throbber
Life Sciences 71 (2002) 667 – 678
`
`www.elsevier.com/locate/lifescie
`
`Expression of dopamine receptors and transporter in
`neuroendocrine gastrointestinal tumor cells
`
`K. Lemmer a, G. Ahnert-Hilger b, M. Ho¨pfner a, S. Hoegerle c, S. Faiss a, P. Grabowski a,
`M. Jockers-Scheru¨bl d, E.O. Riecken a, M. Zeitz a, H. Scheru¨bl a,*
`
`aMed. Klinik I, Universita¨tsklinikum Benjamin Franklin, FU Berlin, Germany
`bInst. Anatomie, HU, Charite´ Mitte, Berlin, Germany
`cAbt. Nuklearmedizin, Albert-Ludwigs Universita¨t, Freiburg, Germany
`dPsych. Klinik, Universita¨tsklinikum Benjamin Franklin, FU Berlin, Germany
`
`Received 17 July 2001; accepted 14 January 2002
`
`Abstract
`
`C-11- or F-18-DOPA positron emission tomography (DOPA PET) is a new sensitive imaging technique for
`small neuroendocrine gastrointestinal tumors which evaluates the decarboxylase activity. To further characterize
`the dopaminergic system in neuroendocrine gastrointestinal tumor cells, we investigated the expression of both
`dopamine receptors and the transmembrane dopamine transporter
`(DAT)
`in the human neuroendocrine
`pancreatic cell line BON and in the neuroendocrine gut cell line STC-1. Both BON and STC-1 cells expressed
`mRNA of the dopamine receptors D2 – D5 and DAT. mRNA of the dopamine receptor D1 was detected in BON
`cells only. Both in BON and STC-1 cells, expression of D2 and D5 receptors and DAT was also demonstrated
`immunocytochemically. For functional receptor characterization intracellular cAMP levels ([cAMP]i) were
`determined. Whereas in STC-1 cells dopamine and the D1-like (D1/D5) receptor agonist SKF 38393 increased
`[cAMP]i, [cAMP]i was decreased by dopamine or the D2-like (D2 – D4) receptor agonist quinpirole in BON
`cells. Functional DAT activity was, however, not detected in either cell line. The presence of both dopamine
`receptors and of the DAT suggests an autocrine and/or paracrine function of dopamine in neuroendocrine
`gastrointestinal tumor cells. Yet neither the transmembrane dopamine transporter nor dopamine receptors are
`likely to contribute to positive DOPA PET imaging of neuroendocrine gastrointestinal tumors. However, these
`
`* Corresponding author. Tel.: +49-30-8445-3534; fax: +49-30-8445-4481.
`E-mail address: hscher@zedat.fu-berlin.de (H. Scheru¨bl).
`
`0024-3205/02/$ - see front matter D 2002 Published by Elsevier Science Inc.
`PII: S 0 0 2 4 - 3 2 0 5 ( 0 2 ) 0 1 7 0 3 - 4
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 001
`
`

`
`668
`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`molecules may be of diagnostic importance when applying other dopaminergic system tracers. D 2002
`Published by Elsevier Science Inc.
`
`Keywords: DOPA PET; Dopamine transporter; Dopamine receptor; Carcinoid; cAMP
`
`Introduction
`
`Neuroendocrine gastrointestinal tumors, i.e. carcinoids and endocrine pancreatic tumors, are often
`small and difficult to localize. Recently, C-11- or F-18-DOPA positron emission tomography (DOPA
`PET) has been introduced as a new sensitive imaging technique for the detection and staging of these
`tumors [1,2]. In view of the possibilities of PET to study metabolic changes (during therapy), its future
`clinical application may well extend beyond diagnostic use.
`Gastrointestinal neuroendocrine tumors belong to the amine precursor uptake and decarboxylation
`(APUD) cell system and have the capacity to uptake and to decarboxylate amine precursors such as 5-
`hydroxytryptophan. The re-uptake of monoamines most likely proceeds via selective sodium/chloride
`dependent monoamine transporters of the plasma membrane [3,4]. Accumulation of monoamines into
`secretory vesicles is achieved by vesicular monoamine transporters [5,6]. Dopa is readily decarboxylated
`to dopamine in peripheral tissues [7] and the decarboxylase activity is thought to be essential for positive
`DOPA PET imaging. Although dopamine, its receptors and the transmembrane dopamine transporter
`(DAT) are known to play an important role in gastrointestinal physiology [8 –11], they have not been
`investigated in neuroendocrine gastrointestinal tumor cells so far.
