competing interests

None declared.

Authors' contributions

YD and IT carried out the molecular studies and the design of study and coordination. BAJP and CC participated in the molecular studies. SW, KS and MF participated in the design of study and coordination. All authors read and approved the final manuscript.
Abkürzungen
BP1/6C3
glutamyl aminopeptidase
BST1/BP3
bone marrow stromal antigen 1(alias ADP-ribosylcyclase 2 precursor)
CPO
coproporphyrinogen oxidase
DMP
deep muscular plexus
ESTs
expressed sequence tags
FISH
fluorescence in situ hybridization
GI
gastrointestinal
ICC
interstitial cells of Cajal
IC-DMP
ICC at the level of the deep muscular plexus
IC-IM
intramuscular ICC
IC-MY
ICC at the level of the myenteric plexus
MCM7
minichromosome maintenance 7
MY
myenteric plexus
p160 ROCK2
p160 Rho-associated, coiled-coil forming protein kinase 2
RBP2
retinol binding protein 2, cellular
RHAMM
hyaluronan-mediated motility receptor
RT-PCR
reverse transcription-polymerase chain reaction
SNP
single nucleotide polymorphism

Background

Interstitial cells of Cajal (ICC) are gastrointestinal (GI) pacemaker cells and intermediaries in enteric motor neurotransmission in the GI tract [1, 2]. ICC express KIT/c-kit and depend on signalling via the gene product protein, KIT, a receptor tyrosine kinase which is essential for development and maintenance of the ICC phenotype [3]. Mutations in the white-spotting locus (i.e. W/W V ) result in reduced KIT expression. These mutant animals develop few ICC at the level of the myenteric plexus (IC-MY) in the small intestine and the reduced ICC numbers is associated with a loss of slow wave activity. Intramuscular ICC (IC-IM) located in the stomach, lower esophageal and pyloric sphincters are absent in W/W V mutant animals [4]. Reduced numbers of ICC have also been reported in several GI motility disorders, such as chronic intestinal pseudo-obstruction [5, 6], infantile hypertrophic pyloric stenosis [79], Hirschsprung's disease [1012], slow-transit constipation and certain forms of gastroparesis [13, 14].
The association between motility disorders and loss of specific populations of ICC suggests that a more complete understanding of the molecular and cell biology of ICC networks within the gastrointestinal tract may help in understanding the etiology of some GI motor pathologies. The aim of the present study was to characterize genetic sequences that are expressed in ICC of the stomach that may encode important functional elements of the GI pacemaker/motor neurotransmission system. We pursued the hypothesis that such genes might show differential expression in the small intestines of wild type mice and W/W V mice [15, 16]. We previously identified fifteen known and novel genes that were differentially expressed in the small intestines of wild type and W/W V mice, which develop few IC-MY using a differential gene expression method [17, 18].
In the present study we hypothesized that there may also be differential expression of genes in the gastric fundus of W/W V mice, where IC-IM are lost. Our gene microarray analysis successfully identified 21 genes that were differentially expressed in the fundus of W/W V mice. The differential expression of these mice was confirmed by semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR).

Methods

Animals and Tissue Preparation

The use and treatment of experimental animals was approved by the Guideline governing Animal Experiment Committee at the University of Yamanashi School of Medicine and at the University of Nevada School of Medicine. Six adult male WBB6F1-+/+ mice (wild type) and age matched six adult male WBB6F1-W/W V mice, weighing 20 to 30 g, were purchased from Japan SLC Inc (Shizuoka, Japan). They were anesthetized with ether or carbon dioxide inhalation and sacrificed by cervical dislocation. For gene analyses, stomachs were removed from the animal and the mucosa with the attached submucosa rapidly sharp dissected from the tunica muscularis. Tissues were subsequently frozen in liquid nitrogen (-196 °C). Tissues were stored at -80 °C until isolation of RNA was preformed [18].

RNA Preparation and Microarray Data Analysis

For gene microarray analysis, poly (A)+ RNA was isolated from each tissue sample by TRIZOL (Life Technologies, Inc., Gaithersburg, MD, USA) and Poly (A)+Isolation Kit from Total RNA (ISOGEN, Nippon Gene, Tokyo, Japan).18 Before starting microarray analysis, differential gene expression of KIT between the gastric fundic tissues of wild type mice and those of W/W V mutant mice were confirmed by semi-quantitative RT-PCR (See the section of Semi-quantitative RT-PCR) (Fig. 1A). Microarrays were hybridized and scanned, and image analysis was performed as described previously [19, 20]. In brief, alterations in gene expression were evaluated by reverse transcription of poly (A)+ RNAs in the presence of Cy3 or Cy5 fluorochromes followed by hybridization to mouse GEM 1 microarray chips (Incyte Genomics, Inc., Porter Drive Palo Alto, CA, USA). To confirm the validity of the assay, the experiments were performed in duplicate. The 8734 cDNAs we used represent 7854 unique genes/UniGene clusters: 3205 are annotated and 4649 are unannotated sequences. We assigned a cut-off value, using a variance analysis, to the microarray slide. Genes whose Cy3- and Cy5-signal intensities were lower than the cut-off values were excluded from further investigation. Balanced differential expression was determined for control wild type mice versus W/W V mutant mice. Values that were < -2.0 or > 2.0 in both two independent experiments were considered significant and the mean values of the relative fluorescence ratio for each gene were calculated.

