| Nyegaard
M, Borglum AD, Bruun TG, Collier DA, Russ C, Mors O, Ewald H, Kruse TA. Novel
polymorphisms in the somatostatin receptor 5 (SSTR5) gene associated with bipolar
affective disorder. Mol Psychiatry 2002;7(7):745-54
"The somatostatin receptor 5 (SSTR5) gene is a candidate gene for bipolar
affective disorder (BPAD) as well as for other neuropsychiatric disorders. The
gene is positioned on chromosome 16p13.3, a region that has been implicated by
a few linkage studies to potentially harbor a disease susceptibility gene for
BPAD. Recent evidence shows that the dopamine D2 receptor (DRD2) and SSTR5 interact
physically to form heterodimers with enhanced functional activity. Brain D2 dopamine
receptors are one of the major targets of neuroleptic treatments in psychiatric
disorders. In this study we systematically screened the promoter and coding region
of the SSTR5 gene for genetic variation that could contribute to the development
of neuropsychiatric disorders. Eleven novel single nucleotide polymorphisms (SNPs)
were identified including four missense SNPs, Leu48Met, Ala52Val, Pro109Ser and
Pro335Leu. We carried out an association study of BPAD using 80 Danish cases and
144 control subjects, and replication analysis using 55 British cases and 88 control
subjects. For the Danish population, association was suggested between silent
SNP G573A and BPAD (P = 0.008). For the British population we found association
to BPAD with missense mutation Leu48Met (P = 0.003) and missense mutation Pro335Leu
(P = 0.004). The statistical significance of the association was, however, greatly
reduced after correcting for multiple testing. When combining genotypes from Leu48Met
and Pro335Leu into haplotypes, association to BPAD was found in the British population
(P = 0.0007). This haplotype association was not replicated in the Danish population.
Our results may indicate that the SSTR5 gene is involved in the etiology of BPAD
or may exist in linkage disequilibrium with a susceptibility gene close to SSTR5.
However, given the marginal statistical significance and the potential for false-positive
results in association studies with candidate genes, further studies are needed
to clarify this hypothesis." [Abstract] Strowski
MZ, Dashkevicz MP, Parmar RM, Wilkinson H, Kohler M, Schaeffer JM, Blake AD. Somatostatin
receptor subtypes 2 and 5 inhibit corticotropin-releasing hormone-stimulated adrenocorticotropin
secretion from AtT-20 cells. Neuroendocrinology 2002 Jun;75(6):339-46 "Somatostatin
(SRIH) regulates pituitary adrenocorticotropin (ACTH) secretion by interacting
with a family of homologous G protein-coupled membrane receptors. The SRIH receptor
subtypes (sst(1)-sst(5)) that control ACTH release remain unknown. Using novel,
subtype-selective SRIH analogs, we have identified the SRIH receptor subtypes
involved in regulating ACTH release from AtT-20 cells, a model for cell line pituitary
corticotropes. Radioligand-binding studies with (125)I-SRIH-14 and (125)I-SRIH-28
showed that SRIH-14 and SRIH-28 recognized specific, high-affinity and saturable
membrane-binding sites. Nonpeptidyl agonists with selectivity for the sst(2) (L-779,976;
compound 2) or sst(1)/sst(5)) (L-817,818; compound 5) receptor subtypes potently
displaced (125)I-SRIH-28 from AtT-20 cell membranes, while agonists selective
for the sst(1) (L-779,591; compound 1), sst(3) (L-796,778; compound 3) or sst(4)
(L-803,087; compound 4) subtypes were inactive. Tyr(11)-SRIH-14, compound 2 (sst(2))
or compound 5 (sst(5)) inhibited forskolin and corticotropin-releasing hormone
(CRH)-induced increases in intracellular cAMP. Furthermore, the sst(2) and sst(5)
agonists potently inhibited CRH-induced ACTH release from AtT-20 cells. These
results provide the first evidence that sst(2) and sst(5) receptor subtypes, but
not sst(1), sst(3) or sst(4), inhibit cAMP accumulation and regulate ACTH secretion
in the AtT-20 cell model of the rodent corticotrope." [Abstract] R
Day, W Dong, R Panetta, J Kraicer, MT Greenwood, and YC Patel Expression
of mRNA for somatostatin receptor (sstr) types 2 and 5 in individual rat pituitary
cells. A double labeling in situ hybridization analysis
Endocrinology 136: 5232-5235, 1995. "To characterize cell specific expression
of sstr subtypes in the pituitary we have analyzed mRNA for sstr1-5 in rat pituitary
somatotrophs by reverse transcriptase polymerase chain reaction and determined
the pattern and level of expression of mRNA for sstr subtypes 2 and 5 in individual
pituitary cell subpopulations by double label in situ hybridization. Purified
somatotrophs expressed mRNA for all 5 sstrs. In situ hybridization analysis revealed
sstr5 mRNA in 70% of somatotrophs, 57% of thyrotrophs, 38% of corticotrophs, 33%
of lactotrophs, and 21% of gonadotrophs. mRNA for sstr2 occurred in 40% of somatotrophs,
36% of thyrotrophs, 26% of lactotrophs, 3% of corticotrophs, and 8% of gonadotrophs.
Not only were more cells positive for sstr5 mRNA but the average number of autoradiographic
grains/cell was also higher for sstr5 than sstr2. These results show expression
of multiple sstr genes in individual pituitary cells. mRNA for sstr2 and 5 occur
in each of the 5 major pituitary cell subsets, sstr5 mRNA being more widely and
more abundantly expressed than sstr2." [Abstract/Full
Text] Shimon I. Somatostatin receptors
in pituitary and development of somatostatin receptor subtype-selective analogs. Endocrine.