`Dopamine receptors belong to the family of G protein-coupled receptors. They modulate the activity
`of adenylate cyclases (AC) via G proteins. Based on their AC interactions the five known dopamine
`receptors are divided into two subfamilies, i.e. the D1-like (D1 and D5) and the D2-like (D2, D3 and D4)
`receptors. D1-like receptors stimulate AC activity via Gs proteins, while D2-like receptors decrease
`intracellular cAMP accumulation ([cAMP]i) via Gi/Go proteins [12,13]. As for the transmembrane
`dopamine transporter different subtypes have not been described so far [3,14].
`In this study we investigated the expression of the dopamine receptors D1– D5 and the trans-
`membrane dopamine transporter DAT in the human pancreatic neuroendocrine tumor cell line BON and
`in the neuroendocrine gut tumor cell line STC-1. In addition to mRNA and immunofluorescence studies,
`we functionally characterized dopamine receptors and the transmembrane dopamine transporter.
`
`Material and methods
`
`Cell cultures
`
`STC-1 cells, derived from a murine neuroendocrine gut tumor, were cultured in Dulbecco’s modified
`Eagle medium (DMEM) supplemented with 15% horse serum, 2.5% fetal calf serum (FCS) and 1% L-
`glutamine at 37 jC in a 5% CO2 humidified atmosphere [17]. The human pancreatic neuroendocrine
`(carcinoid) cell line BON was maintained in a mixture of DMEM and F12K medium (1:1) supplemented
`with 10% FCS and 1% L-glutamine. BON cells were cultured at 37 jC in a 5% CO2 humidified
`atmosphere [15].
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 002
`
`

`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`669
`
`Reverse transcription-polymerase chain reaction (RT-PCR)
`
`RNAs from both cell lines, and from mouse and human brain (cortex) tissues, were isolated with a
`commercial RNA isolation kit (PURE script, Biozym, Oldendorf, Germany). To eliminate residual
`genomic DNA, the RNA samples were treated with DNase I (Gibco, Eggenstein, Germany) for 15 min at
`room temperature. Single strand cDNA was generated from the cellular mRNA by reverse transcription
`using Superscript II RNase H Reverse Transcriptase (Gibco) as described previously [16]. Reverse
`transcription and amplification conditions were carried out as described previously [17]. The oligonu-
`cleotide primers (Amersham Pharmacia, Freiburg, Germany) used for amplification of the dopamine
`receptor subtypes D1–D5 and DAT (Table 1) were designed from GenBank sequences with BLASTN
`2.0 software, NCBI (National Center for Biotechnology Information, Bethesda, MD, USA) or selected
`from published primer sequences [18– 23]. PCR started with an inital denaturation step for 5 min at 94
`jC. Maintaining the samples at 85 jC 1.25 U Taq polymerase (Promega, Mannheim, Germany) was
`added. Cyclic denaturation at 94 jC for 45 s and elongation at 72 jC for 2 min were identical in each
`PCR, with different annealing temperatures for respective primers as shown in Table 1. Amplification
`consisted of 38 cycles. PCR products were visualized in ethidium bromide-stained 1.8% agarose gels
`and band lengths were compared with a 123 bp DNA ladder (Gibco).