Semi-quantitative RT-PCR

The levels of mRNA expression that were decreased or increased by > 2.0-fold or greater, were confirmed by semi-quantitative RT-PCR from fundus of 6 of each wild type mice and W/W V mice. RT-PCR was performed as described previously [18]. In brief, 2 μg aliquots of total RNAs treated with DNase I (Roche Diagnostics, Basel, Switzerland) from the mice fundic tissues were used to synthesize single-strand cDNAs. These cDNAs were used as templates for PCR in a thermal cycler (PerkinElmer, Shelton, CT, USA), using primers listed in Table 1 or KIT (5'-ATG ACG TCA TGA AGA CTT GCT-3' and 5'-CTA CCC TGG AAT AGG ATG CA-3'), and GAPDH specific primer sets (5'-GACAACAGCCTCAAGATCATCA-3' and 5'-GGTCCACCACTGACACTGTG-3'). Expression of GAPDH served as an internal control. Primers were derived from different exons in the same gene, when the corresponding mouse genomic sequence was available at that time. The PCR reactions were optimized for number of cycles to ensure product intensity within the linear phase of amplification.
Table 1
Primer sets used for semi-quantitative RT-PCR.
Gene Name
Accession No.
Forward Primer
Reverse Primer
EST
AA185701
CGA AAG CCT TGA GGT TGA AG
TAG GAA AAC AGG CGT CAC TG
EST
AA166336
GAA GAA AAG GCT GCA GAT CG
CAA GAG GCA AAG AGC AAT CC
EST
AA021806
CAC GAA TTG CAG GAC TAC CT
ACC TGC ACT GTA GGC TGA GT
MCM7
Q61881
GCC CAC TGG ATT GTG AAG AT
AGG AGA CTG GTC CAC ACC AC
p160 ROCK2
U58513
AAG AAC CTG TCA AGC GTG GT
TCC AGG GTC ATC TGG AGT TC
EST
W18585
AGA CTT GGT GGC AGA GGA GA
GCA GCT CAT GAC AGA ACA CC
EST
AA403748
CCT AAA GCA ACC CAA CCT GA
TAG CCT TAT GGG ACC TGG TG
BST1/BP3
Q64277
GAT TTC TTG AGC TGG TGT CG
AAA ACC CTC TCG TGG GAT AG
EST
AA024250
CAG ATA GAG CAA GGG ATG GA
CTG AGC CCA AAC CAG TAG AA
RBP2
Q08652
GAC GAA GGA CCA AAA TGG AA
CGG TGA AAT CCA GGT CGT AG
EST
W46016
AGC AAC AAC AGC TGG ACT TC
ACC TTC TGT TTG GTG CTG AG
BP1/6C3
S30398
TAT CGG CCT CAT CTA ACC AG
ATC TTC AAG CAG CAC CTG AC
EST
AI552444
CAT GCG ATA CTG GAA CAT GA
TGA CTC CAA ATA GCC CTC AG
EST
AA108876
TGG AGC AAG AGA GGA AAG TG
CTA GAC CTG AGC TTG CCT TG
EST
AA049294
ACG TGG AGA AAG TTC TCG TG
GCA ATA GTG TCA CCG AAT CC
EST
AA254777
GCC CTG GAG TTG AGA CTG TA
TGA CAA GCT GCA CAG TAA CC
RHAMM
AF031932 GCA GAA GGA GGA GCA GAG TG
GCA GTG ACG TCC CTC AGA CT
EST
AA002293
AAG TCT TTG TGT GGG CTG AG
AGG AAG CTT CGT CTC TCC AT
EST
AI595081
CAC CAG GAT GTC TGC CTA CT
CCG AGA TCA TGT TCT TCA CC
CPO
D16333
GAA GAC CAA GAT GGA GCT GA
CAG AAA GAT TCC CAT GAA CG
EST
AA000304
TTC ATC TGC TGC TCC TTC TC
TTA TAG ACC TTC CCG CAC AG