2003 Apr;20(3):265-9. "Somatostatin receptor (SSTR) subtypes 1, 2, and
5 are expressed in the normal human pituitary. SSTR2 and SSTR5 are expressed in
almost all growth hormone (GH) cell adenomas, and prolactin (PRL)-secreting tumors
express SSTR5 more than SSTR2. SSTR4 is not detected in all pituitary adenoma
subtypes, and SSTR1 and SSTR3 are expressed in about 50% of tumors. Human GH is
regulated through ligand binding to both SSTR2 and SSTR5, but octreotide and lanreotide,
the two clinically available somatostatin analogs, bind to human SSTR2 much better
than to SSTR5. Novel SSTR2- and SSTR5- selective analogs with improved binding
affinity for these receptor subtypes are highly potent in suppressing GH release
from cultures of human fetal pituitaries or GH-cell adenomas. Only SSTR5-selective
analogs suppress in vitro PRL secretion from cultured prolactinomas. A new SSTR2+5
bispecific analog with high affinity and selectivity for both SSTR2 and SSTR5,
and a somatostatin analog with a unique broad receptor (SSTR1, 2, 3, and 5) binding
profile, are both able to inhibit in vitro GH release in GH cell adenomas partially
sensitive to octreotide. Recently, a somatostatindopamine hybrid molecule was
introduced with potentially functional synergy on GH and PRL release. Using the
expanding knowledge on SSTRs and their ligand activation, the development of novel
pharmacologic concepts may open new opportunities for effective manipulation of
this complex intracellular signaling system. These concepts may achieve better
control of pituitary hormone hypersecretion, pituitary size, as well as antitumor
effects in patients with SSTR-expressing tumors." [Abstract]
Norman M, Moldovan S, Seghers V, Wang XP, DeMayo
FJ, Brunicardi FC. Sulfonylurea receptor knockout causes glucose
intolerance in mice that is not alleviated by concomitant somatostatin subtype
receptor 5 knockout. Ann Surg 2002 Jun;235(6):767-74
"OBJECTIVE: To examine the long-term effects of Sur KO, SSTR5 KO, and double
Sur/SSTR5 KO on insulin secretion and glucose regulation. SUMMARY BACKGROUND DATA:
The sulfonylurea receptor (Sur) and somatostatin receptor type 5 (SSTR5) play
an integral role in the regulatory pathways of the endocrine pancreas. Sur knockout
(KO) and SSTR5 KO mice were generated in the authors' laboratories and crossbred
to generate Sur/SSTR5 KO mice. All mice were genotyped by Southern blotting and
polymerase chain reaction analysis. METHODS: One-year-old Sur KO, Sur/SSTR5 KO,
SSTR5 KO, and age-matched wild-type control mice underwent single-pass perfusion
of isolated pancreata with low and high glucose concentration (n = 4-6/group).
Another group of mice also underwent intraperitoneal glucose tolerance tests with
1.2 g glucose/kg body weight (n = 4/group per time point). RESULTS: Sur1 KO and
Sur/SSTR5 KO mice had profoundly decreased insulin secretion in vitro, whereas
SSTR5 KO had increased insulin secretion compared with wild-type mice. Sur1 KO
and Sur/SSTR5 mice had increased glucose response in vivo compared with wild-type
mice. Sur1 KO and Sur/SSTR5 KO mice exhibit glucose intolerance and SSTR5 KO mice
show increased insulin response in vitro. CONCLUSIONS: Sur1 KO causes glucose
intolerance and SSTR5 KO causes increased insulin secretion. However, Sur/SSTR5
double ablation does not alleviate the diabetic state of the Sur1 KO." [Abstract]
Zambre Y, Ling Z, Chen MC, Hou X, Woon CW, Culler M, Taylor
JE, Coy DH, Van Schravendijk C, Schuit F, Pipeleers DG, Eizirik DL. Inhibition
of human pancreatic islet insulin release by receptor-selective somatostatin analogs
directed to somatostatin receptor subtype 5. Biochem Pharmacol
1999 May 15;57(10):1159-64 "In order to identify the subtype responsible
for inhibition of insulin release by human B cells, SSTR-selective SS analogs
were tested in isolated human islets. Glucose-stimulated insulin secretion in
human islets incubated for 1 hr at 20 mM glucose, and in islets cultured for 24
hr at a near-physiological (6.1 mM) glucose concentration, was inhibited (<50%
of the control) by SSTR5-specific analogs and by SS14 and SS28. SS14, SS28, and
different SSTR5 preferential analogs also inhibited islet amyloid polypeptide
release during the 24-hr culture. On the other hand, a group of SSTR2-selective
analogs failed to inhibit insulin release. Analysis by reverse transcription-polymerase
chain reaction indicated that human islets express similar amounts of SSTR2 and
SSTR5 mRNAs, while human pancreatic ductal cells express much lower levels of
these mRNAs. In conclusion, our data suggest that SSTR5 is an important mediator
of the insulin inhibitory action of SS in cultured human islets." [Abstract] Cattaneo
MG, Taylor JE, Culler MD, Nisoli E, Vicentini LM. Selective stimulation
of somatostatin receptor subtypes: differential effects on Ras/MAP kinase pathway
and cell proliferation in human neuroblastoma cells. FEBS
Lett 2000 Sep 22;481(3):271-6 "In previous studies we have showed that
somatostatin (SST) inhibits cell division, mitogen-activated protein (MAP) kinase
and Ras activity in the human neuroblastoma cell line SY5Y. In the present study,
we have assessed the role of a series of SST analogs, three of which were selective
for SSTR1, SSTR2 or SSTR5, in these cellular events. All the analogs inhibited
forskolin-induced cAMP accumulation. Selective stimulation of SSTR1 or SSTR2 but
not of SSTR5 inhibited platelet-derived growth factor (PDGF)-induced [(3)H]thymidine
incorporation. The three analogs inhibited PDGF-stimulated MAP kinase activity,
at least at an early time. In contrast, none of the analogs used individually
was able to inhibit PDGF-stimulated Ras activity. A combined stimulation of SSTR2
and SSTR5 was necessary to obtain a significant inhibitory effect, suggesting
the possibility of receptor heterodimerization. These results indicate that SST
inhibition of Ras and MAP kinase activities takes place via different pathways
and that SST inhibition of PDGF-induced cell proliferation occurs via a Ras-independent
pathway." [Abstract] Shimokawa
I, Yanagihara K, Higami Y, Okimoto T, Tomita M, Ikeda T, Lee S. Effects
of aging and dietary restriction on mRNA levels of receptors for growth hormone-releasing
hormone and somatostatin in the rat pituitary. J Gerontol
A Biol Sci Med Sci 2000 Jun;55(6):B274-9 "Aging impairs and dietary restriction
may modulate pituitary response to growth hormone (GH)-releasing hormone (GHRH)
and somatostatin (SRIH) for GH secretion. Using the semiquantitative reverse-transcription
polymerase chain reaction method, we analyzed the mRNA levels of the GHRH receptor
(grfr) and SRIH receptor subtype 2 (sstr2) and subtype 5 (sstr5) in anterior pituitaries
of male rats fed ad libitum or 30% dietary restricted. Aging reduced the mRNA
levels of these receptors in a slightly different manner. The levels of grfr progressively
decreased between 6 and 24 months, whereas those of sstr2 and sstr5 declined after
16 months. Dietary restriction did not diminish the aging-dependent changes, although
it slightly augmented the levels of grfr, but not sstr2 and sstr5. The present
results suggest that the aging-dependent impairment in pituitary response for
GH secretion could result mostly from a decline in grfr rather than relative increase
of sstrs. Although DR could slightly enhance the pituitary sensitivity to GHRH,
the antiaging action may be minor at the level of gene expression." [Abstract] Pierre
Cordelier, Jean-Pierre Estève, Corinne Bousquet, Nathalie Delesque, Anne-Marie
O'Carroll, Andrew V. Schally, Nicole Vaysse, Christiane Susini, and Louis Buscail
Characterization of the antiproliferative signal mediated by the
somatostatin receptor subtype sst5 PNAS 94: 9343-9348,
August 1997. "We conclude that, in CHO cells, CCK and somatostatin modulate
cell proliferation and MAP kinase signaling cascade through a cGMP-dependent pathway.
