Figure 3 - uploaded by Jean Rosenbaum
Content may be subject to copyright.
Immunohistochemistry of somatostatin receptors (SSTRs) in hepatocellular carcinoma. (A) Negative control. (B–F) Double staining immunohistochemistry with a smooth muscle actin ( a -SMA) and SSTR1–5 in the same patient. We observed intense immunoreaction with SSTR1–4, and to a lesser degree with SSTR5. Within the tumour, double positive cells were seen (arrow, B insert). These cells are clearly activated hepatic stellate cells. Original magnification 100 6 , inserts 400 6 . 

Immunohistochemistry of somatostatin receptors (SSTRs) in hepatocellular carcinoma. (A) Negative control. (B–F) Double staining immunohistochemistry with a smooth muscle actin ( a -SMA) and SSTR1–5 in the same patient. We observed intense immunoreaction with SSTR1–4, and to a lesser degree with SSTR5. Within the tumour, double positive cells were seen (arrow, B insert). These cells are clearly activated hepatic stellate cells. Original magnification 100 6 , inserts 400 6 . 

Source publication
Article
Full-text available
Somatostatin analogues have been used with conflicting results to treat advanced hepatocellular carcinoma (HCC). The aim of this study was to investigate expression of somatostatin receptor (SSTR) subtypes in human liver, and to examine the effect of selective SSTR agonists on proliferation, apoptosis, and migration of hepatoma cells (HepG2, HuH7)...

Contexts in source publication

Context 1
... by automatic sequencing using the ABI prism 310 (Perkin Elmer). To elucidate effects of somatostatin on tumour progression, SSTR subtype specific agonists were used: L-797,591 (SSTR1 agonist), L-779,976 (SSTR2 agonist), L-796,778 (SSTR3 agonist), L-803,087 (SSTR4 agonist), and L-817,818 (SSTR5 agonist) (Merck Research Laboratories, Rahway, New Jersey, USA). These substances are non-peptide somatostatin agonists with a very high subtype specificity, except for L-817,818 which has affinity not only for SSTR5 but also some for SSTR1. Although these substances have been used in animal cells, they were designed and developed as agonists for human SSTRs. The affinity of the agonists was tested in transfected CHO-K1 cells expressing each of the five human SSTRs. 18 In addition to their specificity, these agonists have a prolonged half life. Biological effects of the agonists has been tested in several cell lines and these effects were SSTR subtype specific. 19 Cell proliferation was measured by using the colorimetric 5-bromo-2 9 deoxyuridine (BrdU) cell proliferation ELISA (Boehringer, Mannheim, Germany), as described previously. 20 After trypsinisation, HepG2, HuH7, and HSCs were plated in 96 well dishes (Falcon, Becton Dickinson Labware, Meylan, France) at a density of 15 6 10 3 , 15 6 10 3 , and 6 6 10 3 cells per well, respectively. All cells were suspended in DMEM supplemented with 2% fetal calf serum. After 24 hours, media were renewed and cells exposed for another 24 hours to different concentrations of SSTR agonists (10 2 12 to 10 2 8 mol/l) in the presence of 10 m mol BrdU. Platelet derived growth factor (PDGF-BB 10 ng/ml), a strong mitogen for HSCs, 21 was added to the medium of HSCs (R & D Systems Europe, Abingdon, UK). In each experiment, all conditions were tested 10 times, and every experiment was performed in triplicate. To allow comparison of proliferation between different groups, results were expressed as percentages and compared with controls normalised to 100%. Mean (SD) values were calculated for all groups. Somatostatin induced apoptosis was studied by the TUNEL reaction according to the manufacturer’s instructions (In Situ Cell Death Detection Kit; Roche diagnostics, Belgium). After trypsinisation, HepG2, HuH7, and HSCs were plated in 96 well dishes at a density of 2.5 6 10 3 , 2.5 6 10 3 , and 1 6 10 3 cells per well, respectively. All cells were suspended in DMEM supplemented with 2% fetal calf serum. As apoptosis has been attributed to stimulation of SSTR2 and 3, different concentrations (10 2 12 to 10 2 6 mol/l) of L-779,976 and L-796,778, agonists of SSTR 2 and 3, respectively, were studied. After 24 and 72 hours of treatment, apoptotic cells were counted and expressed as a percentage of total cells. Experiments were performed in triplicate. The influence of the five SSTR agonists on migration of the hepatoma cell lines and HSCs was investigated, as described previously, 22 using Boyden invasion chambers with 8 m m pore size filters coated with Matrigel basement membrane matrix (Falcon, Becton Dickinson Labware, Bedford, Massachusetts, USA). PDGF-BB is a very strong chemoattractant for human HSCs 21 whereas hepatocyte growth factor (HGF) increases the invasiveness of HepG2 and HuH7 tumour cells. 22 To investigate the possibility that somatostatin inhibits PDGF- BB induced migration of HSCs or HGF induced migration of HepG2 and HuH7, cells were seeded in the upper well of Boyden chambers in the presence of PDGF-BB or HGF in the lower chamber. In these experiments, 2.5 6 10 4 hepatoma cells or HSCs were seeded onto the filter. HGF 20 ng/ml (R & D Systems Europe, Abingdon, UK) was added to the lower chamber when hepatoma cells were seeded in the upper chamber, or PDGF-BB 10 ng/l was added when HSCs were present in the upper chamber. Subsequently, 10 2 9 mol/l of SSTR agonists were added to the cells in the upper chamber. After 12 and 48 hours for experiments with HSCs and hepatoma cells, respectively, cells in the upper chamber were wiped with a cotton swab, and the filters were fixed in methanol and stained with haematoxylin. Cells invading the lower surface of the filter were counted in 10 high power fields using a Zeiss Axioplan microscope. Results were