Results

Microarray Analysis and Semi-quantitative RT-PCR

To identify genes that may be selectively associated with IC-IM, we compared gene expression patterns in gastric fundic tissues derived from wild type and W/W V mice utilizing a gene microarray method [19, 20]. Murine fundus without epithelial tissues derived from wild type and W/W V mice maintained under fasted conditions were used to generate poly (A)+ mRNA for microarray analysis using the mouse GEM 1 microarray. Results of this experiment, highlighting genes that were reproducibly decreased or increased by > 2.0-fold or greater, are shown in Table 2 and Fig. 1B. Several known and novel genes were differentially expressed in the fundus of W/W V mice ( > 2.0 balanced differential expression). To confirm the differential expression profiles, we further examined the expression levels in murine fundic mRNA derived from wild types and W/W V mice maintained under fasted conditions as templates, using semi-quantitative RT-PCR analysis (Representative data were shown in Fig. 1C,1D. All primer sets used for the confirmation were listed in Table 1). Expression of ten genes was considerably reduced in the gastric fundus of W/W V mice compared to age matched wild type mice. Another eleven genes showed an increase in expression in fasted W/W V mice.
Table 2
Analysis of gene expression in the fundus of wild type and W/W V mice
Gene Name
W/W V /wildtype ratio(a)
Accession No.
Subcellular location(b)
Cytoband(c)
EST
2.5
AA185701
  
EST
2.5
AA166336
  
EST
2.4
AA021806
  
MCM7: DNA Replication Licensing Factor
2.4
Q61881
nucleus
7q21.3-q22.1
p160 ROCK2: Rho-Associated Protein Kinase
2.3
U58513
cytoskeleton
2p24
EST
2.3
W18585
  
EST
2.3
AA403748
  
BST1/BP3: ADP-Ribosyl Cyclase 2 Precursor
2.0
Q64277
membrane
4p15
EST
2.0
AA024250
  
RBP2: retinol binding protein 2, cellular
2.0
Q08652
cytoplasm
3q23
EST
2.0
W46016
  
BP1/6C3: Glutamyl Aminopeptitase
-2.1
S30398
membrane
16
EST
-2.2
AI552444
  
EST
-2.3
AA108876
  
EST
-2.4
AA049294
  
EST
-2.4
AA254777
  
RHAMM: Hyaluronan-Mediated Motility Receptor
-2.5
AF031932
membrane
5q33.2-qter
EST
-2.5
AA002293
  
EST
-3.4
AI595081
  
CPO: Coproporphyrinogen Oxidase
-3.6
D16333
mitochondria
3q12
EST
-3.9
AA000304
  
(a) Balanced differential expression for control wild type mice vs W/W V mutant mice. Values that were decreased or increased by > 2-fold or greater in both two independent experiments were considered significant and the mean values were calculated. All of the results were confirmed with semi-quantitative RT-PCR using fundic tissues from six wild type and six W/W V mutant mice. (b) Subcellular location determined by the SOURCE program http://​source.​stanford.​edu. (c) Human chromosomal localization of the gene.