These effects are positively regulated through CCKA receptors and negatively controlled
through sst5 receptors." [Full
Text] L Buscail, J Esteve, N Saint-Laurent,
V Bertrand, T Reisine, A O'Carroll, GI Bell, AV Schally, N Vaysse, and C Susini
Inhibition of Cell Proliferation by the Somatostatin Analogue
RC-160 is Mediated by Somatostatin Receptor Subtypes SSTR2 and SSTR5 Through Different
Mechanisms PNAS 92: 1580-1584. "Cell proliferation
was induced in CHO cells by 10% (vol/vol) fetal calf serum, 1 µM insulin,
or 0.1 µM cholecystokinin (CCK)-8; RC-160 inhibited serum-induced proliferation
of CHO cells expressing SSTR2 and SSTR5 (EC50, 53 and 150 pM, respectively) but
had no effect on growth of cells expressing SSTR1, -3, or -4. In SSTR2-expressing
cells, orthovanadate suppressed the growth inhibitory effect of RC-160. This analogue
inhibited insulin-induced proliferation and rapidly stimulated the activity of
a tyrosine phosphatase in only this cellular clone. This latter effect was observed
at doses of RC-160 (EC50, 4.6 pM) similar to those required to inhibit growth
(EC50, 53 pM) and binding to the receptor (IC50, 170 pM), implicating tyrosine
phosphatase as a transducer of the growth inhibition signal in SSTR2-expressing
cells. In SSTR5-expressing cells, the phosphatase pathway was not involved in
the inhibitory effect of RC-160 on cell growth, since this action was not influenced
by tyrosine and serine/threonine phosphatase inhibitors. In addition, in SSTR5-expressing
cells, RC-160 inhibited CCK-stimulated intracellular calcium mobilization at doses
(EC50, 0.35 nM) similar to those necessary to inhibit somatostatin-14 binding
(IC50, 21 nM) and CCK-induced cell proliferation (EC50, 1.1 nM). This suggests
that the inositol phospholipid/calcium pathway could be involved in the antiproliferative
effect of RC-160 mediated by SSTR5 in these cells. RC-160 had no effect on the
basal or carbachol-stimulated calcium concentration in cells expressing SSTR1
to -4. Thus, we conclude that SSTR2 and SSTR5 bind RC-160 with high affinity and
mediate the RC-160-induced inhibition of cell growth by distinct mechanisms."
[Abstract/Full Text]
Kamal Sharma, Yogesh C. Patel, and Coimbatore B. Srikant
C-Terminal Region of Human Somatostatin Receptor 5 Is Required
for Induction of Rb and G1 Cell Cycle Arrest Mol. Endocrinol.
13: 82-90, 1999. "To determine whether the antiproliferative signaling
via these hSSTRs causes cell cycle arrest and to identify the molecular mediators
involved in this process, we evaluated the effect of SST in CHO-K1 cells expressing
hSSTRs 1, 2, 4, and 5 on cell cycle progression and induction of Rb and p21. We
report here that SST-induced G1 cell cycle arrest in these cells is due mainly
to the induction of Rb. Maximal effect was exerted via hSSTR5 followed by hSSTR
2, 4, and 1. In hSSTR5-expressing cells, a major portion of SST-induced Rb was
hypophosphorylated. SST-induced G1 arrest and induction of Rb were pertussis toxin
sensitive, G protein dependent, and protein tyrosine phosphatase (PTP) dependent.
In octreotide (OCT)-treated cells there was a redistribution of PTP activity from
the cytosol to the membrane. Mutational analysis of the C tail of this receptor
revealed that the C tail of the receptor is essential for PTP-dependent cytostatic
signaling." [Full
Text] Seungjoon Park, Jun Kamegai, Todd A. Johnson,
Lawrence A. Frohman, and Rhonda D. Kineman Modulation of Pituitary
Somatostatin Receptor Subtype (sst15) Messenger Ribonucleic Acid Levels
by Changes in the Growth Hormone Axis Endocrinology 141:
3556-3563, 2000. [Full
Text] Whitney W. Woodmansee, Rhonda L. Mouser,
David F. Gordon, Janet M. Dowding, William M. Wood, and E. Chester Ridgway
Mutational Analysis of the Mouse Somatostatin Receptor Type 5 Gene Promoter
Endocrinology 143: 2268-2276, 2002. "We have
previously characterized the structure of the murine somatostatin receptor type
5 gene (sst5). Initial transient transfection studies in pituitary somatolactotropes
(GH3) mapped the promoter activity of this gene to a region 290 bp upstream of
the transcription start site. The current study identifies the sst5 promoter region
critical for basal activity. A series of deletions was generated, and promoter
activity was localized to a region between -83 and -19. Similar promoter deletion
patterns were evident in five pituitary cell types. Seven 10-bp transversion mutations
encompassing the region between -83 and -19 were generated, and functional activity
was assessed. Promoter activity was reduced by the mutations spanning -67 to -47
compared with the wild-type construct. Another mutation between -26 and -17 resulted
in promoter activity reduction in GH3 cells, but not TtT-97 thyrotropes. Deoxyribonuclease
I protection analysis of the sst5 promoter region between -208/+47 was performed
using GH3 and TtT-97 nuclear extracts. The most striking protected regions, located
between -61 and -41 and -25 and -3, correlated with functionally important regions
identified by transfection studies. In summary, the mouse sst5 gene promoter has
been characterized, and functional activity and nuclear factor interactions were
mapped to two specific promoter regions. The region between -67 and -47 appears
to contain a nucleotide sequence critical for basal transcriptional regulation
of the mouse sst5 gene in pituitary cells." [Abstract]
Ujendra Kumar, Dale Laird, Coimbatore B. Srikant, Emanuel
Escher, and Yogesh C. Patel Expression of the Five Somatostatin
Receptor (SSTR1-5) Subtypes in Rat Pituitary Somatotrophes: Quantitative Analysis
by Double-Label Immunofluorescence Confocal Microscopy
Endocrinology 138: 4473-4476, 1997. "Using quantitative double-label
fluorescence immunocytochemistry and confocal microscopy, we have analysed the
pattern of expression of SSTR1-5 in normal rat pituitary somatotrophes. Antipeptide
rabbit polyclonal antibodies were produced against the extracellular domains of
SSTR1-5. SSTR antigens were colocalized in GH positive cells using rhodamine conjugated
secondary antibody for SSTRs and FITC-conjugated secondary antibody for GH. SSTR5
was the predominant subtype which was expressed in 86 ± 9.7% of GH cells
followed by SSTR2 in 42 ± 6.4% of GH positive cells. SSTR4 and SSTR3 were
modestly expressed in 23 ± 4.7% and 18 ± 3.2% of somatotrophes respectively
whereas SSTR1 was the least expressed subtype occurring in only 5 ± 1.2%
of somatotrophes. These results demonstrate variable expression of the 5 SSTRs
in somatotrophes. The preponderance of the SST-28 preferring SSTR5 subtype correlates
with the reported higher potency of SST-28 than SST-14 for inhibiting GH secretion."