expressed as a percentage, relative to controls normalised to 100%. Experiments were performed in triplicate, except for controls and experiments with L-797,591 (agonist of SSTR1) which were performed six times. One way analysis of variance (ANOVA) with Bonferroni’s correction for multiple comparisons and the Mann-Whitney test were used when appropriate. Results are presented as means (SD). A p value of , 0.05 was considered statistically significant. Analysis was performed using Instat software (GraphPad software inc., San Diego, California, USA). Using well validated specific antibodies against the five recognised SSTRs, we were able to localise the different SSTRs in sections of normal liver (fig 1), cirrhosis (fig 2), and HCC (fig 3). Expression of the various receptors varied between different specimens and cells (table 3). In fig 1, normal liver is presented. Hepatocytes and sinusoidal cells did not stain for any of the five receptors. Cholangiocytes, on the other hand, consistently expressed SSTR subtypes 1, 3, and 4. Immunoreaction with SSTR subtypes 1 and 4 was more intense than with SSTR subtypes 2, 3, and 5 which were detected in, respectively, two, three, and one of the three analysed specimen. Furthermore, a weak immunoreaction of SSTR1 with endothelial cells of a small number of small arteries was found (not shown). In cirrhotic liver, expression of SSTRs significantly changed compared with normal liver. Hepatocytes within cirrhotic nodules expressed SSTR1 (fig 2B, C), and in 4/6 of the examined specimens, specimens also expressed SSTR2 (fig 2F, G), SSTR3 (fig 2J, K), SSTR4 (fig 2N, O), and SSTR5 (fig 2R, S). In most of the studied cases, we also found cells which double stained for all five SSTRs and a -SMA. These cells, clearly activated HSCs, were located at the interface of septa and cirrhotic nodules (fig 2C, G, K, O). In some specimens, we observed HSCs within hepatic nodules that stained for PDGFR- b and SSTR1 (fig 2D) or SSTR3 (fig 2L). In fig 3, immunohistochemistry for HCC is presented. Most of the tumours stained with SSTR subtypes 1 and 2 whereas 50% reacted with SSTR subtypes 3, 4, and 5 (table 3). In some tumours, non-tumoral cells double stained for a -SMA and SSTR subtypes 1, 2, and 3 (fig 3B, table 3). To verify the results of double staining immunohistochemistry, isolated cell lines were exposed to the different SSTR antibodies. Human HSCs were positive for all five SSTRs although immunoreaction with SSTR3 was less intense. Similarly, HepG2 expressed the five SSTR subtypes, again less clearly SSTR3. Finally, HuH7 reacted with all SSTRs, except for SSTR3 (data not shown). Messenger RNA of the five SSTRs was detected both in normal and pathological liver tissue (fig 4). We identified a single band for all receptors, except SSTR4 for which three bands were found in some patients. Sequencing demonstrated that the middle band (635 nucleotides), which was most intense, corresponded to SSTR4. This was in agreement with the findings of Talme et al who used the same primers for SSTR4. 23 All investigated samples were definitely positive for SSTR subtypes 1 and 2. For these receptors, no important variations between different patients or between cirrhosis and tumour samples from the same patient were observed. Expression of mRNA of SSTR subtypes 3, 4, and 5 was almost undetectable in some patients whereas a significant band was observed in others. In two of six patients, surrounding cirrhotic tissue expressed SSTR4 and 5 mRNA more clearly than the tumour of these patients. When examining expression of mRNA of the five SSTRs in different cell lines, we were able to demonstrate the presence of all receptors in HSCs, HepG2, and HuH7 cells (fig 5). In agreement with the results of liver tissue, a single band was demonstrated for SSTR subtypes 1, 2, 3, and 5 whereas multiple bands were found for SSTR4. Again, the middle band (635 nucleotides) was shown to correspond to SSTR4. Proliferation of hepatoma cells and HSCs was stimulated with 2% fetal calf serum or PDGF-BB, respectively. As expected, PDGF-BB induced a marked increase in the proliferation of HSCs. 21 Proliferation of hepatocellular carcinoma cell lines HepG2 and HuH7, and HSCs did not change in the presence of different concentrations of various SSTR agonists compared with cells treated with PDGF-BB alone (data not shown). In preliminary experiments, no apoptotic effects of somatostatin agonists were observed. As somatostatin induced apoptosis has been attributed to activation of SSTR2 and 3, definitive experiments were performed with these agonists. No effects were observed in any of the three cell lines after 24 hours. Even when cells were treated for 72 hours with L-779,976 or L-796,778, no difference in apoptosis between treated and untreated cells was detected (fig 6). In the presence of chemotactic stimuli PDGF-BB or HGF in the lower part of the Boyden migration chamber, all three cell types migrated through the pores to the lower surface of the membrane (fig 7A). Among the five SSTR agonists tested, only addition of L-797,591 (SSTR1 agonist) significantly reduced migration (fig 7B). Compared with untreated control cells, migration of HepG2, HuH7, and HSCs significantly decreased to 88 (7)% (p , 0.05), 83 (11)% (p , 0.05), and 67 (13)% (p , 0.01), respectively. In this study, we have described expression of SSTR subtypes in normal human liver, in cirrhosis, and in HCC. Furthermore, we showed that migration of hepatoma cells and HSCs was reduced via activation of SSTR subtype 1. However, the subtype specific somatostatin agonists used in this study had no effect on proliferation or apoptosis. Expression of SSTRs in human liver has not been ...