Discussion

A comparison of differentially expressed genes from the fundus of W/W V mice using DNA microarray analysis revealed that the expression of eleven genes transcripts were significantly up-regulated in W/W V mice, whereas ten genes transcripts were dramatically suppressed in these animals. We confirmed these results with semi-quantitative RT-PCR of tissues from six each of wild type and W/W V mice. Data obtained from these experiments suggest that expression of the genes were specifically regulated in W/W V mice.
The eleven genes that were up-regulated in the gastric fundus of W/W V mice were identified as MCM7, p160 ROCK2, BST1/BP3, RBP2, and another seven unannotated transcripts. MCM7 is a mammalian homologue of the yeast nuclear protein MCM2/CDC47, which is thought to play an important role in two crucial steps of the cell cycle, namely, onset of DNA replication and cell division [21, 22]. p160 ROCK2, which is an isozyme of ROCK1 is a target for the small GTPase, Rho [23]. ROCK2 is a serine/threonine kinase that regulates cytokinesis, smooth muscle contraction, the formation of actin stress fibers and focal adhesions, and the activation of the FOS serum response element [24]. The up-regulation of p160 ROCK2 may have a compensating effect for the loss of ICC-dependent mechanisms in the gastric fundus. The BST1/BP3, a bone marrow stromal cell surface antigen, is a variably glycosylated glycosyl-phosphatidylinositol (GPI)-linked molecule that is selectively expressed by early B and T lineage cells and a discrete subpopulation of reticular cells in the peripheral lymphoid organs. It is also expressed on the brush border of intestinal epithelial cells, the luminal surface of renal collecting tubules and mature myeloid cells [25]. This protein is supposed to belong to ADP-ribosyl cyclase family [26]. Cellular RBP2 is an abundant 134-residue protein present in the small intestinal epithelium [27]. It is thought to participate in the uptake and/or intracellular metabolism of vitamin A and belong to a protein family that contains liver fatty acid-binding protein. Vitamin A is a fat-soluble vitamin necessary for growth, reproduction, differentiation of epithelial tissues, and vision. Mammals depend on intestinal absorption of this vitamin for their survival. RBP2, which is confined largely to the small intestinal enterocyte, probably plays an important role in the intestinal absorption and/or metabolism of vitamin A [27].
We also confirmed the down-regulation of ten genes in W/W V mice: BP1/6C3, RHAMM, CPO, and another seven unannotated ESTs. The murine β-lymphocyte differentiation antigen, BP1/6C3 was characterized as glutamyl aminopeptidase, which is reported to serve as cell-differentiation marker of lymphomyelocytic lineages and may be involved in cell activation, signal transduction, and cell-matrix adhesion. It is also expressed by capillary endothelial cells, placenta, and epithelial cells of the intestine and proximal renal tubules [28]. RHAMM encodes a hyaluronan receptor protein [29]. When hyaluronan binds to RHAMM, the phosphorylation of a number of proteins including the focal adhesion kinase pp125-FAK occurs [30]. This is a necessary step for disassembly of focal contacts and subsequent motility. CPO is the sixth enzyme of the heme biosynthetic pathway. This soluble protein is localized in the intermembrane space of mitochondria and catalyzes the conversion of two propionate groups at positions two and four of coproporphyrinogen III to two vinyl groups of protoporphyrinogen IX [31]. It was reported that coproporphyria (CPO deficiency) patients showed constipation and abnormal colic that are main symptoms of this disease [32]. Marked elevation of coproporphyria in the feces differentiated the condition from the Swedish type in which stool porphyrins are usually normal and from variegate porphyria in which both coproporphyrin and protoporphyrinogen fractions are increased in the stool [33].
In the 21 genes identified, we determined the subcellular localization and human chromosomal mapping of the 7 known genes using the SOURCE program http://​source.​stanford.​edu (Table 2). These genes showed a variety of cellular localization including 3 membrane, 2 cytoplasmic, 1 mitochondrial and 1 nuclear proteins. Further analyses of these genes might enable us to elucidate not only their relationship with the KIT, a receptor tyrosine kinase, but also the molecular aspects of GI pacemaker system.
We previously identified fifteen genes that were differentially expressed in the small intestines of wild type and W/W V mice which develop few IC-MY using a differential gene expression method [17, 18].™@None of these 15 genes were found in the list of 21 genes that were differentially expressed in the gastric fundus of wild type and W/W V mice which develop few IC-IM using a cDNA microarray. As we confirmed differential gene expression of KIT between the small intestines/gastric fundic tissues of wild type and those of W/W V mutant mice by semi-quantitative RT-PCR, our experimental system could detect genetic aberration in W/W V mice. Therefore our results might reflect the difference of the cellular entity of two types of ICCs, IC-MY and IC-IM, or the difference of the cellular composition between small intestine and fundus. To provide conclusive evidence, which support these speculations, expression profile using purified mRNA from isolated single interstitial cells might be very useful in the next study.
At the present time we do not know whether these genes are important to the function of the gastric pacemaker/neurotransmission apparatus or were down- or up-regulated as a result of the loss of ICC. These data, however, provide clear systemic genetic evidence that several important proteins that have roles in cell cycle, cytokinesis and formation of cytoskeleton, cellular metabolism, oxygen metabolism, cell adhesion, and development and differentiation of gut cells are significantly changed in the gastric fundus of W/W V mice. Generation of transgenic animals that show tissue-specific overexpression of the candidate genes, as well as the gene knockout animals might be helpful for elucidating the role of each gene in gastrointestinal motility.
The discovery of an entire human and mouse genes through the genome project is supposed to revolutionize biological medicine including molecular diagnosis of various diseases and development of novel treatment. The information combined with high throughput technology such as DNA microarray and SNP typing analysis will accelerate discovery of genes susceptible to or causing various diseases and contribute to screening of novel drugs that target these disease-gene products. In this sense, the effort of the molecular profiling project in which we attempt to discover genetic aberration in animal disease models such as W/W V mice will generate very variable resources for further elucidation of motility disorders. As reduced numbers of ICC have also been reported in several GI motility disorders, such as chronic intestinal pseudo-obstruction [5, 6], slow-transit constipation and certain forms of gastroparesis [13, 14], these gene information should aid the development of novel molecular-targeted therapies for these disorders, and may also identify diagnostic molecular markers for these disorders.

Conclusion

We have identified twenty-one genes that encode functional proteins that are significantly up- or down-regulated in the tunica muscularis of the gastric fundus of W/W V mice. Considering that none of these genes has been implicated in any aspect of GI motility, we suggest that many unknown genes could be involved in the cellular changes that lead to motility disorders associated with the loss of ICC. Gene microarray analysis is an effective method for screening the changes that occur in the expression patterns of genes in response to spontaneous or genetic mutations that lead to the loss of a specific cell type. Application of this technique may allow recognition of patterns of gene expression that are common to the several motility disorders in which ICC are lost, thus providing new insights into the molecular mechanisms responsible for the selective loss of ICC populations.