[Abstract]
Stroh T, Kreienkamp HJ, Beaudet A. Immunohistochemical
distribution of the somatostatin receptor subtype 5 in the adult rat brain: predominant
expression in the basal forebrain. J Comp Neurol 1999 Sep
13;412(1):69-82 "Somatostatin exerts its actions by means of a family
of G protein-coupled receptors, five of which have so far been cloned. Whereas
mRNAs for receptor subtypes sst(1)-sst(4) have been unequivocally localized in
the brain, the data concerning the fifth subtype, sst(5), are contradictory. Moreover,
whereas sst(1) and sst(2A) receptor proteins have been localized by immunohistochemistry,
the distribution of sst(3)-sst(5) receptor proteins and/or subtype-specific binding
remains to be determined in the central nervous system. In the present study,
we investigated the distribution of immunoreactive sst(5) in adult rat brain and
pituitary and demonstrated the presence of this receptor protein in the central
nervous system by using an affinity-purified antibody generated against the C-terminus
of the receptor. The specificity of the antibody for sst(5) was established by
immunoblotting experiments on membranes prepared from cells transfected with cDNA
encoding different somatotropin release inhibiting (SRIF) receptor subtypes as
well as from anterior pituitary. In both systems, the antibody specifically recognized
a band at approximately 50 kDa molecular mass, corresponding well to the reported
size of the cloned receptor (48 kDa). Immunofluorescence in COS-7 cells transfected
with individual SRIF receptor subtypes as well as in sections of rat pituitary
demonstrated the antibody's applicability to the immunohistochemical detection
of sst(5) receptors. In rat brain sections, sst(5) immunoreactivity was predominantly
associated with neuronal perikarya and primary dendrites. Immunolabeling was most
prominent in the olfactory tubercle, islands of Calleja, diagonal band of Broca,
substantia innominata, and magnocellular preoptic nucleus of the basal forebrain
as well as in the reticular nucleus of the thalamus. Other, less intensely labeled
areas included the cerebral cortex, hippocampus, amygdala, preoptic area as well
as the lateroanterior nucleus of the hypothalamus. The present findings provide
the first characterization of the anatomic distribution of sst(5) receptors in
the rat brain. They demonstrate a prominent expression of these receptors in the
basal forebrain, suggesting that they may be involved in the mediation of somatostatin
effects on the sleep-wake cycle through their association with cortically projecting
subcortical systems. Copyright 1999 Wiley-Liss, Inc." [Abstract] Akbar
M, Okajima F, Tomura H, Majid MA, Yamada Y, Seino S, Kondo Y. Phospholipase
C activation and Ca2+ mobilization by cloned human somatostatin receptor subtypes
1-5, in transfected COS-7 cells. FEBS Lett 1994 Jul 11;348(2):192-6
"We transfected the COS-7 cells with cDNAs encoding different human somatostatin
receptor (hSSTR) subtypes, and found that hSSTR subtypes mediate not only the
inhibition of forskolin-induced cAMP accumulation but also the stimulation of
phospholipase C (PLC) and Ca2+ mobilization. Activation of PLC by 1 microM somatostatin
(SRIF) was in the order of: hSSTR5 > hSSTR2 > hSSTR3 > hSSTR4 >>
hSSTR1. Pertussis toxin (PTX) treatment completely or partially reversed the PLC
activation. 1 nM SRIF was equally effective for adenylate cyclase (AC) inhibition
in a PTX-sensitive manner, in all the cells expressing different hSSTRs, except
for hSSTR1. Nevertheless, SRIF stimulated AC even in the presence of forskolin
at higher doses of SRIF in PTX-treated hSSTR5-expressing cells. We conclude that
the cloned hSSTRs differentially couple to PTX-sensitive and -insensitive G-proteins
to modulate PLC, Ca2+ mobilization and AC." [Abstract] Philippe
Sarret, Dominique Nouel, Claude Dal Farra, Jean-Pierre Vincent, Alain Beaudet,
and Jean Mazella Receptor-mediated Internalization Is Critical
for the Inhibition of the Expression of Growth Hormone by Somatostatin in the
Pituitary Cell Line AtT-20 J. Biol. Chem. 274: 19294-19300,
July 1999. "The inhibitory effect of the neuropeptide somatostatin on
the expression of growth hormone was measured by quantitative polymerase chain
reaction in the pituitary cell line AtT-20. We demonstrate that this effect is
dependent on the internalization of somatostatin-receptor complexes and that it
is totally independent from the peptide-induced inhibition of adenylate cyclase.