Context 2
... Meylan, France) at a density of 15 6 10 3 , 15 6 10 3 , and 6 6 10 3 cells per well, respectively. All cells were suspended in DMEM supplemented with 2% fetal calf serum. After 24 hours, media were renewed and cells exposed for another 24 hours to different concentrations of SSTR agonists (10 2 12 to 10 2 8 mol/l) in the presence of 10 m mol BrdU. Platelet derived growth factor (PDGF-BB 10 ng/ml), a strong mitogen for HSCs, 21 was added to the medium of HSCs (R & D Systems Europe, Abingdon, UK). In each experiment, all conditions were tested 10 times, and every experiment was performed in triplicate. To allow comparison of proliferation between different groups, results were expressed as percentages and compared with controls normalised to 100%. Mean (SD) values were calculated for all groups. Somatostatin induced apoptosis was studied by the TUNEL reaction according to the manufacturer’s instructions (In Situ Cell Death Detection Kit; Roche diagnostics, Belgium). After trypsinisation, HepG2, HuH7, and HSCs were plated in 96 well dishes at a density of 2.5 6 10 3 , 2.5 6 10 3 , and 1 6 10 3 cells per well, respectively. All cells were suspended in DMEM supplemented with 2% fetal calf serum. As apoptosis has been attributed to stimulation of SSTR2 and 3, different concentrations (10 2 12 to 10 2 6 mol/l) of L-779,976 and L-796,778, agonists of SSTR 2 and 3, respectively, were studied. After 24 and 72 hours of treatment, apoptotic cells were counted and expressed as a percentage of total cells. Experiments were performed in triplicate. The influence of the five SSTR agonists on migration of the hepatoma cell lines and HSCs was investigated, as described previously, 22 using Boyden invasion chambers with 8 m m pore size filters coated with Matrigel basement membrane matrix (Falcon, Becton Dickinson Labware, Bedford, Massachusetts, USA). PDGF-BB is a very strong chemoattractant for human HSCs 21 whereas hepatocyte growth factor (HGF) increases the invasiveness of HepG2 and HuH7 tumour cells. 22 To investigate the possibility that somatostatin inhibits PDGF- BB induced migration of HSCs or HGF induced migration of HepG2 and HuH7, cells were seeded in the upper well of Boyden chambers in the presence of PDGF-BB or HGF in the lower chamber. In these experiments, 2.5 6 10 4 hepatoma cells or HSCs were seeded onto the filter. HGF 20 ng/ml (R & D Systems Europe, Abingdon, UK) was added to the lower chamber when hepatoma cells were seeded in the upper chamber, or PDGF-BB 10 ng/l was added when HSCs were present in the upper chamber. Subsequently, 10 2 9 mol/l of SSTR agonists were added to the cells in the upper chamber. After 12 and 48 hours for experiments with HSCs and hepatoma cells, respectively, cells in the upper chamber were wiped with a cotton swab, and the filters were fixed in methanol and stained with haematoxylin. Cells invading the lower surface of the filter were counted in 10 high power fields using a Zeiss Axioplan microscope. Results were expressed as a percentage, relative to controls normalised to 100%. Experiments were performed in triplicate, except for controls and experiments with L-797,591 (agonist of SSTR1) which were performed six times. One way analysis of variance (ANOVA) with Bonferroni’s correction for multiple comparisons and the Mann-Whitney test were used when appropriate. Results are presented as means (SD). A p value of , 0.05 was considered statistically significant. Analysis was performed using Instat software (GraphPad software inc., San Diego, California, USA). Using well validated specific antibodies against the five recognised SSTRs, we were able to localise the different SSTRs in sections of normal liver (fig 1), cirrhosis (fig 2), and HCC (fig 3). Expression of the various receptors varied between different specimens and cells (table 3). In fig 1, normal liver is presented. Hepatocytes and sinusoidal cells did not stain for any of the five receptors. Cholangiocytes, on the other hand, consistently expressed SSTR subtypes 1, 3, and 4. Immunoreaction with SSTR subtypes 1 and 4 was more intense than with SSTR subtypes 2, 3, and 5 which were detected in, respectively, two, three, and one of the three analysed specimen. Furthermore, a weak immunoreaction of SSTR1 with endothelial cells of a small number of small arteries was found (not shown). In cirrhotic liver, expression of SSTRs significantly changed compared with normal liver. Hepatocytes within cirrhotic nodules expressed SSTR1 (fig 2B, C), and in 4/6 of the examined specimens, specimens also expressed SSTR2 (fig 2F, G), SSTR3 (fig 2J, K), SSTR4 (fig 2N, O), and SSTR5 (fig 2R, S). In most of the studied cases, we also found cells which double stained for all five SSTRs and a -SMA. These cells, clearly activated HSCs, were located at the interface of septa and cirrhotic nodules (fig 2C, G, K, O). In some specimens, we observed HSCs within hepatic nodules that stained for PDGFR- b and SSTR1 (fig 2D) or SSTR3 (fig 2L). In fig 3, immunohistochemistry for HCC is presented. Most of the tumours stained with SSTR subtypes 1 and 2 whereas 50% reacted with SSTR subtypes 3, 4, and 5 (table 3). In some tumours, non-tumoral