Acknowledgements

Supported by 08457165 (to MF) from the Japanese Ministry of Education, Science, Sports and Culture, JAPAN and DK 57236 (to SW) and PO1 DK41315 (to SW and KS) from the National Institutes of Health, USA.

Competing interests

None declared.

Authors' contributions

YD and IT carried out the molecular studies and the design of study and coordination. BAJP and CC participated in the molecular studies. SW, KS and MF participated in the design of study and coordination. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
1.
Zurück zum Zitat Ward SM, Burns AJ, Torihashi S, Sanders KM: Mutation of the proto-oncogene c-kit blocks development of interstitial cells and electrical rhythmicity in murine intestine. J Physiol (Lond). 1994, 480: 91-97.CrossRef Ward SM, Burns AJ, Torihashi S, Sanders KM: Mutation of the proto-oncogene c-kit blocks development of interstitial cells and electrical rhythmicity in murine intestine. J Physiol (Lond). 1994, 480: 91-97.CrossRef
2.
Zurück zum Zitat Burns AJ, Herbert TM, Ward SM, Sanders KM: Interstitial cells of Cajal inhibit neurotransmission in the stomach. Proc Natl Acad Sci USA. 1996, 93: 12008-12013. 10.1073/pnas.93.21.12008.CrossRefPubMedPubMedCentral Burns AJ, Herbert TM, Ward SM, Sanders KM: Interstitial cells of Cajal inhibit neurotransmission in the stomach. Proc Natl Acad Sci USA. 1996, 93: 12008-12013. 10.1073/pnas.93.21.12008.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Huizinga JD, Thuneberg L, Kluppel M, Malysz J, Mikkelesen HB, Bernstein A: W/kit gene required for interstitial cells of Cajal and for intestinal pacemaker activity. Nature (Lond). 1995, 373: 347-349. 10.1038/373347a0.CrossRef Huizinga JD, Thuneberg L, Kluppel M, Malysz J, Mikkelesen HB, Bernstein A: W/kit gene required for interstitial cells of Cajal and for intestinal pacemaker activity. Nature (Lond). 1995, 373: 347-349. 10.1038/373347a0.CrossRef
4.
Zurück zum Zitat Ward SM, Beckett EAH, Wang XY, Baker F, Khoyi M, Sanders KM: Interstitial cells of Cajal mediate enteric excitatory neurotransmission in the murine fundus. J Neurosci. 2000, 20: 1393-1403.PubMed Ward SM, Beckett EAH, Wang XY, Baker F, Khoyi M, Sanders KM: Interstitial cells of Cajal mediate enteric excitatory neurotransmission in the murine fundus. J Neurosci. 2000, 20: 1393-1403.PubMed
5.
Zurück zum Zitat Takayama I, Kojima Y, Ohtsuka H, Sato T, Fujino MA: Pathology of human idiopathic intestinal pseudo-obstruction and W/W v mice in gut motility: A gut pacemaker disease?. Gut. 1995, 37 (Suppl): A85- Takayama I, Kojima Y, Ohtsuka H, Sato T, Fujino MA: Pathology of human idiopathic intestinal pseudo-obstruction and W/W v mice in gut motility: A gut pacemaker disease?. Gut. 1995, 37 (Suppl): A85-
6.
Zurück zum Zitat Isozaki K, Hirota S, Miyagawa J, Taniguchi M, Shiomura Y, Matsuzawa Y: Deficiency of c-kit+ cells in patients with a myopathic form of chronic idiopathic intestinal pseudo-obstruction. Am J Gastroenterol. 1997, 92: 332-334.PubMed Isozaki K, Hirota S, Miyagawa J, Taniguchi M, Shiomura Y, Matsuzawa Y: Deficiency of c-kit+ cells in patients with a myopathic form of chronic idiopathic intestinal pseudo-obstruction. Am J Gastroenterol. 1997, 92: 332-334.PubMed
7.
Zurück zum Zitat Vanderwinden JM, Liu H, Menu R, Conreur JL, De Laet MH, Vanderhagen JJ: The pathology of infantile hypertrophic pyloric stenosis after healing. J Pediatr Surg. 1996, 31: 1530-1534.CrossRefPubMed Vanderwinden JM, Liu H, Menu R, Conreur JL, De Laet MH, Vanderhagen JJ: The pathology of infantile hypertrophic pyloric stenosis after healing. J Pediatr Surg. 1996, 31: 1530-1534.CrossRefPubMed
8.