`
`Table 1
`RT-PCR primers used for the detection of dopamine receptors D1 – D5 or the transmembrane dopamine transporter DAT
`
`Subtype Species Primer sequence
`
`D1
`
`mouse
`
`human
`
`human/
`mouse
`human
`
`D2
`
`D2S
`
`D3
`
`mouse
`
`human
`
`D4
`
`mouse
`
`human
`
`D5
`
`mouse
`
`human
`
`DAT
`
`mouse
`
`human
`
`5V-GTAGCCATTATGATCGTCAC-3V
`5V-GATCACAGACAGTGTCTTCAG-3V
`5V-CAGTCCACGCCAAGAATTGCC-3V
`5V-ATTGCACTCCTTGGAGATGGAGCC-3V
`5V-GCAGCCGAGCTTTCAGGGCC-3V
`5V-GGGATGTTGCAGTCACAGTG-3V
`5V-GAGGGCTCCACTAAAGGAGG-3V
`5V-GGGATGTTGCAGTCACAGTG-3V
`5V-AGGTTTCTGTCAGATGCC-3V
`5V-GTTGCTGAGTTTTCGAACC-3V
`5V-GCGTTACTACAGCATCTGCCAGGAC-3V
`5V-AGACAGGATCTTGAGGAAGG-3V
`5V-CACCAACTACTTCATCGTGA-3V
`5V-AAGGAGCAGACGGACGAGTA-3V
`5V-TGCTGTGCTGGACGCCCTTCTTCG-3V
`5V-CGTTGCGGAACTCGGCGTTGAAGA-3V
`5V-CTACGAGCGCAAGATGACC-3V
`5V-CTCTGAGCATGCTCAGCTG-3V
`5V-GTCGCCGAGGTGGCCGGTTAC-3V
`5V-GCTGGAGTCACAGTTCTCTGCAT-3V
`5V-GGCTTACAGGACCTCAAAG-3V
`5V-TGAACCTCCACTGGTGTCT-3V
`5V-AGCAGAACGGAGTGCAGCT-3V
`5V-GTATGCTCTGATGCCGTCT-3
`
`Annealing
`temperature
`55 jC
`
`RT-PCR
`product
`
`213 bp
`
`GenBank accession
`number/Reference
`
`Acc. no. L20336
`
`60 jC
`
`60 jC
`
`60 jC
`
`55 jC
`
`55 jC
`
`58 jC
`
`55 jC
`
`58 jC
`
`60 jC
`
`58 jC
`
`55 jC
`
`455 bp
`
`529 bp (D2L)
`442 bp (D2S)
`417 bp
`
`Vrana et al. 1995 [20]
`Acc. no. X55760
`Vrana et al. 1995 [20]
`Acc. no. S69899
`Acc. no. S69899
`
`276 bp
`
`Fishburn et al. 1993 [18]
`
`439 bp
`
`Segal et al. 1997 [23]
`
`393 bp
`
`162 bp
`
`359 bp
`
`363 bp
`
`258 bp
`
`Matsumoto et al. 1995 [19]
`Acc. no. U19880
`Mulcrone and Kerwin
`1996 [21]
`Acc. no. L20330
`
`Nagai et al. 1996 [22]
`Acc. no. X58454
`Acc. no. U16265
`
`785 bp
`
`Acc. no. L24178
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 003
`
`

`
`670
`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`Immunocytochemistry
`
`STC-1 and BON cells were grown on 12 mm coverslips for 3–4 days. The cells were gently washed 3
`times for 5 minutes with icecold 0.15 M PBS (pH 7.4) and fixed with 3.5% formaline dissolved in PBS
`for 20 min at room temperature. Membrane permeabilization was achieved by incubating the cells with
`0.2% Triton X-100 for 10 min. Nonspecific binding was blocked with 1% BSA in PBS for 1 h. The cells
`were then incubated for 1 h with polyclonal antisera (1 Ag IgG/ml) diluted with 0.3% BSA in PBS. The
`specific antibodies had been raised against amino- or carboxy-terminal peptides of the D2 receptor (N-
`19; Santa Cruz Biotechnology, Heidelberg, Germany), the D5 receptor (R-18; C-20) or DAT (N-19).
`Each coverslip was overlaid with 100 Al antibody solution. Negative controls were prepared by
`incubation with polyclonal antibodies that had been neutralized by a five fold excess of their antigens
`(blocking peptides; Santa Cruz Biotechnology). Immunocomplexes were detected with fluorescein-
`conjugated secondary antibodies (Santa Cruz Biotechnology). The rinsed coverslips were dried, mounted
`on slides with Aquamount (Polysciences, St. Goar, Germany) and examined on an Axioskop-2
`microscope (Zeiss, Jena, Germany).
`
`cAMP assay
`
`cAMP accumulation was determined with a competitive enzyme immunoassay (BiotrakTM; Amer-
`sham Pharmacia Biotech, Braunschweig, Germany). STC-1 and BON cells were seeded at a concen-
`tration of 50,000 cells/well in 96 well microtitre plates and cultured for 48 h. The cells were washed
`twice with sodium bath solution containing 130 mM NaCl, 5.4 mM KCl, 1 mM MgCl2, 10 mM glucose
`and 10 mM Hepes, pH 7.3 (37 jC). Cells were then incubated in 200 Al sodium bath solution
`supplemented with 100 AM isobutylmethylxanthine (IBMX) (Sigma, Deisenhofen, Germany) in the
`absence or presence of 1 AM forskolin (Sigma) and/or the following drugs: 100 AM dopamine, 100 AM
`SKF 38393 or 100 AM quinpirole (Sigma). Incubation was carried out for 15 min at 37 jC. Following
`two rinses with sodium bath solution 200 Al lysis reagent was added. Cells were agitated for up to 15
`min and cell lysis was verified by microscopic evaluation. 100 Al aliquot of cell lysate were transfered to
`the 96 well microtitre plate and the assay was carried out as described previously [24]. The cAMP values
`(fmol/well) of the samples were calculated by direct read-off from the standard curve.