cells double stained for a -SMA and SSTR subtypes 1, 2, and 3 (fig 3B, table 3). To verify the results of double staining immunohistochemistry, isolated cell lines were exposed to the different SSTR antibodies. Human HSCs were positive for all five SSTRs although immunoreaction with SSTR3 was less intense. Similarly, HepG2 expressed the five SSTR subtypes, again less clearly SSTR3. Finally, HuH7 reacted with all SSTRs, except for SSTR3 (data not shown). Messenger RNA of the five SSTRs was detected both in normal and pathological liver tissue (fig 4). We identified a single band for all receptors, except SSTR4 for which three bands were found in some patients. Sequencing demonstrated that the middle band (635 nucleotides), which was most intense, corresponded to SSTR4. This was in agreement with the findings of Talme et al who used the same primers for SSTR4. 23 All investigated samples were definitely positive for SSTR subtypes 1 and 2. For these receptors, no important variations between different patients or between cirrhosis and tumour samples from the same patient were observed. Expression of mRNA of SSTR subtypes 3, 4, and 5 was almost undetectable in some patients whereas a significant band was observed in others. In two of six patients, surrounding cirrhotic tissue expressed SSTR4 and 5 mRNA more clearly than the tumour of these patients. When examining expression of mRNA of the five SSTRs in different cell lines, we were able to demonstrate the presence of all receptors in HSCs, HepG2, and HuH7 cells (fig 5). In agreement with the results of liver tissue, a single band was demonstrated for SSTR subtypes 1, 2, 3, and 5 whereas multiple bands were found for SSTR4. Again, the middle band (635 nucleotides) was shown to correspond to SSTR4. Proliferation of hepatoma cells and HSCs was stimulated with 2% fetal calf serum or PDGF-BB, respectively. As expected, PDGF-BB induced a marked increase in the proliferation of HSCs. 21 Proliferation of hepatocellular carcinoma cell lines HepG2 and HuH7, and HSCs did not change in the presence of different concentrations of various SSTR agonists compared with cells treated with PDGF-BB alone (data not shown). In preliminary experiments, no apoptotic effects of somatostatin agonists were observed. As somatostatin induced apoptosis has been attributed to activation of SSTR2 and 3, definitive experiments were performed with these agonists. No effects were observed in any of the three cell lines after 24 hours. Even when cells were treated for 72 hours with L-779,976 or L-796,778, no difference in apoptosis between treated and untreated cells was detected (fig 6). In the presence of chemotactic stimuli PDGF-BB or HGF in the lower part of the Boyden migration chamber, all three cell types migrated through the pores to the lower surface of the membrane (fig 7A). Among the five SSTR agonists tested, only addition of L-797,591 (SSTR1 agonist) significantly reduced migration (fig 7B). Compared with untreated control cells, migration of HepG2, HuH7, and HSCs significantly decreased to 88 (7)% (p , 0.05), 83 (11)% (p , 0.05), and 67 (13)% (p , 0.01), respectively. In this study, we have described expression of SSTR subtypes in normal human liver, in cirrhosis, and in HCC. Furthermore, we showed that migration of hepatoma cells and HSCs was reduced via activation of SSTR subtype 1. However, the subtype specific somatostatin agonists used in this study had no effect on proliferation or apoptosis. Expression of SSTRs in human liver has not been studied in detail. In one study, SSTR2 was present in low amounts 24 although in another study using northern blot analysis, no relevant amounts of mRNA for any of the five SSTRs could be detected. 11 With the very sensitive RT-PCR technique, we were able to demonstrate the presence of all five SSTRs in normal human liver. By means of immunohistochemistry, we demonstrated that bile ducts, but not hepatocytes or sinusoids, immunoreacted with specific SSTR antibodies. Furthermore, SSTR1 was present in the endothelial cells of some small blood vessels, which is in agreement with findings in normal extrahepatic human blood vessels. 25 In a somatostatin 14/28 receptor binding study, levels of SSTRs were either absent, low, or very high in hepatitis and cirrhosis. 5 We also found considerable variation in SSTR expression in cirrhosis. Receptor subtypes 1 and 2 were prominent in most of the studied samples but SSTR 3, 4, and 5 were clearly present in some and absent in other specimens. As we did not use a quantitative PCR method, we cannot draw firm conclusions on the variation in mRNA expression. However, care was taken to start with equal amounts of mRNA, as proven by amplification of b -actin mRNA. Additionally, immunohistochemistry results ...

Citations

... In rats, SSTR2 is mainly expressed in the brain, pituitary, pancreas, and adrenal glands [40][41][42][43][44]. In humans, SSTR2 is expressed in the brain, kidney, stomach, duodenum, ileum and liver, and has also been detected in adipose tissue [45][46][47][48]. To our knowledge, this is the first time that SSTR2 has been found to be highly expressed in chicken adipose tissue. ...