Indeed, the inhibitory effect of the peptide on growth hormone mRNA levels was
totally insensitive to pertussis toxin treatment but was totally abolished under
conditions which block somatostatin receptor internalization. Comparative confocal
microscopic imaging of fluorescent somatostatin sequestration and fluorescence
immunolabeling of sst1, sst2A, and sst5 receptors suggests that sst2A is most
probably responsible of the inhibitory effect of somatostatin on growth hormone
expression." [Full
Text] Lamberts, Steven W.J., van der Lely,
Aart-Jan, de Herder, Wouter W., Hofland, Leo J. Octreotide
N Engl J Med 1996 334: 246-254 "The clinical introduction of somatostatin
analogues has resulted in new insights into the physiology of a number of organ
systems. Despite the broad range of physiologic actions of somatostatin, its analogues
control hormonal hypersecretion by somatostatin-receptorpositive endocrine
tumors without serious side effects. The subtypes of somatostatin receptors on
these tumors may turn over at a different rate from those on normal tissue. Therefore,
the inhibitory effects of octreotide on hormonal hypersecretion by endocrine tumors
persist, whereas adaptation occurs in most normal somatostatin target tissues,
thus preventing serious side effects." [Full
Text] | Rocheville
M, Lange DC, Kumar U, Patel SC, Patel RC, Patel YC. Receptors for
dopamine and somatostatin: formation of hetero-oligomers with enhanced functional
activity. Science 2000 Apr 7;288(5463):154-7 "Somatostatin
and dopamine are two major neurotransmitter systems that share a number of structural
and functional characteristics. Somatostatin receptors and dopamine receptors
are colocalized in neuronal subgroups, and somatostatin is involved in modulating
dopamine-mediated control of motor activity. However, the molecular basis for
such interaction between the two systems is unclear. Here, we show that dopamine
receptor D2R and somatostatin receptor SSTR5 interact physically through hetero-oligomerization
to create a novel receptor with enhanced functional activity. Our results provide
evidence that receptors from different G protein (heterotrimeric guanine nucleotide
binding protein)-coupled receptor families interact through oligomerization. Such
direct intramembrane association defines a new level of molecular crosstalk between
related G protein-coupled receptor subfamilies." [Abstract] Jeffrey
C. Liu, Ross E. Baker, Clement Sun, Valdine C. Sundmark, and Harry P. Elsholtz
Activation of Go-coupled Dopamine D2 Receptors Inhibits ERK1/ERK2
in Pituitary Cells. A KEY STEP IN THE TRANSCRIPTIONAL SUPPRESSION
OF THE PROLACTIN GENE J. Biol. Chem. 277: 35819-35825,
September 2002. "In pituitary lactotrophs the prolactin gene is stimulated
by neuropeptides and estrogen and is suppressed by dopamine via D2-type receptors.
Stimulatory signals converge on activation of the mitogen-activated protein kinases
ERK1/2, but dopamine regulation of this pathway is not well defined. Paradoxically,
D2 agonists activate ERK1/2 in many cell types. Here we show that in prolactin-secreting
GH4ZR7 cells and primary pituitary cells, dopamine treatment leads to a rapid,
pronounced, and specific decrease in activated ERK1/2. The response is blocked
by D2-specific antagonists and pertussis toxin. Interestingly, in stable lines
expressing specific pertussis toxin-resistant G subunits, toxin treatment blocks
dopamine suppression of MAPK in Gi2- but not Go-expressing
cells, demonstrating that Go-dependent pathways can effect the inhibitory MAPK
response. At the nuclear level, the MEK1 inhibitor U0126 mimics the D2-agonist
bromocryptine in suppressing levels of endogenous prolactin transcripts. Moreover,
a good correlation is seen between the IC50 values for inhibition of MEK1 and
suppression of prolactin promoter function (PD184352 > U0126 > U0125). Both
dopamine and U0126 enhance the nuclear localization of ERF, a MAPK-sensitive ETS
repressor that inhibits prolactin promoter activity. In addition, U0126 suppression
is transferred by tandem copies of the Pit-1-binding site, consistent with mapping
experiments for dopamine responsiveness. Our data suggest that ERK1/2 suppression
is an obligatory step in the dopaminergic control of prolactin gene transcription
and that bidirectional control of ERK1/2 function in the pituitary may provide
a key mechanism for endocrine gene control." [Abstract]
Ramesh C. Patel, Ujendra Kumar, Don C. Lamb, John S. Eid,
Magalie Rocheville, Michael Grant, Aruna Rani, Theodore Hazlett, Shutish C. Patel,
Enrico Gratton, and Yogesh C. Patel Ligand binding to somatostatin
receptors induces receptor-specific oligomer formation in live cells
PNAS 99: 3294-3299, March 2002. "Heptahelical receptors (HHRs) are generally
thought to function as monomeric entities. Several HHRs such as somatostatin receptors
(SSTRs), however, form homo- and heterooligomers when activated by ligand binding.
By using dual fluorescent ligands simultaneously applied to live cells monotransfected
with SSTR5 (R5) or SSTR1 (R1), or cotransfected with R5 and R1, we have analyzed
the ligand receptor stoichiometry and aggregation states for the three receptor
systems by fluorescence resonance energy transfer and fluorescence correlation
spectroscopy. Both homo- and heterooligomeric receptors are occupied by two ligand
molecules. We find that monomeric, homooligomeric, and heterooligomeric receptor
species occur in the same cell cotransfected with two SSTRs, and that oligomerization
of SSTRs is regulated by ligand binding by a selective process that is restricted
to some (R5) but not other (R1) SSTR subtypes. We propose that induction by ligand
of different oligomeric states of SSTRs represents a unique mechanism for generating
signaling specificity not only within the SSTR family but more generally in the
HHR family." [Full
Text] Komatsuzaki K, Terashita K, Kinane
TB, Nishimoto I. Somatostatin type V receptor activates c-Jun N-terminal
kinases via Galpha(12) family G proteins. Biochem Biophys
Res Commun 2001 Dec 21;289(5):1211-7 "Somatostatin is a neurotransmitter
with diverse effects including anti-proliferation in a wide range of normal and
neoplastic cells, and occasionally growth stimulatory and neurotrophic actions.
Stress-activated protein kinase or c-Jun N-terminal kinase (SAPK/JNK) can also
induce growth arrest and occasionally growth stimulation. However, the relationship
between somatostatin and SAPK/JNK is less clear. Here we report that the binding
of somatostatin to the somatostatin receptor type V (SSTR5) upregulates SAPK/JNK
activity. We also show that this activation is mediated by Galpha(12) and Galpha(13).