Zurück zum Zitat Vanderwinden JM, Liu H, DeLaet MH, Vanderhagen JJ: Study of the interstitial cells of Cajal in infantile hypertrophic pyloric stenosis. Gastroenterol. 1996, 111: 279-288.CrossRef Vanderwinden JM, Liu H, DeLaet MH, Vanderhagen JJ: Study of the interstitial cells of Cajal in infantile hypertrophic pyloric stenosis. Gastroenterol. 1996, 111: 279-288.CrossRef
9.
Zurück zum Zitat Yamataka A, Fujiwara T, Kato Y, Okazaki T, Sunagawa M, Miyano T: Lack of intestinal pacemaker (C-KIT-positive) cells in infantile hypertrophic pyloric stenosis. J Pediatr Surg. 1996, 31: 96-98.CrossRefPubMed Yamataka A, Fujiwara T, Kato Y, Okazaki T, Sunagawa M, Miyano T: Lack of intestinal pacemaker (C-KIT-positive) cells in infantile hypertrophic pyloric stenosis. J Pediatr Surg. 1996, 31: 96-98.CrossRefPubMed
10.
Zurück zum Zitat Yamataka A, Kato Y, Tibboel D: A lack of intestinal pacemaker (c-kit) in aganglionic bowel of patients with Hirschsprung's disease. J Pediatr Surg. 1995, 30: 441-444.CrossRefPubMed Yamataka A, Kato Y, Tibboel D: A lack of intestinal pacemaker (c-kit) in aganglionic bowel of patients with Hirschsprung's disease. J Pediatr Surg. 1995, 30: 441-444.CrossRefPubMed
11.
Zurück zum Zitat Yamataka A, Ohshiro K, Kobayashi H, Fujiwara T, Sunagawa M, Miyano T: Interstitial C-KIT+ cells and synapse in allied Hirschsprung's disorders. J Pediatr Surg. 1997, 30: 1069-1074.CrossRef Yamataka A, Ohshiro K, Kobayashi H, Fujiwara T, Sunagawa M, Miyano T: Interstitial C-KIT+ cells and synapse in allied Hirschsprung's disorders. J Pediatr Surg. 1997, 30: 1069-1074.CrossRef
12.
Zurück zum Zitat Vanderwinden JM, Liu H, DeLaet NH, Vanderhagen JJ: Interstitial cells of Cajal in human colon and in Hirschsprung's disease. Gastroenterol. 1996, 111: 901-910.CrossRef Vanderwinden JM, Liu H, DeLaet NH, Vanderhagen JJ: Interstitial cells of Cajal in human colon and in Hirschsprung's disease. Gastroenterol. 1996, 111: 901-910.CrossRef
13.
Zurück zum Zitat He CL, Burgart L, Wang L, Pemberton J, Young-Fadok L, Szurszewski J, Farrugia G: Decreased interstitial cell of Cajal volume in patients with slow-transit constipation. Gastroenterol. 2000, 118: 14-21.CrossRef He CL, Burgart L, Wang L, Pemberton J, Young-Fadok L, Szurszewski J, Farrugia G: Decreased interstitial cell of Cajal volume in patients with slow-transit constipation. Gastroenterol. 2000, 118: 14-21.CrossRef
14.
Zurück zum Zitat Ordog T, Takayama I, Cheung WKT, Ward SM, Sanders KM: Remodeling of networks of interstitial cells of Cajal in a murine model of diabetic gastroparesis. Diabetes. 2001, 49: 1731-1739.CrossRef Ordog T, Takayama I, Cheung WKT, Ward SM, Sanders KM: Remodeling of networks of interstitial cells of Cajal in a murine model of diabetic gastroparesis. Diabetes. 2001, 49: 1731-1739.CrossRef
15.
Zurück zum Zitat Takayama I, Daigo Y, Kojima Y, Fujino MA: Gastrointestinal pacemaker system. Nippon Shoukakibyo Gakkai Zasshi. 2001, 98: 922-934. Takayama I, Daigo Y, Kojima Y, Fujino MA: Gastrointestinal pacemaker system. Nippon Shoukakibyo Gakkai Zasshi. 2001, 98: 922-934.
16.
Zurück zum Zitat Takayama I, Horiguchi K, Daigo Y, Mine T, Fujino MA, Ohno S: The interstitial cells of Cajal and a gastrointestinal pacemaker system. Arch Histol Cytol. 2002, 65: 1-26.CrossRefPubMed Takayama I, Horiguchi K, Daigo Y, Mine T, Fujino MA, Ohno S: The interstitial cells of Cajal and a gastrointestinal pacemaker system. Arch Histol Cytol. 2002, 65: 1-26.CrossRefPubMed
17.
Zurück zum Zitat Takayama I, Daigo Y, Ward SM, Sanders KM, Yamanaka T, Fujino MA: Differential gene expression in the small intestines of wildtype and W/W V mice. Neurogastroenterol Motil. 2001, 13: 163-168. 10.1046/j.1365-2982.2001.00256.x.CrossRefPubMed Takayama I, Daigo Y, Ward SM, Sanders KM, Yamanaka T, Fujino MA: Differential gene expression in the small intestines of wildtype and W/W V mice. Neurogastroenterol Motil. 2001, 13: 163-168. 10.1046/j.1365-2982.2001.00256.x.CrossRefPubMed