Article
Full-text available
Somatostatin shows an anti-lipolytic effect in both chickens and ducks. However, its molecular mediator remains to be identified. Here, we report that somatostatin type 2 receptor (SSTR2) is expressed at a high level in chicken adipose tissue. In cultured chicken adipose tissue, the inhibition of glucagon-stimulated lipolysis by somatostatin was blocked by an SSTR2 antagonist (CYN-154086), supporting an SSTR2-mediated anti-lipolytic effect. Furthermore, a significant pro-proliferative effect was detected in SST28-treated immortalized chicken preadipocytes (ICP-1), and this cell proliferative effect may be mediated through the MAPK/ERK signaling pathway activated by SSTR2. In summary, our results demonstrate that SSTR2 may regulate adipose tissue development by affecting the number and volume of adipocytes in chickens.
... Эти рецепторы показали высокое сродство как к природному соматостатину, так и к октреотиду [1]. В цирротической печени и ГЦК экспрессируются все пять рецепторов соматостатина (SSTR) как на уровне белка, так и на уровне мРНК, но в нормальной печени все они (SSTR) отрицательны [2]. Более того, похоже, что все случаи ГЦК не демонстрируют сходные паттерны экспрессии SSTR. ...
Article
The process of tumor differentiation is a central aspect of the histopathological classification of solid malignancies and is closely related to the biological behavior of the tumor (poorly differentiated tumors/dedifferentiated tumors are known to be more aggressive than more differentiated tumors). The mechanisms by which tumor cell differentiation is disrupted are poorly understood, but pathologists and molecular tumor biologists have introduced the concept of dedifferentiation to explain the phenotypic changes that occur in solid tumors. In this review, we discuss a case of detection of dedifferentiated cancer, where even with the help of molecular testing of a tumor sample, we were not completely able to unambiguously determine the original source of the process, which entailed difficulties in choosing treatment tactics. And the main question that we asked ourselves during the treatment process: should treatment be based on molecular markers identified in the tumor, or should treatment be carried out according to the recommendations for the treatment of tumors of an undetected primary location with empirically selected chemotherapy?
... It has been suggested that this decrease may be attributed to distinct internalization patterns of SSTRs in normal tissues and tumour cells, resulting in downregulation of SSTRs in the normal liver tissue [6,7]. On the other hand, a study by Reynaert et al. did not identify any SSTR expression in normal hepatocytes and hepatic stellate cells [8]. ...
... [5][6][7] But in our study, obtained median SUV max and SUV mean values for 68 Ga-DOTATATE were significantly higher as compared with the results of Kunikowska et al. 5 In vitro studies have demonstrated that hepatocytes and hepatic stellate cells do not express any SSTR subtypes. 10 Boy et al. 11 reported that the main SSTR subtype in liver is SSTR1 (94.2%), followed by SSTR2 (5.7%) and SSTR5 (0.1%), while negative for SSTR3 and SSTR4. To an accepted consensus, the uptake of 68 Ga-DOTATATE and 68 Ga-DOTANOC in the liver is related to the metabolism and elimination of peptides. ...
Article
Full-text available
Objective: In this study, we aimed to investigate the physiological biodistribution pattern of 68Ga- DOTATATE and DOTANOC using standardized uptake value (SUV) parameters (maximum and mean) in nontumorous tissues on positron emission tomography/computed tomography (PET/CT) images. Methods: Images of 40 patients (female, n=23 and male, n=17; mean age, 52.50+-15.82 years) who had undergone 68Ga-DOTA labeled (TATE or NOC) PET/CT imaging with the diagnosis of GEPneuroendocrine tumor (NET), non-GEP-NET and thyroid cancer were retrospectively evaluated. SUVmax and SUVmean measurements were performed from areas of physiologic uptake including pituitary gland, parotid and submandibular gland, palatine tonsils, thyroid, lungs, blood pool, thymus, lymph nodes, liver, pancreas (from head, body and tail parts), spleen, stomach, both adrenal gland, kidney, small bowel and colon, bone marrow, prostate, glandular breast tissue and muscle tissue. Results: The highest uptake for 68Ga-DOTATATE and 68Ga-DOTATANOC was noted in the spleen, adrenal, kidney, liver, pituitary gland and head of the pancreas, respectively. Lung, muscle, blood pool, bone marrow, lymph node and breast showed low uptake (SUVmax-mean <2), while moderate uptake was observed in the remaining organs. Conclusion: We think that determination of uptake patterns and range of SUVmax-mean values for both agents in many organs will aid in discriminating between physiologic and pathologic uptake during the interpretation of images.
... Besides, 68 Ga-DOTATATE is excreted in the renal tract, and, therefore, renal parenchymal uptake might be overestimated due to intense excretory tracer activity in the medullary pyramids and possible signal crossover from the renal pelvis. Although in vitro studies have shown that hepatocytes and hepatic stellate cells of the normal liver parenchyma do not express any of the SSTR subtypes [25], high 68 Ga-DOTATATE uptake in the liver was observed in this study. However, as an organ related to metabolism and elimination of radiotracer, nonsomatostatin receptor-mediated uptake in liver [26] may have contributed to this phenomenon. ...