This study demonstrates that SSTR5 is the heptahelical receptor that activates
SAPK/JNK via the G(12) family G proteins." [Abstract] Saveanu
A, Morange-Ramos I, Gunz G, Dufour H, Enjalbert A, Jaquet P. A luteinizing
hormone-, alpha-subunit- and prolactin-secreting pituitary adenoma responsive
to somatostatin analogs: in vivo and in vitro studies.
Eur J Endocrinol 2001 Jul;145(1):35-41 "RESULTS: This adenoma presented
with high levels of SSTR2, SSTR3 and SSTR5 mRNAs, as compared with a series of
gonadotroph adenomas. In cell culture studies, PRL, LH and alpha-subunit were
inhibited by 60%, 47% and 33% respectively by SRIF-14 at a concentration of 10
nmol/l. The SSTR2 (BIM-23197, lanreotide) and SSTR5 (BIM-23268) preferential analogues
both produced a partial 21-38% inhibition of PRL, LH, and alpha-subunit release.
DISCUSSION: In this plurihormonal-secreting adenoma, the high efficacy of somatostatin
analogues to inhibit PRL, LH and alpha-subunit secretion in vivo may be explained
by the unusually high level of expression and by the functionality of both SSTR2
and SSTR5 receptor subtypes." [Abstract] Patel
YC. Somatostatin and its receptor family.
Front Neuroendocrinol 1999 Jul;20(3):157-98 "Somatostatin (SST), a regulatory
peptide, is produced by neuroendocrine, inflammatory, and immune cells in response
to ions, nutrients, neuropeptides, neurotransmitters, thyroid and steroid hormones,
growth factors, and cytokines. The peptide is released in large amounts from storage
pools of secretory cells, or in small amounts from activated immune and inflammatory
cells, and acts as an endogenous inhibitory regulator of the secretory and proliferative
responses of target cells that are widely distributed in the brain and periphery.
These actions are mediated by a family of seven transmembrane (TM) domain G-protein-coupled
receptors that comprise five distinct subtypes (termed SSTR1-5) that are endoded
by separate genes segregated on different chromosomes. The five receptor subtypes
bind the natural SST peptides, SST-14 and SST-28, with low nanomolar affinity.
Short synthetic octapeptide and hexapeptide analogs bind well to only three of
the subtypes, 2, 3, and 5. Selective nonpeptide agonists with nanomolar affinity
have been developed for four of the subtypes (SSTR1, 2, 3, and 4) and putative
peptide antagonists for SSTR2 and SSTR5 have been identified. The ligand binding
domain for SST ligands is made up of residues in TMs III-VII with a potential
contribution by the second extracellular loop. SSTRs are widely expressed in many
tissues, frequently as multiple subtypes that coexist in the same cell. The five
receptors share common signaling pathways such as the inhibition of adenylyl cyclase,
activation of phosphotyrosine phosphatase (PTP), and modulation of mitogen-activated
protein kinase (MAPK) through G-protein-dependent mechanisms. Some of the subtypes
are also coupled to inward rectifying K(+) channels (SSTR2, 3, 4, 5), to voltage-dependent
Ca(2+) channels (SSTR1, 2), a Na(+)/H(+) exchanger (SSTR1), AMPA/kainate glutamate
channels (SSTR1, 2), phospholipase C (SSTR2, 5), and phospholipase A(2) (SSTR4).
SSTRs block cell secretion by inhibiting intracellular cAMP and Ca(2+) and by
a receptor-linked distal effect on exocytosis. Four of the receptors (SSTR1, 2,
4, and 5) induce cell cycle arrest via PTP-dependent modulation of MAPK, associated
with induction of the retinoblastoma tumor suppressor protein and p21. In contrast,
SSTR3 uniquely triggers PTP-dependent apoptosis accompanied by activation of p53
and the pro-apoptotic protein Bax. SSTR1, 2, 3, and 5 display acute desensitization
of adenylyl cyclase coupling. Four of the subtypes (SSTR2, 3, 4, and 5) undergo
rapid agonist-dependent endocytosis. SSTR1 fails to be internalized but is instead
upregulated at the membrane in response to continued agonist exposure. Among the
wide spectrum of SST effects, several biological responses have been identified
that display absolute or relative subtype selectivity. These include GH secretion
(SSTR2 and 5), insulin secretion (SSTR5), glucagon secretion (SSTR2), and immune
responses (SSTR2). Copyright 1999 Academic Press." [Abstract] Tulipano
G, Bonfanti C, Milani G, Billeci B, Bollati A, Cozzi R, Maira G, Murphy WA, Poiesi
C, Turazzi S, Giustina A. Differential inhibition of growth hormone
secretion by analogs selective for somatostatin receptor subtypes 2 and 5 in human
growth-hormone-secreting adenoma cells in vitro. Neuroendocrinology
2001 May;73(5):344-51 "We conclude that the availability of somatostatin
analogs selective for SSTR5 will enhance the treatment potency and spectrum in
acromegaly." [Abstract] Rohrer
SP, Schaeffer JM. Identification and characterization of subtype
selective somatostatin receptor agonists. J Physiol Paris
2000 May-Aug;94(3-4):211-5 "High affinity, subtype selective non-peptide
agonists of somatostatin receptor subtypes 1-5 were identified in combinatorial
libraries constructed based on molecular modeling of known peptide agonists. Simultaneous
traditional chemical synthesis yielded an additional series of somatostatin subtype-2
receptor (SSTR2) selective agonists. These compounds have been used to further
define the physiological functions of the individual somatostatin receptor subtypes.
In vitro experiments demonstrated the role of the SSTR2 in inhibition of glucagon
release from mouse pancreatic alpha-cells and the somatostatin subtype-5 receptor
(SSTR5) as a mediator of insulin secretion from pancreatic beta-cells. Both SSTR2
and SSTR5 regulated growth hormone release from the rat anterior pituitary gland.
In vivo studies performed with SSTR2 receptor selective compounds demonstrated
effective inhibition of pulsatile growth hormone release in rats. The SSTR2 selective
compounds also lowered plasma glucose levels in normal and diabetic animal models.
The availability of high affinity, subtype selective non-peptide agonists for
each of the somatostatin receptors provides a direct approach to defining their
physiological function both peripherally and in the central nervous system."