`
`[3H] dopamine uptake
`
`Primary neuronal cultures were prepared from embryonic (ED 15) mouse mesencephalon and
`cultured for 2 weeks as described previously [25]. BON and STC-1 cells were grown for 48 h. Cells
`were preloaded with [3H] dopamine in serum free medium supplemented with 1 mM ascorbic acid in the
`absence or presence of 0.1–1 AM GBR 12783, a selective DAT inhibitor. Uptake of [3H] dopamine was
`stopped by several washes after 4 h of incubation at 37 jC. Radioactivity as well as protein content were
`estimated after lysing the cells in 0.1% Triton X-100.
`
`Statistics
`
`Results are expressed as means F standard error. Data were analyzed using Student’s non-paired two-
`tailed t-test.
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 004
`
`

`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`671
`
`Results and discussion
`
`Expression of dopamine receptor and DAT mRNAs
`
`RT-PCR revealed mRNA expression of dopamine receptors and DAT in STC-1 and BON cells (Fig.
`1). While STC-1 cells expressed mRNA of D2–D5 receptors only, BON cells expressed mRNA of all
`five dopamine receptors. In the two cell lines mRNA of both the long and the short D2 isoforms, D2L
`and D2S, generated by alternative splicing [26] were present. In STC-1 cells, D2L mRNA amplification
`resulted in an abundant band, whereas D2S mRNA showed a weaker amplification, which suggests a
`predominance of the longer D2 isoform in STC-1 cells [20]. In BON cells, mRNA amplification with the
`primer pair D2 resulted in an abundant D2L band but in contrast to brain no D2S amplification could be
`observed. By use of a D2S mRNA selective forward primer (D2S primer pair listed in Table 1) mRNA
`amplification of BON cells resulted in a marked D2S band. These findings suggest that D2S mRNA
`amplification with the primer pair D2 was competitively inhibited by large amounts of D2L mRNA.
`
`Fig. 1. Agarose gel electrophoresis with PCR products of dopamine receptors and the dopamine transporter in STC-1 and
`BON cells. Top panel: Amplification was performed with cDNA from mouse brain as control (C) and from STC-1 cells (S).
`The primers used corresponded to the dopamine receptors D1, D2 (amplifying both the long and short isoforms D2L and
`D2S), D3, D4 or D5 and to the transmembrane dopamine transporter DAT. M, 123 bp DNA ladder. Bottom panel:
`Amplification was performed with cDNA from human brain as control (C) and from BON cells (B). The primers used
`corresponded to the dopamine receptors D1, D2L/S, D2S, D3, D4 or D5 and to the transmembrane dopamine transporter DAT.
`M, 123 bp DNA ladder.
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 005
`
`

`
`672
`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`Thus, STC-1 cells as well as (mouse and human) brain seem to express lower amounts of D2L mRNA in
`relation to D2S mRNA than BON cells do. Although both D3 and D4 mRNAs are rarely found in the
`gastrointestinal tract of the rat [9,11], both D3 and D4 mRNAs were expressed in the human BON and in
`the murine STC-1 cells. In STC-1 cells only a D3 mRNA band of 276 bp length was obtained (D3L),
`while the shorter splice variant of 213 bp length [18] was not observed. Human D3 mRNA was
`amplified in low amount in BON cells, resulting in a single band equivalent in length to D3L mRNA.
`Both cell lines expressed mRNA of the D5 receptor which has been described as the most prevalent
`dopamine receptor in the upper gastrointestinal tract [27–30]. The expression patterns of dopamine
`receptor mRNAs in STC-1 and BON cells were reproducible with cells of different passages. Thus,
`similar to the situation in neurons [20], STC-1 as well as BON cells co-express mRNAs of both
`subfamilies of dopamine receptors, i.e. the [cAMP]i increasing D1-like (D1, D5) receptors and the
`[cAMP]i decreasing D2-like (D2–D4) receptors. While DAT mRNA was persistently expressed in STC-
`1 cells, BON cells did not express DAT mRNA stably in different cell passages.