Article
Full-text available
Objective: To investigate the biodistribution and kinetic constants of 68Ga-DOTATATE in normal organs through dynamic total-body positron emission tomography/computed tomography (PET/CT). Methods: Seven patients who experienced endoscopic resection of gastric neuroendocrine tumor were enrolled. Dynamic total-body PET/CT scans over 60 min were performed. Time-activity curves were obtained by drawing regions of interest in normal organs. Rate constants, including K 1, k 2, k 3, and vB, were computed using a two-tissue compartment model. Factor analysis was used to compare the rate constants among subjects and regions. Hierarchical cluster analysis was performed to identify organs with similar kinetic characteristics. Results: The highest uptake of 68Ga-DOTATATE was observed in the spleen followed by kidneys, adrenals, liver, pituitary gland, pancreas head, prostate, pancreas body, and thyroid, parotid, and submandibular glands. Low background level of 68Ga-DOTATATE uptake was observed in the nasal mucosa, bone, blood pool, and cerebrum. In addition, the uptake in the pancreas head was noted to be higher than the pancreas body (P < 0.001) on the basis of each time point of dynamic PET. There were differences of rate constants among different organs. The mean K 1 ranged from 0.0507 min-1 in the left nasal mucosa to 1.21 min-1 in the left kidney, and mean k 2 ranged from 0.0174 min-1 in the spleen to 4.4487 min-1 in the left cerebrum. The mean k 3 ranged from 0.0563 min-1 in the right cerebrum to 4.6309 min-1 in the left adrenal, and mean vB ranged from 0.0001 in the left cerebrum to 0.2489 in the right adrenal. However, none of the rate constants was significantly different among subjects or among different sites within a single organ. Three groups of organs with similar kinetic characteristics were identified: (1) cerebrum; (2) pituitary gland, liver, adrenal, and prostate; and (3) nasal mucosa, parotid and submandibular glands, thyroid, spleen, pancreas, kidney, and bone. Conclusion: Uptake and clearance of 68Ga-DOTATATE, in terms of kinetic constants, were different in different organs. The kinetic parameters of 68Ga-DOTATATE in different organs provide a reference for future dynamic PET imaging.
... The expression of SSTR has been reported in various liver diseases such as acute hepatitis, chronic hepatitis, cirrhosis, and HCC (Kouroumalis et al., 1998). SSTR was found to express in 41% of HCC patients (24/59), whereas expression of SSTR was absent in normal liver cells such as hepatocytes and hepatic stellate cells, as evidenced from the study (Reynaert et al., 2004). The expression levels of different subtypes such as SSTR1, SSTR2, SSTR3, SSTR4, and SSTR5 of SSTR receptor in HCC were found to be 46, 41, 64, 0, and 75%, respectively (Reynaert et al., 2004). ...
... SSTR was found to express in 41% of HCC patients (24/59), whereas expression of SSTR was absent in normal liver cells such as hepatocytes and hepatic stellate cells, as evidenced from the study (Reynaert et al., 2004). The expression levels of different subtypes such as SSTR1, SSTR2, SSTR3, SSTR4, and SSTR5 of SSTR receptor in HCC were found to be 46, 41, 64, 0, and 75%, respectively (Reynaert et al., 2004). These findings were further supported by several other studies (Notas 2004;Blaker 2004). ...
Chapter
Full-text available
Hepatocellular carcinoma (HCC), the most common liver malignancy, has been a significant cause of cancer-related deaths worldwide. Cirrhosis, hepatic viral infections, fatty liver, and alcohol consumption are notable risk factors associated with HCC. Furthermore, a crucial challenge in the therapeutic management of HCC patients is the late-stage diagnosis, primarily due to the asymptomatic early stage. Despite the availability of various preventive techniques, diagnoses, and several treatment options, the mortality rate persists. Ongoing investigation on exploring molecular pathogenesis of HCC and identifying different prognostic and diagnostic markers may intervene in the conventional mode of treatment option for better therapeutic management of the disease. Subsequently, tumor site and its size, extrahepatic spread, and liver function are the underlying fundamental factors in treating treatment modality. The development in both surgical and non-surgical methods has resulted in admirable benefits in the survival rates. Understanding the mechanism(s) of tumor progression and the ability of the tumor cells to develop resistance against drugs is extremely important for designing future therapy concerning HCC. This chapter has accumulated the current literature and provided a vivid description of HCC based on its classification, risk factors, stage-based diagnosis systems, molecular pathogenesis, prognostic/diagnostic markers, and the existing conventional treatment approaches.
... Some data point to a potential anti-fibrotic action of cortistatin in liver. Thus, cortistatin receptors (somatostatin-receptors SST 1 -SST 5 and the ghrelin receptor) are expressed in fibroblasts and HSCs, and various SST receptor/ghrelin receptor-agonists exert anti-fibrotic responses in liver (Borie et al., 2008;Fort et al., 1998;Moreno et al., 2010;Reynaert et al., 2001Reynaert et al., , 2004Sikiric et al., 1993;Tracy et al., 1993). Moreover, in several non-fibroblastic cells, cortistatin inhibited signalling pathways that are involved in fibrogenic responses (Duran-Prado et al., 2013;Liu et al., 2018;Morell et al., 2014). ...
... Moreover, in several non-fibroblastic cells, cortistatin inhibited signalling pathways that are involved in fibrogenic responses (Duran-Prado et al., 2013;Liu et al., 2018;Morell et al., 2014). Finally, evidence indicates that cortistatin is expressed by various types of liver cells, including Kupffer cells (KCs) and hepatocytes (Reynaert et al., 2004;Xidakis et al., 2007), and the Human Protein Atlas (www. ...