[Abstract] Suich
DJ, Mousa SA, Singh G, Liapakis G, Reisine T, DeGrado WF. Template-constrained
cyclic peptide analogues of somatostatin: subtype-selective binding to somatostatin
receptors and antiangiogenic activity. Bioorg Med Chem
2000 Sep;8(9):2229-41 "Template-constrained cyclic peptides in which
the ends of the -Tyr-D-Trp-Lys-Val-tetrapeptide were linked by scaffolds based
on either an N,N'-dimethyl-N,N'-diphenylurea or a substituted biphenyl system
(DJS631 and DJS811, respectively), bound selectively to mouse SSTR2B and rat and
human SSTR5 with affinities as high as 1 nM. DJS811, at a dose of 3 mg/kg/day,
was shown in a mouse Matrigel model to inhibit angiogenesis to a level of 79%.
The development of structured turn scaffolds allows beta-turn sequences to be
contained in the context of a compact structure, with less peptidic nature and
potentially greater bioavailability than cyclic hexapeptides. These systems can
be used to study the determinants of beta-turn formation, as well as to probe
the importance of turn sequences occurring in molecular recognition interactions.
The antiangiogenic activity of DJS811 suggests that it may have antitumor activity
as well. In addition, because SSTR2 is overexpressed on many types of tumors,
DJS631 and DJS811 may be useful in the development of agents for tumor imaging
or the radiotherapy of cancer." [Abstract] Emilia
Ballarè, Luca Persani, Andrea G. Lania, Marcello Filopanti, Enza Giammona,
Sabrina Corbetta, Simona Mantovani, Maura Arosio, Paolo Beck-Peccoz, Giovanni
Faglia, and Anna Spada Mutation of Somatostatin Receptor Type 5
in an Acromegalic Patient Resistant to Somatostatin Analog Treatment
J. Clin. Endocrinol. Metab. 86: 3809-3814, 2001. "Introduction of somatostatin
analogs has greatly contributed to improving the prognosis of acromegaly. Although
the majority of patients are effectively treated by these agents, resistance occurs
in a subset of patients. So far, resistance to somatostatin has never been associated
with mutations of the somatostatin receptor subtypes (sst2 and sst5) that inhibit
GH secretion. Molecular analysis of genomic DNA from pituitary tumor and peripheral
blood obtained from an acromegalic resistant to octreotide showed a somatic activating
mutation of Gs (Arg201Cys), no mutation in sst2, and one polymorphism (Pro109Ser)
and one germ line mutation (Arg240Trp) in sst5. Wild-type (WT) and mutant sst5
PCR products were cloned and transfected into Chinese hamster ovary K1 cells.
In Chinese hamster ovary K1 cells stably expressing mutant sst5, somatostatin-28
was less potent in inhibiting cyclic AMP levels than in WT cells. Proliferation
of mutant cells exceeded that of WT by 50%. Moreover, somatostatin reduced cell
growth and MAPK activity in WT but not in mutant cells in which the peptide even
increased MAPK activity. We suggest that this mutation that abrogates the antiproliferative
action of somatostatin and activates mitogenic pathways may be involved in the
resistance to somatostatin treatment." [Full
Text] S. Petersenn, A. C. Rasch, C. Böhnke,
and H. M. Schulte Identification of an Upstream Pituitary-Active
Promoter of Human Somatostatin Receptor Subtype 5 Endocrinology
143: 2626-2634, 2002. "Somatostatin receptor subtype 5 (sst5) has been
linked to inhibition of PRL and insulin secretion. We characterized the genomic
structure of the human sst5. The transcription start site was located 94 nucleotides
upstream of the initiator ATG codon. Sequence analysis of 5'-inverse PCR products
revealed the presence of a 6.1-kb intron in the 5'-untranslated region. RT-PCR
analysis indicated tissue-specific activation of the newly identified upstream
promoter in pituitary, but not in small intestine, lung, or placenta. A -1741
promoter directed significant levels of luciferase expression in GH4 rat pituitary
cells, Skut-1B endometrium cells, and JEG3 chorion carcinoma cells, which was
absent in COS-7 monkey kidney cells. A minimal -101 promoter was sufficient to
allow tissue-specific expression. Its activity in COS-7 cells was not enhanced
by cotransfection of the pituitary-specific transcription factor Pit-1. Analysis
of deletion constructs revealed a GC-rich region immediately upstream of the transcription
start site, which is necessary for promoter activity. Somatostatin led to a significant
inhibition, and forskolin and thyroid hormone to a significant stimulation of
pituitary-specific promoter activity. Further mapping suggested a cAMP-responsive
element located between -101 and the transcription start site, and thyroid hormone-responsive
elements between -1741 and -1269 and between -317 and -101. These studies identified
an upstream promoter of the sst5 gene with tissue-specific activity." [Abstract]
Freeman, Marc E., Kanyicska, Bela, Lerant, Anna, Nagy, Gyorgy
Prolactin: Structure, Function, and Regulation of Secretion
Physiol. Rev. 2000 80: 1523-1631 [Full
Text] Nedim Hukovic, Rosemarie Panetta, Ujendra
Kumar, Magalie Rocheville, and Yogesh C. Patel The Cytoplasmic
Tail of the Human Somatostatin Receptor Type 5 Is Crucial for Interaction with
Adenylyl Cyclase and in Mediating Desensitization and Internalization
J. Biol. Chem. 273: 21416-21422, August 1998. [Full
Text] Thomas Stroh, Alexander C. Jackson,
Philippe Sarret, Claude Dal Farra, Jean-Pierre Vincent, Hans-Jürgen Kreienkamp,
Jean Mazella, and Alain Beaudet Intracellular Dynamics of sst5
Receptors in Transfected COS-7 Cells: Maintenance of Cell Surface Receptors during
Ligand-Induced Endocytosis Endocrinology 141: 354-365,
2000. [Full
Text] Martinez V, Coy DH, Lloyd KC, Tache Y.