`
`Immunocytochemical studies of dopamine receptors and DAT
`
`D2 and D5 receptors are known to be widely distributed in the upper gastrointestinal tract [11,29].
`The antibodies used specifically recognize epitops of the extracellular N terminus or cytoplasmic
`C-terminus of the receptors or the intracellularly localized N-terminus of DAT. To access cytoplasmic
`targets,
`the cell membranes were permeabilized by incubation with 0.2% Triton X-100. Fig. 2
`demonstrates the immunoreactivity of STC-1 and BON cells after incubation with polyclonal antibodies
`against the D2 receptor (Fig. 2A and 2E) and the D5 receptor (Fig. 2C and 2G).
`Both cell lines were also immunopositive for DAT (Fig. 3A and 3C). But in contrast to the STC-1 cell
`line only about 25% of BON cells showed a bright fluorescence for DAT (in three independent
`experiments). This may correspond to the observed unstable expression of DAT mRNA or the diversity
`of BON cells. The immunofluorescence signals of DAT, D2 or D5 receptors were interspersed
`homogeneously only sparing nuclei. Preabsorption of the antibodies with immunizing peptides
`eliminated the strong immunofluorescences (Figs. 2B, 2D, 2F, 2H, 3B and 3D). Hence, both STC-1
`and BON cells do co-express D2 and D5 receptors as well as DAT. Interestingly D2, D5 and DAT
`immunoreactivity had been described previously in the gastrointestinal tract. Thus, gastric and duodenal
`mucosa and pancreatic cells express DAT together with tyrosine hydroxylase activity, high levels of
`dopamine, vesicular monoamine transporters (VMAT) and D5 mRNA [9,11,27,29,30]. Similarly,
`VMAT1 and VMAT2 expression could be detected immunocytochemically in BON cells [6].
`
`Adenylate cyclase activity and dopamine receptors
`
`To study dopamine receptor function, adenylate cyclase (AC) activity both of untreated and of
`forskolin-stimulated STC-1 and BON cells was investigated in the absence or presence of either
`dopamine, the D1-like receptor agonist SKF 38393 or the D2-like receptor agonist quinpirole. To block
`phosphodiesterase activity, cells were treated with 1 AM IBMX. In unstimulated STC-1 cells 100 AM
`dopamine as well as 100 AM SKF 38393 increased basal [cAMP]i by about 35% (p < 0.05) (Fig. 4).
`Incubation with 1 AM forskolin, a direct activator of the catalytic subunit of AC resulted in about a two
`fold increase of [cAMP]i (p < 0.05). Co-incubation with dopamine or SKF 38393 potentiated the
`forskolin-stimulated cAMP accumulation by about 230% and 150%, respectively (p < 0.05). The
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 006
`
`

`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`673
`
`Fig. 2. Expression of D2 and D5 dopamine receptors in STC-1 (A – D) and in BON cells (E – H). Cells were fixed,
`permeabilized and immunostained as described in materials and methods. Immunofluorescence microscopy was performed
`using a conventional fluorescence microscope. STC-1 cells were incubated with the D2-specific polyclonal antibody N-19 (A)
`or the D5-specific (mouse reactive) polyclonal antibody R-18 (C) and for control with neutralized antibodies (B, D). Similarly,
`BON cells were incubated with the D2-specific antibody N-19 (E) or the D5-specific (human reactive) antibody C-20 (G) and
`for control with neutralized antibodies (F, H). The immunocomplexes were detected with fluorescein-conjugated secondary
`antibodies. Magnification: 630.
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 007
`
`

`
`674
`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`Fig. 3. Expression of the transmembrane dopamine transporter DAT in STC-1 (A, B) and in BON cells (C, D). Cells were fixed,
`permeabilized and immunostained as described in materials and methods. Immunofluorescence microscopy was performed
`using a conventional fluorescence microscope. STC-1 (A) and BON cells (C) were incubated with the DAT-specific polyclonal
`antibody N-19 or for control with the neutralized antibody (B, D). The immunocomplexes were detected with a fluorescein-
`conjugated secondary antibody. Magnification 400 (A, B), 630 (C, D).