18.
Zurück zum Zitat Takayama I, Daigo Y, Ward SM, Sanders KM, Walker RL, Horowitz B, Yamanaka T, Fujino MA: Novel human and mouse genes encoding an acid phosphatase family member and its down regulation in W/W V mouse jejunum. Gut. 2002, 50: 790-796. 10.1136/gut.50.6.790.CrossRefPubMedPubMedCentral Takayama I, Daigo Y, Ward SM, Sanders KM, Walker RL, Horowitz B, Yamanaka T, Fujino MA: Novel human and mouse genes encoding an acid phosphatase family member and its down regulation in W/W V mouse jejunum. Gut. 2002, 50: 790-796. 10.1136/gut.50.6.790.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Liang P, Pardee AB: Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science. 1992, 257: 967-971.CrossRefPubMed Liang P, Pardee AB: Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science. 1992, 257: 967-971.CrossRefPubMed
20.
Zurück zum Zitat Ljubimova JY, Khazenzon NM, Chen Z, Neyman YI, Turner L, Riedinger MS, Black KL: Gene expression abnormalities in human glial tumors identified by gene array. Int J Oncol. 2001, 18: 287-295.PubMed Ljubimova JY, Khazenzon NM, Chen Z, Neyman YI, Turner L, Riedinger MS, Black KL: Gene expression abnormalities in human glial tumors identified by gene array. Int J Oncol. 2001, 18: 287-295.PubMed
21.
Zurück zum Zitat Nakatsuru S, Sudo K, Nakamura Y: Isolation and mapping of a human gene (MCM2) encoding a product homologous to yeast proteins involved in DNA replication. Cell Genet. 1995, 68: 226-230.CrossRef Nakatsuru S, Sudo K, Nakamura Y: Isolation and mapping of a human gene (MCM2) encoding a product homologous to yeast proteins involved in DNA replication. Cell Genet. 1995, 68: 226-230.CrossRef
22.
Zurück zum Zitat Labib K, Tercero JA, Diffley JFX: DNA replication fork progression requires uninterrupted MCM2-7 function. Science. 2000, 288: 1643-1647. 10.1126/science.288.5471.1643.CrossRefPubMed Labib K, Tercero JA, Diffley JFX: DNA replication fork progression requires uninterrupted MCM2-7 function. Science. 2000, 288: 1643-1647. 10.1126/science.288.5471.1643.CrossRefPubMed
23.
Zurück zum Zitat Takahashi N, Tuiki H, Saya H, Kaibuchi K: Localization of the gene coding for ROCK II/Rho kinase on human chromosome 2p24. Genomics. 1999, 55: 235-237. 10.1006/geno.1998.5344.CrossRefPubMed Takahashi N, Tuiki H, Saya H, Kaibuchi K: Localization of the gene coding for ROCK II/Rho kinase on human chromosome 2p24. Genomics. 1999, 55: 235-237. 10.1006/geno.1998.5344.CrossRefPubMed
24.
Zurück zum Zitat Maekawa M, Ishizaki T, Boku S, Watanabe N, Fujita A, Iwamatsu A, Obinata T, Ohashi K, Mizuno K, Narumiya S: Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science. 1999, 285: 895-898. 10.1126/science.285.5429.895.CrossRefPubMed Maekawa M, Ishizaki T, Boku S, Watanabe N, Fujita A, Iwamatsu A, Obinata T, Ohashi K, Mizuno K, Narumiya S: Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science. 1999, 285: 895-898. 10.1126/science.285.5429.895.CrossRefPubMed
25.
Zurück zum Zitat Dong C, Wang J, Neame P, Cooper MD: The murine BP-3 gene encodes a relative of the CD38/NAD glycohydrolase family. Int Immunol. 1994, 6: 1353-1360.CrossRefPubMed Dong C, Wang J, Neame P, Cooper MD: The murine BP-3 gene encodes a relative of the CD38/NAD glycohydrolase family. Int Immunol. 1994, 6: 1353-1360.CrossRefPubMed
26.
Zurück zum Zitat Itoh M, Ishihara K, Tomizawa H, Tanaka H, Kobune Y, Ishikawa J, Kaisho T, Hirano T: Molecular cloning of murine BST-1 having homology with CD38 and Aplysia ADP-ribosyl cyclase. Biochem Biophys Res Commun. 1994, 203: 1309-1317. 10.1006/bbrc.1994.2325.CrossRefPubMed Itoh M, Ishihara K, Tomizawa H, Tanaka H, Kobune Y, Ishikawa J, Kaisho T, Hirano T: Molecular cloning of murine BST-1 having homology with CD38 and Aplysia ADP-ribosyl cyclase. Biochem Biophys Res Commun. 1994, 203: 1309-1317. 10.1006/bbrc.1994.2325.CrossRefPubMed