... Whereas fibrosis in hepatotoxic liver injury is attributed to activated HSCs, activated PFs are implicated in liver fibrosis caused by cholestatic liver injury. Our and other studies showed that cortistatin and its receptors are expressed in HSCs and PFs, and therefore, it could act in both fibrogenic cell types in an autocrine/paracrine manner (Borie et al., 2008;Moreno et al., 2010;Reynaert et al., 2004). In fact, treatment with cortistatin reversed the activated myofibroblastic phenotype observed in cortistatin-deficient hepatic cells and impaired the activation of human and mouse HSCs, pointing to these cells as major targets for the anti-fibrotic effect of cortistatin. ...
Article
Full-text available
Background and Purpose Liver fibrosis induced by chronic hepatic injury remains a major cause of morbidity and mortality worldwide. Identification of susceptibility/prognosis factors and new therapeutic tools for treating hepatic fibrotic disorders are urgent medical needs. Cortistatin is a neuropeptide with potent anti‐inflammatory and anti‐fibrotic activities in lung that binds to receptors that are expressed in liver fibroblasts and hepatic stellate cells. We evaluated the capacity of cortistatin to regulate liver fibrosis. Experimental Approach We experimentally induced liver fibrosis in mice by chronic CCl4 exposure and bile duct ligation and evaluated the histopathological signs and fibrotic markers. Key Results Hepatic expression of cortistatin inversely correlated with liver fibrosis grade in mice and humans with hepatic disorders. Cortistatin‐deficient mice showed exacerbated signs of liver damage and fibrosis and increased mortality rates when challenged by hepatotoxic and cholestatic injury. Compared with wild‐type mice, non‐parenchymal liver cells isolated from cortistatin‐deficient mice showed increased presence of cells with activated myofibroblast phenotypes and a differential genetic signature that is indicative of activated hepatic stellate cells and periportal fibroblasts and of myofibroblasts with active contractile apparatus. Cortistatin treatment reversed in vivo and in vitro these exaggerated fibrogenic phenotypes and protected from progression to severe liver fibrosis in response to hepatic injury. Conclusion and Implications We identify cortistatin as an endogenous molecular brake on liver fibrosis and its deficiency as a potential poor‐prognosis marker for chronic hepatic disorders that link with fibrosis. Cortistatin‐based therapies emerge as attractive strategies for ameliorating severe hepatic fibrosis of various aetiologies.
... Somatostatin exerts its effects by binding to a family of five Gicoupled somatostatin receptors (SSTR1 to SSTR5) (55). HSCs of cirrhotic livers and culture-activated HSCs express all five SSTRs, whereas SSTRs are not detected in HSCs of the normal liver (55,56). ...
... Somatostatin exerts its effects by binding to a family of five Gicoupled somatostatin receptors (SSTR1 to SSTR5) (55). HSCs of cirrhotic livers and culture-activated HSCs express all five SSTRs, whereas SSTRs are not detected in HSCs of the normal liver (55,56). Interestingly, an SSTR1 agonist, L-797,591, decreases the migration of HSCs but does not affect HSC proliferation or apoptosis (55). ...
... HSCs of cirrhotic livers and culture-activated HSCs express all five SSTRs, whereas SSTRs are not detected in HSCs of the normal liver (55,56). Interestingly, an SSTR1 agonist, L-797,591, decreases the migration of HSCs but does not affect HSC proliferation or apoptosis (55). Another study suggests that the effect of octreotide, a SSTR2/5 agonist, on liver fibrosis depends on the cytokine microenvironment of HSCs (57). ...
Article
Full-text available
The prevalence of non-alcoholic fatty liver disease (NAFLD) is globally increasing. Gaining control over disease-related events in non-alcoholic steatohepatitis (NASH), an advanced form of NAFLD, is currently an unmet medical need. Hepatic fibrosis is a critical prognostic factor in NAFLD/NASH. Therefore, a better understanding of the pathophysiology of hepatic fibrosis and the development of related therapies are of great importance. G protein-coupled receptors (GPCRs) are cell surface receptors that mediate the function of a great variety of extracellular ligands. GPCRs represent major drug targets, as indicated by the fact that about 40% of all drugs currently used in clinical practice mediate their therapeutic effects by acting on GPCRs. Like many other organs, various GPCRs play a role in regulating liver function. It is predicted that more than 50 GPCRs are expressed in the liver. However, our knowledge of how GPCRs regulate liver metabolism and fibrosis in the different cell types of the liver is very limited. In particular, a better understanding of the role of GPCRs in hepatic stellate cells (HSCs), the primary cells that regulate liver fibrosis, may lead to the development of drugs that can improve hepatic fibrosis in NAFLD/NASH. In this review, we describe the functions of multiple GPCRs expressed in HSCs, their roles in liver fibrogenesis, and finally speculate on the development of novel treatments for NAFLD/NASH.
... Somatostatin also reduces the levels of proinflammatory cytokines in rat liver stellate cells [51], and this antiinflammatory action may be beneficial in HCC. Other effects on stellate cells have also been reported, but this remains subject to further research [52,53]. Furthermore, somatostatin may reduce the activity of MMPs which are associated with Kupffer cells [43]. ...