Intracerebroventricular injection of somatostatin sst5 receptor agonist
inhibits gastric acid secretion in rats. Eur J Pharmacol
1996 Jan 25;296(2):153-60 "Somatostatin and its analogs act in the brain
to influence gastric acid secretion. Five different somatostatin receptor subtypes
have been characterized (sst1 to sst5). We studied the influence of somatostatin
(0.18-0.6 nmol/rat) and selective sst2, sst3 and sst5 receptor ligands on basal
gastric acid secretion in conscious rats equipped with chronic gastric and intracerebroventricular
(i.c.v.) cannulae. Somatostatin-14 (0.36 nmol/rat), the sst2, sst3 and sst5 receptor
agonist, Des-AA1,2,4,5,12,13-[D-Tryp8,D-Cys14]somatostatin (SMS 201-995) (0.18-0.36
nmol/rat) and the sst5 receptor agonist, BIM-23052, (0.8-1.2 nmol/rat) injected
i.c.v. inhibited gastric acid secretion. Maximal inhibition reaching 42%, 60%
and 42% was induced by somatostatin-14 (0.36 nmol/rat), SMS 201-995 (0.18 nmol/rat)
and BIM-23052 (0.8 nmol/rat) respectively. The sst2 receptor agonist, DC 32-87
(0.2-0.8 nmol/rat) and sst3 receptor agonist, BIM-23056 (0.2-1.2 nmol/rat), did
not modify gastric acid secretion, except the sst3 receptor agonist at 0.4 nmol/rat
which increased acid output at 20 min post-injection. The sst2 receptor agonists
(0.4 nmol/rat) co-injected i.c.v. with a subthreshold dose of sst5 (0.4 nmol/rat)
inhibited gastric acid secretion. These results show that i.c.v. injection of
somatostatin-14 inhibits basal gastric acid secretion in conscious rats through
an action on sst5 receptor subtype which can be potentiated by sst2 receptor subtype."
[Abstract] Fagan
SP, Azizzadeh A, Moldovan S, Ray MK, Adrian TE, Ding X, Coy DH, Brunicardi FC.
Insulin secretion is inhibited by subtype five somatostatin receptor
in the mouse. Surgery 1998 Aug;124(2):254-8; discussion
258-9 "BACKGROUND: Recently five somatostatin receptor subtypes (SSTRs)
were cloned, allowing the development of highly specific agonists to these SSTRs.
Previous studies have shown a species specificity phenomenon with respect to the
inhibition of insulin secretion by these selective agonists. This study was undertaken
to determine which SSTR (2 or 5) is responsible for the inhibitory effect of somatostatin
on glucose-stimulated mouse insulin secretion. METHODS: Intact mouse islets (n
= 10) were stimulated with D-glucose in the presence or absence of receptor-specific
somatostatin agonists. RESULTS: D-glucose (16.7 mmol/L) augmented insulin secretion
by 158% above that seen with 3.9 mmol/L D-glucose. In the presence of DC 32-92
(SSTR5) selective agonist, D-glucose (16.7 mmol/L) augmented insulin secretion
by 64% above that seen with 3.9 mmol/L D-glucose. The presence of SSTR 5 selective
agonist resulted in a significant (P < .05) inhibition of glucose-stimulated
insulin secretion. The identification of SSTR5 within the mouse pancreas was established
by reverse transcriptase polymerase chain reaction and confirmed by Southern blot
analysis. CONCLUSIONS: These results suggest that the inhibitory effect of somatostatin
on insulin secretion is mediated through the subtype 5 receptor within the mouse
islet." [Abstract] Chisholm
C, Greenberg GR. Somatostatin-28 regulates GLP-1 secretion via somatostatin
receptor subtype 5 in rat intestinal cultures. Am J Physiol
Endocrinol Metab 2002 Aug;283(2):E311-7 "Five somatostatin receptors
(SSTRs) bind somatostatin-14 (S-14) and somatostatin-28 (S-28), but SSTR5 has
the highest affinity for S-28. To determine whether S-28 acting through SSTR5
mediates inhibition of glucagon-like peptide-1 (GLP-1), fetal rat intestinal cell
cultures were treated with somatostatin analogs with relatively high specificity
for SSTRs 2-5. S-28 dose-dependently inhibited GLP-1 secretion stimulated by gastrin-releasing
peptide more potently than S-14 (EC50 0.01 vs. 5.8 nM). GLP-1 secretion was inhibited
by an SSTR5 analog, BIM-23268, more potently than S-14 and nearly as effectively
as S-28. The SSTR5 analog L-372,588 also suppressed GLP-1 secretion equivalent
to S-28, but a structurally similar peptide, L-362,855 (Tyr to Phe at position
7), was ineffective. An SSTR2-selective analog was less effective than S-28, and
an SSTR3 analog was inactive. Separate treatment with GLP-1-(7-36)-NH2 increased
S-28 and S-14 secretion by three- and fivefold; BIM-23268 abolished S-28 without
altering S-14, whereas the SSTR2 analog was inactive. The results indicate that
somatostatin regulation of GLP-1 secretion occurs via S-28 through activation
of SSTR5. GLP-1-stimulated S-28 secretion is also autoregulated by SSTR5 activation,
suggesting a feedback loop between GLP-1 and S-28 modulated by SSTR5." [Abstract] Zatelli
MC, Tagliati F, Taylor JE, Piccin D, Culler MD, Degli Uberti EC. Somatostatin,
but not Somatostatin Receptor Subtypes 2 and 5 Selective Agonists, Inhibits Calcitonin
Secretion and Gene Expression in the Human Medullary Thyroid Carcinoma Cell Line,
TT. Horm Metab Res 2002 May;34(5):229-33 "Somatostatin
(SRIH) analogs are commonly used to treat symptoms in medullary thyroid carcinoma
(MTC), that expresses SRIH receptors (SSTR1 to SSTR5), as does the human MTC cell
line TT. The aim of this work was to evaluate whether SRIH, SSTR2 and SSTR5-selective
agonists influence calcitonin (CT) secretion and gene expression in the TT cell
line. CT secretion was evaluated by chemiluminescence, and gene expression was
analyzed by Northern blot. TT cell line proliferation was also assessed by [ (3)H]
thymidine ([ (3)H]thy) incorporation and viable cell number count. SRIH significantly
(p < 0.05) reduced [ (3)H]thy incorporation (approx. 50 %), viable cell number
(approx. 20 %), CT secretion (- 30 %) and CT gene expression (approx. 2-fold).
Exposure to the SSTR2-selective agonist, BIM-23 120, and to the SSTR5-selective
agonist, BIM-23 206, did not modify CT secretion and mRNA levels in TT cells.
Thus, SRIH inhibits DNA synthesis, cell proliferation, CT secretion and CT gene
expression in the TT cell line, while SSTR2 and 5 selective agonists, although
influencing DNA synthesis and cell proliferation, do not modify CT gene expression,
suggesting that SRIH may influence gene expression acting through SSTRs other
than subtypes 2 and 5. Furthermore, these findings may explain the erratic response
of MTC patients in terms of CT plasma levels to treatment with SRIH analogs, like
octreotide and lanreotide, which interact mainly with SSTR2 and 5." [Abstract]
|