27.
Zurück zum Zitat Demmer LA, Birkenmeier EH, Sweetser DA, Levin MS, Zollman S, Sparkes RS, Mohandas T, Lusis AJ, Gordon JI: The cellular retinol binding protein II gene: sequence analysis of the rat gene, chromosomal localization in mice and humans, and documentation of its close linkage to the cellular retinol binding protein gene. J Biol Chem. 1987, 262: 2458-2467.PubMed Demmer LA, Birkenmeier EH, Sweetser DA, Levin MS, Zollman S, Sparkes RS, Mohandas T, Lusis AJ, Gordon JI: The cellular retinol binding protein II gene: sequence analysis of the rat gene, chromosomal localization in mice and humans, and documentation of its close linkage to the cellular retinol binding protein gene. J Biol Chem. 1987, 262: 2458-2467.PubMed
28.
Zurück zum Zitat Wu Q, Li L, Cooper MD, Pierres M, Gorvel JP: Aminopeptidase A activity of the murine B-lymphocyte differentiation antigen BP-1/6C3. Proc Nat Acad Sci. 1991, 88: 676-680.CrossRefPubMedPubMedCentral Wu Q, Li L, Cooper MD, Pierres M, Gorvel JP: Aminopeptidase A activity of the murine B-lymphocyte differentiation antigen BP-1/6C3. Proc Nat Acad Sci. 1991, 88: 676-680.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Hardwick C, Hoare K, Owens R, Hohn HP, Moore D, Cripps V, Austen L, Nance DM, Turley EA: Molecular cloning of a novel hyaluronan receptor that mediates tumor cell motility. J Cell Biol. 1992, 117: 1343-1350.CrossRefPubMed Hardwick C, Hoare K, Owens R, Hohn HP, Moore D, Cripps V, Austen L, Nance DM, Turley EA: Molecular cloning of a novel hyaluronan receptor that mediates tumor cell motility. J Cell Biol. 1992, 117: 1343-1350.CrossRefPubMed
30.
Zurück zum Zitat Hall CL, Wang C, Lange LA, Turley EA: Hyaluronan and the hyaluronan receptor RHAMM promote focal adhesion turnover and transient tyrosine kinase activity. J Cell Biol. 1994, 126: 575-588.CrossRefPubMed Hall CL, Wang C, Lange LA, Turley EA: Hyaluronan and the hyaluronan receptor RHAMM promote focal adhesion turnover and transient tyrosine kinase activity. J Cell Biol. 1994, 126: 575-588.CrossRefPubMed
31.
Zurück zum Zitat Martasek P, Camadro JM, Delfau-Larue MH, Dumas JB, Montagne JJ, De Verneuil H, Labbe P, Grandchamp B: Molecular cloning, sequencing, and functional expression of a cDNA encoding human coproporphyrinogen oxidase. Proc Nat Acad Sci. 1994, 91: 3024-3028.CrossRefPubMedPubMedCentral Martasek P, Camadro JM, Delfau-Larue MH, Dumas JB, Montagne JJ, De Verneuil H, Labbe P, Grandchamp B: Molecular cloning, sequencing, and functional expression of a cDNA encoding human coproporphyrinogen oxidase. Proc Nat Acad Sci. 1994, 91: 3024-3028.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Macy JA, Gilroy J, Perrin JC: Hereditary coproporphyria: a imitator of multiple sclerosis. Arch Phys Med Rehabil. 1991, 72: 703-704.PubMed Macy JA, Gilroy J, Perrin JC: Hereditary coproporphyria: a imitator of multiple sclerosis. Arch Phys Med Rehabil. 1991, 72: 703-704.PubMed
Metadaten
Titel
Differential gene expression in the murine gastric fundus lacking interstitial cells of Cajal
verfasst von
Yataro Daigo
Ichiro Takayama
Bruce AJ Ponder
Carlos Caldas
Sean M Ward
Kenton M Sanders
Masayuki A Fujino
Publikationsdatum
01.12.2003
Verlag
BioMed Central
Erschienen in
BMC Gastroenterology / Ausgabe 1/2003
Elektronische ISSN: 1471-230X
DOI
https://doi.org/10.1186/1471-230X-3-14

Weitere Artikel der Ausgabe 1/2003

BMC Gastroenterology 1/2003 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.