... Whereas octreotide and lanreotide have a great affinity for SST 2 and SST 5 and to a lesser extent SST 3 [70], pasireotide has a high affinity for receptors SST 1 , SST 3 , and SST 5 [71]. Since different types of SSTs have been identified in different types of HCC, it may be of paramount importance to identify the type of SST or SSTs which are more prevalent and adjust the SSA treatment accordingly [52,[72][73][74][75]. ...
... The numerous experiments on the action of somatostatin and its analogs (e.g., [44,52,[76][77][78][79][80][81][82][83][84][85]) did not always exhibit a unified corpus of conclusions. While many researchers replicated the originally determined antiproliferative and/or apoptotic effects, others did not manage that, and some even had conflicting results, depending on the dose of somatostatin or SSA employed. ...
Article
Full-text available
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and affects about 8% of cirrhotic patients, with a recurrence rate of over 50%. There are numerous therapies available for the treatment of HCC, depending on cancer staging and condition of the patient. The complexity of the treatment is also justified by the unique pathogenesis of HCC that involves intricate processes such as chronic inflammation, fibrosis, and multiple molecular carcinogenesis events. During the last three decades, multiple in vivo and in vitro experiments have used somatostatin and its analogs (SSAs) to reduce the proliferative and metastatic potential of hepatoma cells by inducing their apoptosis and reducing angiogenesis and the inflammatory component of HCC. Most experiments have proven successful, revealing several different pathways and mechanisms corresponding to the aforementioned functions. Moreover, a correlation between specific effects and expression of somatostatin receptors (SSTRs) was observed in the studied cells. Clinical trials have tested either somatostatin or an analog, alone or in combination with other drugs, to explore the potential effects on HCC patients, in various stages of the disease. While the majority of these clinical trials exhibited minor to moderate success, some other studies were inconclusive or even reported negative outcomes. A complete evaluation of the efficacy of somatostatin and SSAs is still the matter of intense debate, and, if deemed useful, these substances may play a beneficial role in the management of HCC patients.
... Some data point-out to a potential anti-fibrotic action of cortistatin in liver. Thus, cortistatin-receptors (somatostatin-receptors sstr1-5 and ghrelin-receptor GHSR) are expressed in fibroblasts and HSCs, and various sstr/GHSR-agonists were described that exert antifibrotic responses in liver (13)(14)(15)(16)(17)(18)(19)(20). Moreover, in several non-fibroblastic cells, cortistatin inhibited signaling pathways that are involved in fibrogenic responses (20)(21)(22)(23). ...
... Whereas fibrosis in hepatotoxic liver injury is attributed to activated HSCs, activated PFs are implicated in liver fibrosis caused by cholestatic liver injury (4)(5)(6)49). Our and other studies showed that cortistatin and its receptors are expressed in HSCs and PFs, and therefore, it could act in both fibrogenic cells in an autocrine/paracrine manner (14,18,20). In fact, treatment with cortistatin reversed the activated myofibroblastic phenotype observed in cortistatin-deficient hepatic cells, and impaired the activation of human cell line of HSCs, pointing to these cells as major targets for the anti-fibrotic effect of cortistatin. ...
... Noteworthy is that cortistatin-treatment has a favorable safety profile in humans and demonstrated clinical efficacy in Cushing's disease (52), and that the interest of pharmaceutical industry in developing cortistatin-based analogues with improved half-life in serum and clinical efficiency has increased lately (53). Previous studies described the therapeutic effect on fibrogenic responses by various agonists that signal through receptors that are recognized by cortistatin, including sstr and GHSR (13)(14)(15)(16)(17)(18)(19)(20), suggesting that binding of cortistatin to both receptor-classes could allow a kind of synergic anti-fibrotic effect in liver, a hypothesis that is confirmed here in activated LX2 cells. ...
Preprint
Full-text available
Liver fibrosis induced by chronic hepatic injury remains as a major cause of morbidity and mortality worldwide. Identification of susceptibility/prognosis factors and new therapeutic tools for treating hepatic fibrotic disorders of various etiologies are urgent medical needs. Cortistatin is a neuropeptide with potent anti-inflammatory and anti-fibrotic activities in lung that binds to receptors that are expressed in liver fibroblasts and hepatic stellate cells. Here, we evaluated the capacity of cortistatin to regulate liver fibrosis. We initially found that hepatic expression of cortistatin inversely correlated with liver fibrosis grade in mice and humans with hepatic disorders. Cortistatin-deficient mice showed exacerbated signs of liver damage and fibrosis and increased mortality rates when challenged to hepatotoxic and cholestatic injury. Compared to wild-type mice, non-parenchymal liver cells isolated from cortistatin-deficient mice showed increased presence of cells with activated myofibroblast phenotypes and a differential genetic signature that is indicative of activated hepatic stellate cells and periportal fibroblasts and of myofibroblasts with active contractile apparatus. Cortistatin treatment reversed in vivo and in vitro these exaggerated fibrogenic phenotypes and protected from progression to severe liver fibrosis in response to hepatic injury. In conclusion, we identify cortistatin as an endogenous molecular break of liver fibrosis and its deficiency as a potential poor-prognosis marker for chronic hepatic disorders that course with fibrosis. Cortistatin-based therapies emerge as attractive strategies for ameliorating severe hepatic fibrosis.