`
`selective D2 receptor agonist quinpirole did not decrease cAMP accumulation in STC-1 cells (data not
`shown).
`In contrast to the stimulatory effect of dopamine in STC-1 cells, dopamine decreased cAMP
`accumulation in BON cells (Fig. 5). In unstimulated BON cells 100 AM dopamine reduced basal
`[cAMP]i by about 40% (p < 0.05). Similar results were obtained with 100 AM quinpirole (p < 0.05).
`Forskolin (1 AM) enhanced [cAMP]i accumulation in BON cells by about 7 fold (p < 0.05). Co-
`incubation with 100 AM dopamine or 100 AM quinpirole decreased the (1 AM) forskolin-induced
`[cAMP]i by about 30% and 20%, respectively (n = 6, p < 0.05). The D1-like receptor agonist SKF 38393
`did not increase either basal or forskolin-induced cAMP accumulation in BON cells (data not shown).
`These results suggest that in BON cells functional D2-like receptors prevail, while in STC-1 cells
`functional D1-like receptors do so.
`
`[ 3H] dopamine uptake
`
`DAT activity in both BON and STC-1 cells was assessed by measurement of [3H] dopamine uptake.
`For comparison [3H] dopamine uptake was also studied in mouse mesencephalon [25] which is known to
`contain dopaminergic neurons from substantia nigra. Intracellular dopamine concentrations differed
`significantly in mesencephalic neurons in the absence or presence of 1 AM of the selective DAT inhibitor
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 008
`
`

`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`675
`
`Fig. 4. Effects of dopamine (A) and SKF 38393 (B) on the intracellular cAMP level of STC-1 cells. Basal and (1 AM) forskolin-
`stimulated cAMP accumulation were measured in the absence or presence of 100 AM dopamine or 100 AM SKF 38393. Means
`F standard errors (n = 6) are given. *, p < 0.05.
`
`GBR 12783 (9.5 F 1.9 versus 2.7 F 1.1 pmol [3H] dopamine/mg protein, n = 10). In contrast, [3H]
`dopamine uptake was not affected significantly by the DAT inhibitor GBR 12783 (1 AM) neither in BON
`cells (0.13 F 0.01 versus 0.19 F 0.05 pmol/mg protein, n = 10) nor in STC-1 cells (1.5 F 0.9 versus
`0.85 F 0.13 pmol/mg protein; n = 10). Thus, functional DAT activity could not be detected in either
`BON or STC-1 cells.
`The results of this study provide hitherto undescribed evidence for the presence of both dopamine
`receptors and the transmembrane dopamine transporter in neuroendocrine gastrointestinal tumor cells.
`As neuroendocrine tumor cells have the capacity for uptake and decarboxylation of amine precursors
`such as 5-hydroxytryptophan and L-dopa and as they can secrete serotonin and dopamine [31–33], the
`presence of dopamine receptors and of the DAT suggests an autocrine and/or paracrine function of
`dopamine in neuroendocrine gastrointestinal tumor cells.
`Due to the relatively low affinity (high off-rate constant) and competition with relatively high levels
`of endogenous dopamine the binding of F-18-dopamine formed from F-18-DOPA to either DAT or
`dopamine receptors will not produce interpretable PET images. Thus, neither the transmembrane
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 009
`
`

`
`676
`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`Fig. 5. Effects of dopamine (A) and quinpirole (B) on the intracellular cAMP level of BON cells. Basal and (1 AM) forskolin-
`stimulated cAMP accumulation were measured in the absence or presence of 100 AM dopamine or 100 AM quinpirole. Means
`F standard errors (n = 6) are given. *, p < 0.05.
`
`dopamine transporter nor dopamine receptors are likely to contribute to positive DOPA PET imaging of
`neuroendocrine gastrointestinal tumors. However, these molecules may be of diagnostic importance
`when applying other dopaminergic system tracers [34,35].
`
`Acknowledgements
`
`The study was supported by the Deutsche Forschungsgemeinschaft, the Deutsche Krebshilfe and the
`Sonnenfeld-Stiftung. We are indebted to the Institute of Physiology, FU Berlin for lab facilities.
`
`References
`
`[1] Ahlstro¨m H, Eriksson B, Bergstro¨m M, Bjurling P, La¨ngstro¨m B, O¨ berg K. Pancreatic neuroendocrine tumors: Diagnosis
`with PET. Radiology 1995;195:333 – 7.
