Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Mol Med Rep ; 12(4): 5003-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26151624

ABSTRACT

Medullary thyroid carcinoma (MTC) originates from the C­cells of the thyroid and is not sensitive to radiation or chemotherapy. Therefore, surgical removal of the tumor tissue in its entirety is the only curative treatment for MTC. The present study aimed to examine the potential mechanisms of action of extracts of Trailliaedoxa gracilis (TG; WW Smith & Forrest), a plant from the province of Sichuan, China, and of ursolic acid (UA), a pentacyclic triterpen present in TG, on the MTC­SK MTC cell line. A total of 13 TG fractions and UA were examined in vitro for their effects on cell morphology, cell number, proliferation and rates of apoptosis. Reverse transcription­quantitative polymerase chain reaction of nuclear factor­κB essential modifier (NEMO) was performed to delineate the role of the apoptotic pathway following treatment with UA. TG and UA were examined in vivo in xenotransplanted MTC­bearing severe combined immunodeficient mice. The TG fractions exhibited antiproliferative effects, with inhibition of mitochondrial activity in the tumor cells at concentrations, which caused no impairment of the normal control cells. The apoptotic rates of the MTC­SK cells treated with the TG fractions and UA were determined, in which no marked tumor inhibition was observed in the treated MTC­mice, and no change in the expression of NEMO was detected in the treated MTC­SK cells. The observation of early­onset activation of caspase 8 suggested that the responsible factor was linked to NEMO, an anti­apoptotic protein. However, no differences in the mRNA transcription levels of NEMO were detected in MTC­SK cells treated with UA, suggesting that this protein was not associated with the signal transducer and activator of transcription 3 pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Carcinoma, Neuroendocrine/drug therapy , Rubiaceae/chemistry , Thyroid Neoplasms/drug therapy , Triterpenes/pharmacology , Animals , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Caspase 8/genetics , Caspase 8/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Child, Preschool , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Founder Effect , Gene Expression/drug effects , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Male , Mice , Mice, SCID , Middle Aged , Plant Extracts/chemistry , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Ursolic Acid
2.
PLoS One ; 8(11): e81111, 2013.
Article in English | MEDLINE | ID: mdl-24260544

ABSTRACT

INTRODUCTION: Chromogranin A is a neuroendocrine secretory product and its loss is a feature of malignant NEN de-differentiation. We hypothesized that chromogranin A fragments were differentially expressed during NEN metastasis and played a role in the regulation of NEN proliferation. METHODS: Chromogranin A mRNA (PCR) and protein (ELISA/western blot) were studied in 10 normal human mucosa, 5 enterochromaffin cell preparations, 26 small intestinal NEN primaries and 9 liver metastases. Cell viability (WST-1 assay), proliferation (bromodeoxyuridine ELISA) and expression of AKT/AKT-P (CASE ELISA/western blot) in response to chromogranin A silencing, inhibition of prohormone convertase and mTOR inhibition (RAD001/AKT antisense) as well as different chromogranin A fragments were examined in 4 SI-NEN cell lines. RESULTS: Chromogranin A mRNA and protein levels were increased (37-340 fold, p<0.0001) in small intestinal NENs compared to normal enterochromaffin cells. Western blot identified chromogranin A-associated processing bands including vasostatin in small intestinal NENs as well as up-regulated expression of prohormone convertase in metastases. Proliferation in small intestinal NEN cell lines was decreased by silencing chromogranin A as well as by inhibition of prohormone convertase (p<0.05). This inhibition also decreased secretion of chromogranin A (p<0.05) and 5-HT (p<0.05) as well as expression of vasostatin. Metastatic small intestinal NEN cell lines were stimulated (50-80%, p<0.05) and AKT phosphorylated (Ser473: p<0.05) by vasostatin I, which was completely reversed by RAD001 (p<0.01) and AKT antisense (p<0.05) while chromostatin inhibited proliferation (~50%, p<0.05). CONCLUSION: Chromogranin A was differentially regulated in primary and metastatic small intestinal NENs and cell lines. Chromogranin A fragments regulated metastatic small intestinal NEN proliferation via the AKT pathway indicating that CgA plays a far more complex role in the biology of these tumors than previously considered.


Subject(s)
Chromogranin A/genetics , Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/genetics , Liver Neoplasms/genetics , Neuroendocrine Tumors/genetics , Proto-Oncogene Proteins c-akt/genetics , Calreticulin/metabolism , Calreticulin/pharmacology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chromogranin A/antagonists & inhibitors , Chromogranin A/metabolism , Everolimus , Humans , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/secondary , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Phosphorylation , Proprotein Convertases/antagonists & inhibitors , Proprotein Convertases/genetics , Proprotein Convertases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
3.
PLoS One ; 8(4): e62607, 2013.
Article in English | MEDLINE | ID: mdl-23638125