`[2] Hoegerle S, Altehoefer C, Ghanem N, Koehler G, Waller C, Scheru¨bl H, Moser E, Nitzsche E. Whole-body 18F dopa PET
`for detection of gastrointestinal carcinoid tumors. Radiology 2001;220:373 – 80.
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 010
`
`

`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`677
`
`[3] Shimada S, Kitayama S, Lin C, Patel A, Nanthakumar E, Gregor P, Kuhar M, Uhl G. Cloning and expression of a cocaine-
`sensitive dopamine transporter complementary DNA. Science 1991;254:576 – 8.
`[4] Giros B, El Mestikawy S, Godinot N, Zheng K, Han H, Yang-Feng T, Caron M. Cloning, pharmacological character-
`ization, and chromosome assignment of the human dopamine transporter. Molecular Pharmacology 1992;42:383 – 90.
`[5] De Giorgio R, Su D, Peter D, Edwards R, Brecha N, Sternini C. Vesicular monoamine transporter 2 expression in enteric
`neurons and enterochromaffin-like cells of the rat. Neuroscience Letters 1996;217:77 – 80.
`[6] Holtje M, von Jagow B, Pahner I, Lautenschlager M, Hortnagl H, Nu¨rnberg B, Jahn R, Ahnert-Hilger G. The neuronal
`monoamine transporter VMAT2 is regulated by the trimeric GTPase Go(2). Journal of Neuroscience 2000;20:2131 – 41.
`[7] Borelli M, Villar M, Orezzoli A, Gagliardino J. Presence of DOPA decarboxylase and its localisation in adult rat panceatic
`islet cells. Diabetes Metabolism 1997;23:161 – 3.
`[8] Hernandez D, Mason G, Walker C, Valenzuela J. Dopamine receptors in human gastrointestinal mucosa. Life Sciences
`1987;41:2717 – 23.
`[9] Mezey E´ , Eisenhofer G, Hansson S, Hunyady B, Hoffman B. Dopamine produced by the stomach may act as a paracrine/
`autocrine hormone in the rat. Neuroendocrinology 1998;67:336 – 48.
`[10] Flemstro¨m G, Safsten B, Knutson L. Dopamine and the brain-gut axis. Advances in Pharmacology 1998;42:846 – 51.
`[11] Mezey E´ , Eisenhofer G, Hansson S, Harta G, Hoffman B, Gallatz K, Palkovits M, Hunyady B. Non-neuronal dopamine in
`the gastrointestinal system. Clinical and Experimental Pharmacology and Physiology 1999;26:14 – 22.
`[12] Neve K, Neve R. The Dopamine Receptors. Totowa, New Jersey: Humana Press, 1997.
`[13] Missale C, Nash S, Robinson S, Jaber M, Caron M. Dopamine receptors: From structure to function. Physiological
`Reviews 1998;78:189 – 225.
`[14] Sugamori K, Lee F, Pristupa Z, Niznik H. A cognate dopamine transporter-like activity endogenously expressed in a COS-
`7 kidney-derived cell line. FEBS Letters 1999;451:169 – 74.
`[15] Ahnert-Hilger G, Stadtba¨umer A, Stru¨bing C, Scheru¨bl H, Schultz G, Riecken E, Wiedenmann B. Gamma-aminobutyric
`acid secretion from pancreatic neuroendocrine cells. Gastroenterology 1996;110:1595 – 604.
`[16] Jansen A, Ho¨pfner M, Herzig K, Riecken E, Scheru¨bl H. GABAC receptors in neuroendocrine gut cells: a new GABA
`binding site in the gut. Pflu¨gers Archiv European Journal of Physiology 2000;440:1120 – 7.
`[17] Glassmeier G, Herzig K, Ho¨pfner M, Lemmer K, Jansen A, Scheru¨bl H. Expression of functional GABAA receptors in
`cholecystokinin-secreting gut neuroendocrine murine STC-1 cells. Journal of Physiology 1998;510:805 – 14.
`[18] Fishburn C, Belleli D, David C, Carmon S, Fuchs S. A novel short isoform of the D3 dopamine receptor generated by
`alternative splicing in the third cytoplasmic loop. The Journal of Biological Chemistry 1993;268:5872 – 8.
`[19] Matsumoto M, Hidaka K, Tada S, Tasaki Y, Yamaguchi T. Full-length cDNA cloning and distribution of human dopamine
`D4 receptor. Molecular Brain Research 1995;29:157 – 62.