ABSTRACT

OBJECTIVE: We recently demonstrated that hypoxia, a key feature of IBD, increases enterochromaffin (EC) cell 5-HT secretion, which is also physiologically regulated by the ADORA2B mechanoreceptor. Since hypoxia is associated with increased extracellular adenosine, we wanted to examine whether this nucleotide amplifies HIF-1α-mediated 5-HT secretion. DESIGN: The effects of hypoxia were studied on IBD mucosa, isolated IBD-EC cells, isolated normal EC cells and the EC cell tumor derived cell line KRJ-1. Hypoxia (0.5% O2) was compared to NECA (adenosine agonist), MRS1754 (ADORA2B receptor antagonist) and SCH442146 (ADORA2A antagonist) on HIF signaling and 5-HT secretion. Antisense approaches were used to mechanistically evaluate EC cells in vitro. PCR and western blot were used to analyze transcript and protein levels of HIF-1α signaling and neuroendocrine cell function. An animal model of colitis was evaluated to confirm hypoxia:adenosine signaling in vivo. RESULTS: HIF-1α is upregulated in IBD mucosa and IBD-EC cells, the majority (~90%) of which express an activated phenotype in situ. Hypoxia stimulated 5-HT release maximally at 30 mins, an effect amplified by NECA and selectively inhibited by MRS1754, through phosphorylation of TPH-1 and activation of VMAT-1. Transient transfection with Renilla luciferase under hypoxia transcriptional response element (HRE) control identified that ADORA2B activated HIF-1α signaling under hypoxic conditions. Additional signaling pathways associated with hypoxia:adenosine included MAP kinase and CREB. Antisense approaches mechanistically confirmed that ADORA2B signaling was linked to these pathways and 5-HT release under hypoxic conditions. Hypoxia:adenosine activation which could be reversed by 5'-ASA treatment was confirmed in a TNBS-model. CONCLUSION: Hypoxia induced 5-HT synthesis and secretion is amplified by ADORA2B signaling via MAPK/CREB and TPH-1 activation. Targeting ADORA2s may decrease EC cell 5-HT production and secretion in IBD.


Subject(s)
Enterochromaffin Cells/metabolism , Inflammatory Bowel Diseases/metabolism , Oxygen/pharmacology , Receptor, Adenosine A2B/metabolism , Serotonin/biosynthesis , Serotonin/metabolism , Adenosine/pharmacology , Adult , Aged , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Colitis/metabolism , Colitis/pathology , Disease Models, Animal , Enterochromaffin Cells/drug effects , Enterochromaffin Cells/pathology , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Response Elements/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Trinitrobenzenesulfonic Acid
4.
Cancer Sci ; 104(7): 844-55, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23578138

ABSTRACT

Platelet-derived serotonin (5-HT) is involved in liver regeneration. The liver is also the metastatic site for malignant enterochromaffin (EC) cell "carcinoid" (neuroendocrine) neoplasms, the principal cellular source of 5-HT. We hypothesized that 5-HT produced by metastatic EC cells played a role in the hepatic tumor-microenvironment principally via 5-HT7 receptor-mediated activation of hepatocyte IGF-1 synthesis and secretion. Using isolated rat hepatocytes, we evaluated 5-HT7 receptor expression (using PCR, sequencing and western blot). ELISA, cell transfection and western blots delineated 5-HT-mediated signaling pathways (pCREB, AKT and ERK). IGF-1 synthesis/secretion was evaluated using QPCR and ELISA. IGF-1 was tested on small intestinal neuroendocrine neoplasm proliferation, while IGF-1 production and 5-HT7 expression were examined in an in vivo SCID metastasis model. Our results demonstrated evidence for a functional 5-HT7 receptor. 5-HT activated cAMP/PKA activity, pCREB (130-205%, P < 0.05) and pERK/pAKT (1.2-1.75, P < 0.05). Signaling was reversed by the 5-HT7 receptor antagonist SB269970. IGF-1 significantly stimulated proliferation of two small intestinal neuroendocrine neoplasm cell lines (EC50: 7-70 pg/mL) and could be reversed by the small molecule inhibitor BMS-754807. IGF-1 and 5-HT were elevated (40-300×) in peri-tumoral hepatic tissue in nude mice, while 5-HT7 was increased fourfold compared to sham-operated animals. We conclude that hepatocytes express a cAMP-coupled 5-HT7 receptor, which, at elevated 5-HT concentrations that occur in liver metastases, signals via CREB/AKT and is linked to IGF-1 synthesis and secretion. Because IGF-1 regulates NEN proliferation, identification of a role for 5-HT7 in the hepatic metastatic tumor microenvironment suggests the potential for novel therapeutic strategies for amine-producing mid-gut tumors.


Subject(s)
Hepatocytes/metabolism , Insulin-Like Growth Factor I/metabolism , Intestinal Neoplasms/metabolism , Intestine, Small/metabolism , Neuroendocrine Tumors/metabolism , Receptors, Serotonin/metabolism , Serotonin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Hepatocytes/pathology , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MAP Kinase Signaling System , Mice , Mice, SCID , Neuroendocrine Tumors/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/physiology , Tumor Microenvironment/physiology
5.
Am J Physiol Gastrointest Liver Physiol ; 303(10): G1164-73, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22936271

ABSTRACT

The chemomechanosensory function of the gut enterochromaffin (EC) cell enables it to respond to dietary agents and mechanical stretch. We hypothesized that the EC cell, which also sensed alterations in luminal or mucosal oxygen level, was physiologically sensitive to fluctuations in O(2). Given that low oxygen levels induce 5-HT production and secretion through a hypoxia inducible factor 1α (HIF-1α)-dependent pathway, we also hypothesized that increasing O(2) would reduce 5-HT production and secretion. Isolated normal EC cells as well as the well-characterized EC cell model KRJ-I were used to examine HIF signaling (luciferase-assays), hypoxia transcriptional response element (HRE)-mediated transcription (PCR), signaling pathways (Western blot), and 5-HT release (ELISA) during exposure to different oxygen levels. Normal EC cells and KRJ-I cells express HIF-1α, and transient transfection with Renilla luciferase under HRE control identified a hypoxia-mediated pathway in these cells. PCR confirmed activation of HIF-downstream targets, GLUT1, IGF2, and VEGF under reduced O(2) levels (0.5%). Reducing O(2) also elevated 5-HT secretion (2-3.2-fold) as well as protein levels of HIF-1α (1.7-3-fold). Increasing O(2) to 100% inhibited HRE-mediated signaling, transcription, reduced 5-HT secretion, and significantly lowered HIF-1α levels (∼75% of control). NF-κB signaling was also elevated during hypoxia (1.2-1.6-fold), but no significant changes were noted in PKA/cAMP. We concluded that gut EC cells are oxygen responsive, and alterations in O(2) levels differentially activate HIF-1α and tryptophan hydroxylase 1, as well as NF-κB signaling. This results in alterations in 5-HT production and secretion and identifies that the chemomechanosensory role of EC cells extends to oxygen sensing.


Subject(s)
Enterochromaffin Cells/physiology , Oxygen/pharmacology , Serotonin/physiology , Signal Transduction/physiology , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , NF-kappa B/physiology , Oxygen/administration & dosage , Serotonin/metabolism , Tryptophan Hydroxylase/metabolism
6.
BMC Res Notes ; 5: 318, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22720672

ABSTRACT

BACKGROUND: Accurate neuroendocrine neoplasia (NEN) staging is vital for determining prognosis and therapeutic strategy. The great majority of NENs express chromogranin A (CgA) which can be detected at a protein or transcript level. The current standards for lymph node metastasis detection are histological examination after Hematoxylin and Eosin (H&E) and CgA immunohistochemical (IHC) staining. We hypothesized that detection of CgA mRNA transcripts would be a more sensitive method of detecting these metastases. FINDINGS: We compared these traditional methods with PCR for CgA mRNA extracted from formalin fixed paraffin embedded slides of lymph nodes (n = 196) from small intestinal NENs, other gastrointestinal cancers and benign gastrointestinal disease. CgA PCR detected significantly more NEN lymph nodes (75%) than H&E (53%) or CgA IHC (57%) (p = 0.02). PCR detected CgA mRNA in 50% (14 of the 28) of SI-NEN lymph nodes previously considered negative. The false positive rate for detection of CgA mRNA was 19% in non-neuroendocrine cancers, and appeared to be due to occult neuroendocrine differentiation or contamination by normal epithelium during histological processing. CONCLUSIONS: Molecular pathological analysis demonstrates the limitations of observer-dependent histopathology. CgA PCR analysis detected the presence of CgA transcripts in lymph nodes without histological evidence of tumor metastasis. Molecular node positivity (stage molN1) of SI-NEN lymph nodes could confer greater staging accuracy and facilitate early and accurate therapeutic intervention. This technique warrants investigation using clinically annotated tumor samples with follow-up data.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Neuroendocrine/diagnosis , Chromogranin A/genetics , Intestinal Neoplasms/diagnosis , Intestine, Small , Lymph Nodes/chemistry , RNA, Messenger/analysis , Real-Time Polymerase Chain Reaction , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/secondary , Chromogranin A/analysis , Feasibility Studies , Humans , Immunohistochemistry , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Lymph Nodes/pathology , Lymphatic Metastasis , Neoplasm Micrometastasis , Neoplasm Staging , Observer Variation , Paraffin Embedding , Predictive Value of Tests , Reproducibility of Results , Tissue Fixation
7.
Cancer Sci ; 103(7): 1230-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22435758

ABSTRACT

Basic cancer research is dependent on reliable in vitro and in vivo tumor models. The serotonin (5-HT) producing small intestinal neuroendocrine tumor cell line KRJ-1 has been used in in vitro proliferation and secretion studies, but its use in in vivo models has been hampered by problems related to the xeno-barrier and tumor formation. This may be overcome by the encapsulation of tumor cells into alginate microspheres, which can function as bioreactors and protect against the host immune system. We used alginate encapsulation of KRJ-1 cells to achieve long-term functionality, growth and survival. Different conditions, including capsule size, variations in M/G content, gelling ions (Ca(2+) /Ba(2+)) and microcapsule core properties, and variations in KRJ-1 cell condition (single cells/spheroids) were tested. Viability and cell growth was evaluated with MTT, and confocal laser scanner microscopy combined with LIVE/DEAD viability stains. 5-HT secretion was measured to determine functionality. Under all conditions, single cell encapsulation proved unfavorable due to gradual cell death, while encapsulation of aggregates/spheroids resulted in surviving, functional bioreactors. The most ideal spheroids for encapsulation were 200-350 µm. Long-term survival (>30 days) was seen with solid Ca(2+) /Ba(2+) microbeads and hollow microcapsules. Basal 5-HT secretion was increased (sixfold) after hollow microcapsule encapsulation, while Ca(2+) /Ba(2+) microbeads was associated with normal basal secretion and responsiveness to cAMP/PKA activation. In conclusion, encapsulation of KRJ-1 cells into hollow microcapsules produces a bioreactor with a high constitutively activate basal 5-HT secretion, while Ca(2+) /Ba(2+) microbeads provide a more stable bioreactor similar to non-encapsulated cells. Alginate microspheres technology can thus be used to tailor different functional bioreactors for both in vitro and in vivo studies.


Subject(s)
Alginates/chemistry , Intestine, Small/pathology , Neuroendocrine Tumors/pathology , Spheroids, Cellular/pathology , Adrenergic beta-Agonists/pharmacology , Alginates/pharmacology , Barium/chemistry , Barium/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Calcium/chemistry , Calcium/pharmacology , Capsules , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colforsin/pharmacology , Glucuronic Acid/chemistry , Glucuronic Acid/pharmacology , Hexuronic Acids/chemistry , Hexuronic Acids/pharmacology , Humans , Intestine, Small/metabolism , Isoproterenol/pharmacology , Microscopy, Confocal , Microspheres , Models, Biological , Neuroendocrine Tumors/metabolism , Serotonin/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Time Factors
8.
Cancer ; 118(10): 2763-75, 2012 May 15.
Article in English | MEDLINE | ID: mdl-21990041

ABSTRACT

BACKGROUND: Although gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) exhibit widely divergent behavior, limited biologic information (apart from Ki-67) is available to characterize malignancy. Therefore, the identification of alternative biomarkers is a key unmet need. Given the role of internexin alpha (INA) in neuronal development, the authors assessed its function in neuroendocrine cell systems and the clinical implications of its expression as a GEP-NEN biomarker. METHODS: Functional assays were undertaken to investigate the mechanistic role of INA in the pancreatic BON cell line. Expression levels of INA were investigated in 50 pancreatic NENs (43 primaries, 7 metastases), 43 small intestinal NENs (25 primaries, 18 metastases), normal pancreas (n = 10), small intestinal mucosa (n = 16), normal enterochromaffin (EC) cells (n = 9), mouse xenografts (n = 4) and NEN cell lines (n = 6) using quantitative polymerase chain reaction, Western blot, and immunostaining analyses. RESULTS: In BON cells, decreased levels of INA messenger RNA and protein were associated with the inhibition of both proliferation and mitogen-activated protein kinase (MAPK) signaling. INA was not expressed in normal neuroendocrine cells but was overexpressed (from 2-fold to 42-fold) in NEN cell lines and murine xenografts. In pancreatic NENs, INA was overexpressed compared with pancreatic adenocarcinomas and normal pancreas (27-fold [P = .0001], and 9-fold [P = .02], respectively). INA transcripts were correlated positively with Ki-67 (correlation coefficient [r] = 0.5; P < .0001) and chromogranin A (r = 0.59; P < .0001). INA distinguished between primary tumors and metastases (P = .02), and its expression was correlated with tumor size, infiltration, and grade (P < .05). CONCLUSIONS: INA is a novel NEN biomarker, and its expression was associated with MAPK signaling and proliferation. In clinical samples, elevated INA was correlated with Ki-67 and identified malignancy. INA may provide additional biologic information relevant to delineation of both pancreatic NEN tumor phenotypes and clinical behavior.


Subject(s)
Gastrointestinal Neoplasms/chemistry , Intermediate Filament Proteins/analysis , Neuroendocrine Tumors/chemistry , Neurofilament Proteins/analysis , Pancreatic Neoplasms/chemistry , Animals , Biomarkers, Tumor , Cell Line, Tumor , Gastrointestinal Neoplasms/pathology , Humans , Intermediate Filament Proteins/physiology , Mice , Neuroendocrine Tumors/pathology , Neurofilament Proteins/genetics , Neurofilament Proteins/physiology , Pancreas/chemistry , Pancreatic Neoplasms/pathology
9.
Bioorg Chem ; 40(1): 108-113, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22024042

ABSTRACT

In this study, a completely water soluble tri-cationic porphyrin-EDTA conjugate was synthesized. We present data demonstrating the tumoristatic effects of the novel fully water soluble cationic porphyrin TMPy(3)PhenEDTA-P-Cl(4) in the dark, in the medullary thyroid carcinoma cell lines MTC-SK and SHER-I and weaker effects in the small intestinal neuroendocrine tumor cell line KRJ-I. In addition, cytotoxic effects were also studied in normal human fibroblasts that represent normal tissue and the results are compared to the tumor cell lines.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Edetic Acid/analogs & derivatives , Edetic Acid/chemistry , Porphyrins/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Edetic Acid/pharmacology , Edetic Acid/therapeutic use , Humans , Neuroendocrine Tumors/pathology , Porphyrins/chemistry , Porphyrins/therapeutic use , Water/chemistry
10.
PLoS One ; 6(8): e22457, 2011.
Article in English | MEDLINE | ID: mdl-21853033

ABSTRACT

Small intestinal (SI) neuroendocrine tumors (NET) are increasing in incidence, however little is known about their biology. High throughput techniques such as inference of gene regulatory networks from microarray experiments can objectively define signaling machinery in this disease. Genome-wide co-expression analysis was used to infer gene relevance network in SI-NETs. The network was confirmed to be non-random, scale-free, and highly modular. Functional analysis of gene co-expression modules revealed processes including 'Nervous system development', 'Immune response', and 'Cell-cycle'. Importantly, gene network topology and differential expression analysis identified over-expression of the GPCR signaling regulators, the cAMP synthetase, ADCY2, and the protein kinase A, PRKAR1A. Seven CREB response element (CRE) transcripts associated with proliferation and secretion: BEX1, BICD1, CHGB, CPE, GABRB3, SCG2 and SCG3 as well as ADCY2 and PRKAR1A were measured in an independent SI dataset (n = 10 NETs; n = 8 normal preparations). All were up-regulated (p<0.035) with the exception of SCG3 which was not differently expressed. Forskolin (a direct cAMP activator, 10(-5) M) significantly stimulated transcription of pCREB and 3/7 CREB targets, isoproterenol (a selective ß-adrenergic receptor agonist and cAMP activator, 10(-5) M) stimulated pCREB and 4/7 targets while BIM-53061 (a dopamine D(2) and Serotonin [5-HT(2)] receptor agonist, 10(-6) M) stimulated 100% of targets as well as pCREB; CRE transcription correlated with the levels of cAMP accumulation and PKA activity; BIM-53061 stimulated the highest levels of cAMP and PKA (2.8-fold and 2.5-fold vs. 1.8-2-fold for isoproterenol and forskolin). Gene network inference and graph topology analysis in SI NETs suggests that SI NETs express neural GPCRs that activate different CRE targets associated with proliferation and secretion. In vitro studies, in a model NET cell system, confirmed that transcriptional effects are signaled through the cAMP/PKA/pCREB signaling pathway and that a SI NET cell line was most sensitive to a D(2) and 5-HT(2) receptor agonist BIM-53061.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Gene Regulatory Networks/genetics , Intestinal Neoplasms/genetics , Intestine, Small/pathology , Neuroendocrine Tumors/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/genetics , Computational Biology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Databases, Genetic , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Intestine, Small/metabolism , Models, Genetic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Response Elements/genetics , Transcription, Genetic
11.
Cancer ; 117(18): 4141-54, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21387274

ABSTRACT

BACKGROUND: Treatment of small intestinal neuroendocrine tumors (SINETs) with mammalian target of rapamycin (mTOR) inhibitors alone or with somatostatin analogs has been proposed as effective therapy, because both agents have been reported to exhibit antiproliferative activity. Because adenocarcinomas escape mTOR inhibition, we examined whether the escape phenomenon occurred in SINETs and whether usage of somatostatin analogs with mTOR inhibitors surmounted loss of inhibition. METHODS: The effects of the somatostatin analog octreotide (OCT), the mTOR inhibitor RAD001 (RAD), or the combination were evaluated in SINET cell lines (KRJ-I, H-STS) using cell viability assays, western blotting, enzyme-linked immunosorbent assay, and reverse-transcription polymerase chain reaction to assess antiproliferative signaling pathways and feedback regulation. RESULTS: RAD (10(-9) M) incompletely decreased cell viability (-40% to +15%); growth escape (P < .001) was noted at 72 hours in both cell lines. Phosphorylated (p)mTOR/mTOR and pp70S6K/p70S6K ratios were decreased but were associated with increases in phosphorylated extracellular signal-regulated kinase (pERK)/ERK and pAKT/AKT in both cell lines, whereas phosphorylated insulin-like growth factor 1 receptor (pIGF-1R)/IGF-1R levels were elevated only in H-STS cells. Increased (P < .05) transcript levels for AKT1, MAPK, mTOR, IGF-1R, IGF-1, and TGFß1 were evident. OCT (10(-6) M) itself had no significant effect on growth signaling in either cell line. An antiproliferative effect (66 ± 5%) using OCT+RAD was only noted in the KRJ-I cells (P < .05). CONCLUSIONS: SINET treatment with the mTOR inhibitor RAD had no antiproliferative effect based on activation of pAKT and pERK1/2. A combinatorial approach using OCT and RAD failed to overcome this escape phenomenon. However, differences in RAD response rates in individual NET cell lines suggested that pretreatment identification of different tumor sensitivity to mTOR inhibitors could provide the basis for individualized treatment.


Subject(s)
Intestinal Neoplasms/drug therapy , Intestine, Small , Mitogen-Activated Protein Kinase 1/metabolism , Neuroendocrine Tumors/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Everolimus , Humans , Intestinal Neoplasms/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuroendocrine Tumors/metabolism , Octreotide/pharmacology , Phosphatidylinositol 3-Kinases , Phosphorylation , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
12.
Endocrinol Metab Clin North Am ; 40(1): 1-18, vii, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21349409

ABSTRACT

In this article, updated analyses of the National Cancer Institute Surveillance, Epidemiology and End Results (SEER) registry (1973-2007) are presented and compared with epidemiologic GEP-NET data from Europe and Asia. Several studies have demonstrated a steadily increasing incidence of GEP-NETs, and this escalation is still ongoing (SEER data 2004-2007). The common primary GEP-NET sites exhibit unique epidemiologic profiles with distinct patterns of incidence, age at diagnosis, stage, and survival. Overall, GEP-NET survival has improved over the past 3 decades, although the outcome for poorly differentiated tumors remains dismal.


Subject(s)
Gastrointestinal Neoplasms/epidemiology , Neuroendocrine Tumors/epidemiology , Pancreatic Neoplasms/epidemiology , Gastrointestinal Neoplasms/mortality , Humans , Incidence , Neuroendocrine Tumors/mortality , Pancreatic Neoplasms/mortality , SEER Program , Time Factors , United States/epidemiology
13.
Endocrinol Metab Clin North Am ; 40(1): 111-34, viii, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21349414

ABSTRACT

Chromogranin A, although it exhibits limitations, is currently the most useful general tumor biomarker available for use in the diagnosis and management of gastroenteropancreatic neuroendocrine tumors (NETs). The value of the chromogranin A lies in its universal cosecretion by the majority of neuroendocrine cells that persists after malignant transformation. Clinicians aware of the physiologic role of chromogranin A and its secretion in a variety of non-NET-related pathologic conditions can use this protein as a moderately effective tumor biomarker in the management of GEP-NETs.


Subject(s)
Biomarkers, Tumor/physiology , Chromogranin A/physiology , Gastrointestinal Neoplasms/diagnosis , Neuroendocrine Tumors/diagnosis , Pancreatic Neoplasms/diagnosis , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Chromogranin A/analysis , Chromogranin A/blood , Chromogranin A/metabolism , Diagnostic Techniques, Endocrine/standards , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/metabolism , Humans , Models, Biological , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/metabolism , Predictive Value of Tests
14.
Langenbecks Arch Surg ; 396(3): 273-98, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21274559

ABSTRACT

BACKGROUND: Recent data demonstrate that the incidence of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) has increased exponentially (overall ~500%) over the last three decades, thus refuting the erroneous concept of rarity. GEP-NETs comprise 2% of all malignancies and in terms of prevalence, are the second commonest gastrointestinal malignancy after colorectal cancer. Diagnosis is usually late since there is no biochemical screening test and symptoms are protean and overlooked. As a consequence, 60-80% exhibit metastases with a consequent suboptimal outcome. DISCUSSION: The gastrointestinal tract and pancreas exhibit ~17 different neuroendocrine cell types, but neither the cell of origin nor the biological basis of GEP-NETs is understood. This review examines GEP-NETs from the cellular and molecular perspective and addresses the distinct patterns of functional tumor biology pertinent to clinicians. Although grouped as a neoplastic entity (NETs), each lesion is derived from distinct cell precursors, produces specific bioactive products, exhibits distinct chromosomal abnormalities and somatic mutation events and has uniquely dissimilar clinical presentations. GEP-NETs demonstrate very different survival rates reflecting the intrinsic differences in malignant potential and variations in proliferative regulation. Apart from the identification of the inhibitory role of the somatostatin receptors, there is limited biological knowledge of the key regulators of proliferation and hence a paucity of successful targeted therapeutic agents. IGF-I, TGFß and a variety of tyrosine kinases have been postulated as key regulatory elements; rigorous data is still required to define predictably effective and rational therapeutic strategy in an individual tumor. A critical issue in the clinical management of GEP-NETs is the need to appreciate both the neuroendocrine commonalities of the disease as well as the unique characteristics of each tumor. The further acquisition of a detailed biological and molecular appreciation of GEP-NETs is vital to the development of effective management strategy.


Subject(s)
Gastrointestinal Neoplasms/pathology , Neuroendocrine Tumors/epidemiology , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Female , Gastrointestinal Neoplasms/epidemiology , Gastrointestinal Neoplasms/therapy , Humans , Male , Neuroendocrine Tumors/therapy , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/therapy , Prevalence , Prognosis , Risk Assessment , Serotonin/metabolism , Somatostatin/metabolism , Somatostatin/therapeutic use , Survival Rate
15.
Curr Gastroenterol Rep ; 13(1): 101-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21080245

ABSTRACT

The incidence of gastric neuroendocrine tumors (NETs) has increased exponentially based on widespread use of endoscopy and a greater pathological awareness of the condition. A key concern is the potential association with hypergastrinemia induced by proton pump inhibitor administration. Previous confusion regarding diagnosis and therapy has been diminished by a series of international consensus statements defining the biology and management strategies for the disease. Overall, gastric NETs are categorized as well-differentiated or poorly differentiated neoplasms. Well-differentiated gastric NETs are enterochromaffin-like (ECL) cell tumors subclassified into three types based on their relationship to gastrin, a key regulator of ECL cell neoplastic transformation. The treatment of type 1 and type 2 tumors depends on the size and invasiveness of the tumor, whereas type 3 tumors and poorly differentiated neuroendocrine carcinomas warrant aggressive surgical resection. The disease-specific 5-year survival ranges from about 95% in type 1 gastric carcinoids to about 25% in poorly differentiated gastric NECs. Elucidation of the precise biology of a gastric NET is critical to diagnosis and delineation of a type-specific management strategy.


Subject(s)
Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Carcinoid Tumor/classification , Carcinoid Tumor/epidemiology , Carcinoid Tumor/pathology , Carcinoid Tumor/therapy , Carcinoma, Neuroendocrine/classification , Carcinoma, Neuroendocrine/epidemiology , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/therapy , Cell Transformation, Neoplastic , Enterochromaffin-like Cells/pathology , Enterochromaffin-like Cells/physiology , Gastroscopy , Humans , Incidence , Neoplasm Invasiveness , Neoplasm Staging , Neuroendocrine Tumors/classification , Neuroendocrine Tumors/epidemiology , Prognosis , Stomach Neoplasms/classification , Stomach Neoplasms/epidemiology
16.
Neuroendocrinology ; 92(3): 143-57, 2010.
Article in English | MEDLINE | ID: mdl-20733279

ABSTRACT

Neuroendocrine tumors (NETs) are a heterogeneous group of cancers of which the commonest site is the small intestine (SI). Most information available to determine tumor behavior reflects univariate assessment of factors or is anecdotal or experience based. There currently exists no objective multivariate analysis of indices that defines SI NET prognosis. A key unmet need is the lack of a rigorous mathematical-based tool - a nomogram - for the assessment of parameters that define progress, determine prognosis and can guide therapy. Since prediction of NET behavior is a critical criterion in determining clinical strategy, we constructed a NET nomogram (Modlin Score) for prognosis prediction, patient group comparisons and a guide for stratification of treatment and surveillance. We used hazard ratio (HR), Cox analysis and Kaplan-Meier analysis of published data and the current Surveillance, Epidemiology and End Results (SEER) database (approx. 20,000 patients) to develop a nomogram from 15 variables demonstrated to provide significant multivariate HRs. These included age, gender, ethnicity, symptoms, urinary 5-hydroxyindoleacetic acid, plasma chromogranin A, liver function tests, tumor size, invasion, metastasis, histology, Ki-67 index, carcinoid heart disease and therapy (surgery or long-acting somatostatin analogs). Internal validation was assessed using 33 SI NET patients. A NET nomoscore (Modlin Score) was developed by HR weighting and stratification into low (<75), medium (75-95) and high risk (>95). This identified significant differences (p <0.03, Kaplan-Meier) in survival (15.5 ± 4.3, 9.7 ± 2.5 and 6.4 ± 1.1 years, respectively). The Modlin Score was significantly elevated (p <0.01) in deceased compared to alive patients. This nomogram represents an optimized construct based upon currently analyzable data, and application will facilitate accurate stratification for comparison in clinical trials. External validation and amplification by identification of additional indices, e.g. molecular biomarkers, are necessary. The development of a mathematically validated nomogram provides a platform for objective assessment of SI NET disease, a finite basis for precise prognostication and a tool to guide management strategy.


Subject(s)
Intestinal Neoplasms/mortality , Intestine, Small/pathology , Neuroendocrine Tumors/mortality , Aged , Aged, 80 and over , Female , Humans , Intestinal Neoplasms/pathology , Kaplan-Meier Estimate , Male , Middle Aged , Neuroendocrine Tumors/pathology , Nomograms , Prognosis , Proportional Hazards Models , SEER Program
17.
Cancer ; 116(12): 2902-12, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20564397

ABSTRACT

BACKGROUND: Fibrosis is a cardinal feature of small intestinal neuroendocrine tumors (SI-NETs) both in local peritumoral tissue and systemic sites (cardiac). 5-HT, a commonly secreted NET amine, is a known inducer of fibrosis, although the mechanistic basis for it and growth factors regulating fibrosis and proliferation in the tumor microenvironment are unclear. We hypothesized that targeting 5-HT(2B) receptors on tumor cells would inhibit SI-NET 5-HT release and, thereby, fibroblast activation in the tumor microenvironment. METHODS: We studied the 5-HT(2B) receptor antagonist PRX-08066 in NET cell lines (KRJ-I, H720) and in the coculture system (KRJ-I cells: fibroblastic HEK293 cells) using real time polymerase chain reaction, ELISA, Ki67 immunostaining, and flow cytometry-based caspase 3 assays to assess antiproliferative and profibrotic signaling pathways. RESULTS: In the 5-HT(2B) expressing SI-NET cell line, KRJ-I, PRX-08066 inhibited proliferation (IC(50) 4.6 x 10(-9)M) and 5-HT secretion (6.9 x 10(-9)M) and decreased ERK1/2 phosphorylation and profibrotic growth factor synthesis and secretion (transforming growth factor beta 1 [TGFbeta1], connective tissue growth factor [CTGF] and fibroblast growth factor [FGF2]). In the KRJ-I:HEK293 coculture system, PRX-08066 significantly decreased 5-HT release (>60%), Ki67 (transcript and immunostaining: 20%-80%), TGFbeta1, CTGF, and FGF2 transcription (20%-50%) in the KRJ-I cell line. 5-HT itself stimulated HEK293 proliferation (25%) and synthesis of TGFbeta1, CTGF and FGF2. PRX-08066 inhibition of KRJ-I function reversed these effects in the coculture system. CONCLUSIONS: Targeting the 5-HT(2B) receptor may be an effective antiproliferative and antifibrotic strategy for SI-NETs because it inhibits tumor microenvironment fibroblasts as well as NET cells. Fibrosis and proliferation appear to be biologically interfaced neuroendocrine neoplasia domains.


Subject(s)
Fibroblasts/drug effects , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Receptor, Serotonin, 5-HT2C/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Fibroblasts/pathology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Ki-67 Antigen/metabolism , Serotonin/metabolism , Serotonin 5-HT2 Receptor Antagonists , Signal Transduction/drug effects
18.
Anticancer Res ; 30(1): 55-64, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20150617

ABSTRACT

BACKGROUND: Small intestinal (SI) neuroendocrine tumors (NETs) are rare neoplasms derived from neuroendocrine cells presenting distinct clinical symptoms according to the ability to secrete neuroamines. Nevertheless, many are asymptomatic and misdiagnosed. As response rates to chemotherapy are low, surgery remains the only effective treatment. Because many tumors have metastasized at the time of diagnosis, curative surgery is rarely achieved. Consequently, a substantial need for new therapeutic options has emerged. MATERIALS AND METHODS: The effects of novel plant extracts from Trailliaedoxa gracilis (W.W. Smith & Forrest) were investigated in the SI-NET cell line KRJ-I and in KRJ-I transplanted mice. Proliferation and viability were analyzed using cell counting and WST-1 cell proliferation assay. Apoptosis was determined by DAPI staining and electron microscopy, and quantified by luminescence assays for caspases 3/7, 6, 8, 9 and 2. RESULTS: Extracts of Trailliaedoxa gracilis showed a dose-dependent reduction of proliferation and induction of apoptosis in the KRJ-I cells. Normal fibroblasts were not impaired. Tumor growth inhibition was also observed in heterotransplanted SCID (severe combined immunodeficiency) mice. CONCLUSION: The in vitro and in vivo outcomes suggest a potential clinical effect of Trailliaedoxa gracilis in SI-NETs.


Subject(s)
Carcinoid Tumor/drug therapy , Intestinal Neoplasms/drug therapy , Neuroendocrine Tumors/drug therapy , Rubiaceae/chemistry , Animals , Carcinoid Tumor/pathology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Intestinal Neoplasms/pathology , Intestine, Small/pathology , Mice , Mice, SCID , Neuroendocrine Tumors/pathology , Plant Extracts/pharmacology , Xenograft Model Antitumor Assays
19.
Eur J Med Chem ; 45(2): 760-73, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19939515

ABSTRACT

In this study we demonstrate anticancer activity of novel fully water soluble cationic porphyrins. The two cationic porphyrins 5,10,15-tris(N-methylpyridinium-4-yl)-20-[1-phenyl-4-(3-N-phenylsulfonylindolyl)]-21H,23H-porphyrin chloride (TMPy(3)PhenIndolprot(1)P-Cl(3)) and 5-{5-[2-(9,9-Dimethyl)fluorenyl]-N-methylpyridinium-3-yl}-10,15,20-tris(N-methyl-pyridinium-4-yl)-21H,23H-porphyrin chloride (TMPy(3)PyFluorenyl(1)P-Cl(4)) were prepared and their antiproliferative effects were studied in two human tumor cell lines and a normal human fibroblast cell line. Effects of the novel porphyrin compounds were evaluated in the small intestinal neuroendocrine tumor cell line KRJ-I, the medullary thyroid carcinoma cell line MTC-SK and the normal human fibroblast cell line HF-SAR by cell counting, cell proliferation assays and cell cytotoxicity analyses. TMPy(3)PhenIndolprot(1)P-Cl(3) and TMPy(3)PyFluorenyl(1)P-Cl(4) showed antiproliferative effects in the tumor cell lines MTC-SK and KRJ-I; cell viability was decreased and cytotoxic effects were quantified, while no significant alterations of the human fibroblasts were noted. With the advantage of full water solubility and antiproliferative effects in tumor cell lines, the novel porphyrin compounds TMPy(3)PhenIndolprot(1)P-Cl(3) and TMPy(3)PyFluorenyl(1)P-Cl(4) could be a new therapeutic option in anticancer treatment.


Subject(s)
Fluorenes/chemistry , Indoles/chemistry , Neuroendocrine Tumors/pathology , Porphyrins/chemistry , Porphyrins/pharmacology , Thyroid Neoplasms/pathology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Base Sequence , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/genetics , DNA/metabolism , Humans , Neuroendocrine Tumors/drug therapy , Porphyrins/chemical synthesis , Porphyrins/metabolism , Solubility , Thyroid Neoplasms/drug therapy , Water/chemistry
20.
Cancer ; 115(21): 4934-45, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19634160

ABSTRACT

BACKGROUND: : Survival rates for gastrointestinal (GI) and bronchopulmonary (BP) neuroendocrine tumors (NETs) have not altered significantly (5-year survival rate: GI NETs, 64.1%; BP NETs, 87%-89%) in 30 years (from 1973 to 2004). No effective or specific antineoplastic agents are available to date, although somatostatin analogs inhibit NET 5-hydroxytryptophan (5-HT) secretion. Given the expression of 5-HT receptors on NETs, the authors hypothesized that 5-HT autoregulated NET proliferation. METHODS: : Proliferation was evaluated in 3 NET cell lines using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide uptake; in addition, real-time polymerase chain reaction analyses and enzyme-linked immunosorbent assay studies were performed to delineate 5-HT-mediated signaling pathways. To determine the receptor and role of endogenous 5-HT production, the effects of ketanserin (5-HT receptor subtypes 2A and 2C [5-HT(2A/2C)]); ondansetron (5-HT(3)); and the suicide inhibitor of the rate-limiting enzyme for 5-HT synthesis, tryptophan hydroxylase (7-HTP) were investigated. RESULTS: : Exogenously added 5-HT stimulated proliferation in atypical BP NET NCI-H720 cells (+50%; half-maximal stimulatory concentration [EC(50)] = 10 nM), in typical BP NET NCI-H727 cells (+40%; EC(50) = 0.01 nM), and in GI NET KRJ-I cells (+60%; EC(50) = 25 nM). In NCI-H720 cells, proliferation was inhibited by ketanserin (half-maximal inhibitory concentration [IC(50)] = 0.06 nM) and ondansetron (IC(50) = 0.4 nM) and also was inhibited by 7-HTP (IC(50) = 0.3 nM). In NCI-H727 cells, ketanserin and 7-HTP inhibited proliferation (IC(50) = 0.3 nM and IC(50) = 2.3 nM, respectively), whereas ondansetron had no effect. In KRJ-I cells, ketanserin (IC(50) = 0.1 nM) and 7-HTP (IC(50) = 0.6 nM), but not ondansetron, inhibited proliferation. In all cell lines, 5-HT activated proliferation through extracellular signal-regulated kinase 1 (ERK1) and ERK2 phosphorylation and c-Jun N-terminal kinase (JNK)-mediated pathways (c-JUN and Ki-67 transcription). An autoregulatory effect was indicated by the 7-HTP-mediated inhibition of extracellular 5-HT and downstream effects on NET proliferation. CONCLUSIONS: : Lung and GI NET proliferation was autoregulated by 5-HT through alterations in ERK and JNK signaling. Targeting NET cells with 5-HT(2) receptor antagonists and 7-HTP reversed proliferation. The current results indicated that 5-HT(2) receptor subtype-specific antagonists may represent a viable antiproliferative therapeutic strategy. Cancer 2009. (c) 2009 American Cancer Society.


Subject(s)
Cell Proliferation/drug effects , Intestinal Neoplasms/metabolism , Lung Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Serotonin/pharmacology , Signal Transduction , Cell Line, Tumor , Homeostasis , Humans , Intestinal Neoplasms/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Ketanserin/analogs & derivatives , Ketanserin/pharmacology , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinase 3/metabolism , Neuroendocrine Tumors/pathology , Phosphorylation , Receptors, Serotonin/metabolism , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...