`[20] Vrana S, Kluttz B, Vrana K. Application of quantitative RT-PCR to the analysis of dopamine receptor mRNA levels in rat
`striatum. Molecular Brain Research 1995;34:127 – 34.
`[21] Mulcrone J, Kerwin R. No difference in the expression of the D4 gene in post-mortem frontal cortex from controls and
`schizophrenics. Neuroscience Letters 1996;219:163 – 6.
`[22] Nagai Y, Ueno S, Saeki Y, Soga F, Hirano M, Yanagihara T. Decrease of the D3 dopamine receptor mRNA expression in
`lymphocytes from patients with Parkinson’s disease. Neurology 1996;46:791 – 5.
`[23] Segal D, Moraes C, Mash D. Up-regulation of D3 dopamine receptor mRNA in the nucleus accumbens of human cocaine
`fatalities. Molecular Brain Research 1997;45:335 – 9.
`[24] Ho¨pfner M, Maaser K, von Lampe B, Hanski C, Riecken E, Scheru¨bl H. Growth inhibition and apoptosis induced by
`P2Y2 receptors in colorectal carcinoma cells: Involvement of intracellular calcium and cyclic AMP. International Journal
`Colorectal Disease 2001;16:154 – 66.
`[25] Ahnert-Hilger G, Kutay U, Chahoud I, Rapoport T, Wiedenmann B. Synaptobrevin is essential for secretion but not for the
`development of synaptic processes. European Journal of Cell Biology 1996;70:1 – 11.
`[26] Dal Toso R, Sommer B, Ewert M, Herb A, Pritchett D, Bach A, Shivers B, Seeburg P. The dopamine D2 receptor: two
`molecular forms generated by alternative splicing. The EMBO Journal 1989;8:4025 – 34.
`[27] Mezey E´ , Eisenhofer G, Harta G, Hansson S, Gould L, Hunyady B, Hoffman B. A novel nonneuronal catecholaminergic
`system: Exocrine pancreas synthesizes and releases dopamine. Proceedings of National Academy of Sciences 1996;93:
`10377 – 82.
`[28] Eisenhofer G, Aneman A, Friberg P, Hooper D, Fandriks L, Lonroth H, Hunyady B, Mezey E´ . Substantial production of
`dopamine in the human gastrointestinal tract. Journal of Clinical Endocrinology and Metabolism 1997;82:3864 – 71.
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 011
`
`

`
`678
`
`K. Lemmer et al. / Life Sciences 71 (2002) 667–678
`
`[29] Hunyady B, Hoffmann B, Eisenhofer G, Hansson S, Mezey E´ . Dopaminergic characteristics of isolated parietal cells from
`rats. Journal of Physiology Paris 1997;91:247 – 56.
`[30] Hunyady B, Young III W, Mezey E´ . Distribution of dopamine receptor subtypes in the upper GI tract of the rat. Gastro-
`enterology 1998;114:A1150.
`[31] Feldmann J. Increased dopamine production in patients with carcinoid tumors. Metabolism 1985;34:255 – 60.
`[32] Kema I, de Vries E, Slooff M, Biesma B, Muskiet F. Serotonin, catecholamines, histamine, and their metabolites in urine,
`platelets, and tumor tissue of patients with carcinoid tumors. Clinical Chemistry 1994;40:86 – 95.
`[33] O¨ rlefors H, Sundin A, Ahlstro¨m H, Bjurling P, Bergstro¨m M, Lilja A, Langstro¨m B, O¨ berg K, Eriksson B. Positron
`emission tomography with 5-hydroxytryptophan in neuroendocrine tumors. Journal of Clinical Oncology 1998;16:2534 –
`41.
`[34] Kugaya A, Fujita M, Innis R. Applications of SPECT imaging of dopaminergic neurotransmission in neuropsychiatric
`disorders. Annals of Nuclear Medicine 2000;14:1 – 9.
`[35] de Herder W, Reijs A, de Swart J, Kaandorp Y, Lamberts S, Krenning E, Kwekkeboom D. Comparison of iodine-123
`epidepride and iodine-123 IBZM for dopamine D2 receptor imaging in clinically non-functioning pituitary macroadeno-
`mas and macroprolactinomas. European Journal of Nuclear Medicine 1999;26:46 – 50.
`
`Roxane Labs., Inc.
`Exhibit 1037
`Page 012

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket