Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Neuroendocrinology ; 103(3-4): 383-401, 2016.
Article in English | MEDLINE | ID: mdl-26338447

ABSTRACT

BACKGROUND/AIMS: The hepatocyte growth factor/transmembrane tyrosine kinase receptor c-Met has been defined as a potential target in antitumoral treatment of various carcinomas. We aimed to investigate the direct effect of c-Met inhibition on neuroendocrine tumor cells in vitro. METHODS: The effects of the multi-tyrosine kinase inhibitors cabozantinib and tivantinib and of the highly specific c-Met inhibitor INC280 were investigated in human pancreatic neuroendocrine BON1, bronchopulmonary NCI-H727 and midgut GOT1 cells in vitro. RESULTS: INC280, cabozantinib and tivantinib inhibited c-Met phosphorylation, respectively. However, while equimolar concentrations (10 µM) of cabozantinib and tivantinib inhibited cell viability and cell migration, INC280 had no inhibitory effect. Knockdown experiments with c-Met siRNA also did not demonstrate effects on cell viability. Cabozantinib and tivantinib caused a G2 arrest in neuroendocrine tumor cells. CONCLUSIONS: Our in vitro data suggest that c-Met inhibition alone is not sufficient to exert direct antitumoral or antimigratory effects in neuroendocrine tumor cells. The multi-tyrosine kinase inhibitors cabozantinib and tivantinib show promising antitumoral and antimigratory effects in neuroendocrine tumor cells, which are most probably 'off-target' effects, not mediated by c-Met.


Subject(s)
Anilides/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Pyridines/pharmacology , Pyrrolidinones/pharmacology , Quinolines/pharmacology , Benzamides , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , ErbB Receptors/metabolism , Flow Cytometry , Humans , Imidazoles/pharmacology , Mitogen-Activated Protein Kinase Kinases/metabolism , Neuroendocrine Tumors/pathology , Oncogene Protein v-akt/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Time Factors , Transfection , Triazines/pharmacology
2.
Nephrol Dial Transplant ; 30(8): 1272-6, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25230708

ABSTRACT

Remarkable progress has been achieved in the field of diabetes with the development of incretin analogues, dipeptidyl peptidase IV inhibitors and novel insulin analogues; nevertheless, there is an unmet need for additional therapeutic options. Individualization of HbA1c target levels is a recent progress within the field. Approximately 50% of diabetics do not reach a previously aspired treatment goal of glycosylated HbA1 levels below 7% and often face a vicious circle with accelerated weight gain. Current antidiabetic therapeutics mainly target the decline in insulin secretion and ameliorate insulin resistance. In this regard a new generation of drugs, denoted gliflozines, that specifically interfere with sodium-glucose cotransporters (SGLT)-2 and exhibit a favourable impact on glucose metabolism in patients with type 2 diabetes are emerging as hopeful avenues. The resultant negative energy balance caused by glucosuria results in long-term weight losses, significantly reduced HbA1c levels approximating 0.5-1.0% and may in addition exert beneficial effects on blood pressure, reactive oxygen products and inflammatory mediators. Recent studies indicate improvement in ß-cell glucose sensitivity and insulin sensitivity in patients treated with gliflozines, a decrease in tissue glucose disposal and interestingly an increase in endogenous glucose production. The list of side effects observed under SGLT2 inhibition includes increased rates of genitourinary infections, balanitis, vulvovaginitis, hypotensive episodes and acute deterioration of kidney function. Main questions towards the safety profile are still unanswered given that long-term clinical outcome data with SGLT2 inhibition are lacking and the cardiovascular safety profile is under scrutiny in large trials. Thus, the successful development of selective SGLT2 inhibitors for therapeutic use in diabetics has a huge potential to meet patients' needs. However, it awaits quick results from clinical trials with meaningful clinical endpoints.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/therapeutic use , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Sodium-Glucose Transporter 2 Inhibitors , Animals , Humans
3.
Hormones (Athens) ; 13(4): 498-508, 2014.
Article in English | MEDLINE | ID: mdl-25402373

ABSTRACT

Metformin is a widely used oral antidiabetic drug with good tolerability. Recent studies suggest that it also possesses adjuvant potent anticancer properties in a variety of tumors. Neuroendocrine tumors (NETs) of the gastro-entero-pancreatic system (GEP) comprise a heterogeneous group of tumors with increasing incidence and limited effective therapeutic options. Here we report the antiproliferative effects of metformin in neuroendocrine tumor cells in vitro. Treatment of human pancreatic BON1, bronchopulmonary NCI-H727, and midgut GOT1 neuroendocrine tumor cells with increasing concentrations of metformin (0.1-10 mM) dose-dependently suppressed cell viability and cell counts. Metformin induced AMPK phosphorylation in pancreatic BON1 and midgut GOT1 but suppressed AMPK activity in bronchopulmonary NCI-H727. Thus, AMPK-dependent and AMPK-independent properties may be operative in NETs of different origin. Metformin suppressed mTORC1 signaling in all three tumor cell types, evidenced by suppression of 4EBP1, pP70S6K, and S6 phosphorylation, and was associated with compensatory AKT activity. We observed induction of ERK phosphorylation in BON1 and NCI-H727 and inhibition of ERK in midgut GOT1 cells, while all three tumor cell types responded with induction of GSK3 phosphorylation. This suggests a central role for GSK3 in metformin-mediated signal transduction. Inhibition of cell proliferation by metformin was associated with apoptosis induction only in midgut GOT1, evidenced by increased subG0/1 fraction and PARP cleavage. These results suggest a potential role of metformin as a (adjuvant) therapeutic for patients with NETs.


Subject(s)
Antineoplastic Agents/pharmacology , Metformin/pharmacology , Neuroendocrine Cells/drug effects , Neuroendocrine Tumors/pathology , AMP-Activated Protein Kinases/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Neuroendocrine Cells/pathology , Neuroendocrine Cells/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
4.
World J Gastroenterol ; 20(29): 10038-49, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25110431

ABSTRACT

AIM: To investigate the effect of aspirin on neuroendocrine tumor (NET) cell growth and signaling in vitro. METHODS: Human pancreatic BON1, bronchopulmonary NCI-H727 and midgut GOT1 neuroendocrine tumor cells were treated with different concentrations of aspirin (from 0.001 to 5 mmol/L), and the resulting effects on metabolic activity/cell proliferation were measured using cell proliferation assays and SYBR-DNA-labeling after 72, 144 and 216 h of incubation. The effects of aspirin on the expression and phosphorylation of several critical proteins that are involved in the most common intracellular growth factor signaling pathways (especially Akt protein kinase B) and mammalian target of rapamycin (mTOR) were determined by Western blot analyses. Propidium iodide staining and flow cytometry were used to evaluate changes in cell cycle distribution and apoptosis. Statistical analysis was performed using a 2-tailed Student's t-test to evaluate the proliferation assays and cell cycle analyses. The results are expressed as the mean ± SD of 3 or 4 independently performed experiments. Statistical significance was set at P < 0.05. RESULTS: Treatment with aspirin suppressed the viability/proliferation of BON1, NCI-H727 and GOT1 cells in a time- and dose-dependent manner. Significant effects were observed at starting doses of 0.5-1 mmol/L and peaked at 5 mmol/L. For instance, after treatment with 1 mmol/L aspirin for 144 h, the viability of pancreatic BON1 cells decreased to 66% ± 13% (P < 0.05), the viability of bronchopulmonary NCI-H727 cells decreased to 53% ± 8% (P < 0.01) and the viability of midgut GOT1 cells decreased to 89% ± 6% (P < 0.01). These effects were associated with a decreased entry into the S phase, the induction of the cyclin-dependent kinase inhibitor p21 and reduced expression of cyclin-dependent kinase 4 and cyclin D3. Aspirin suppressed mTOR downstream signaling, evidenced by the reduced phosphorylation of the mTOR substrates 4E binding protein 1, serine/threonine kinase P70S6K and S6 ribosomal protein and inhibited glycogen synthase kinase 3 activity. We observed the (compensatory) activation of tuberous sclerosis 2, the serine/threonine specific protein kinase AKT and extracellular signal-regulated kinases. CONCLUSION: Aspirin demonstrates promising anticancer properties for NETs in vitro. Further preclinical and clinical studies are needed.


Subject(s)
Antineoplastic Agents/pharmacology , Aspirin/pharmacology , Lung Neoplasms/enzymology , Neuroendocrine Tumors/enzymology , Pancreatic Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/pathology , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Phosphorylation , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Time Factors
5.
Endocrinology ; 155(7): 2377-90, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24762141

ABSTRACT

Currently, there are no reliably effective therapeutic options for metastatic pheochromocytoma (PCC) and paraganglioma. Moreover, there are no therapies that may prevent the onset or progression of tumors in patients with succinate dehydrogenase type B mutations, which are associated with very aggressive tumors. Therefore, we tested the approved and well-tolerated drugs lovastatin and 13-cis-retinoic acid (13cRA) in vitro in an aggressive PCC mouse cell line, mouse tumor tissue-derived (MTT) cells, and in vivo in a PCC allograft nude mouse model, in therapeutically relevant doses. Treatment was started 24 hours before sc tumor cell injection and continued for 30 more days. Tumor sizes were measured from outside by caliper and sizes of viable tumor mass by bioluminescence imaging. Lovastatin showed antiproliferative effects in vitro and led to significantly smaller tumor sizes in vivo compared with vehicle treatment. 13cRA promoted tumor cell growth in vitro and led to significantly larger viable tumor mass and significantly faster increase of viable tumor mass in vivo over time compared with vehicle, lovastatin, and combination treatment. However, when combined with lovastatin, 13cRA enhanced the antiproliferative effect of lovastatin in vivo. The combination-treated mice showed slowest tumor growth of all groups with significantly slower tumor growth compared with the vehicle-treated mice and significantly smaller tumor sizes. Moreover, the combination-treated group displayed the smallest size of viable tumor mass and the slowest increase in viable tumor mass over time of all groups, with a significant difference compared with the vehicle- and 13cRA-treated group. The combination-treated tumors showed highest extent of necrosis, lowest median microvessel density and highest expression of α-smooth muscle actin. The combination of high microvessel density and low α-smooth muscle actin is a predictor of poor prognosis in other tumor entities. Therefore, this drug combination may be a well-tolerated novel therapeutic or preventive option for malignant PCC.


Subject(s)
Adrenal Gland Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Isotretinoin/pharmacology , Lovastatin/pharmacology , Pheochromocytoma/drug therapy , Actins/metabolism , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Animals , Antigens, CD34/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Chromogranin A/metabolism , Female , Immunohistochemistry , Isotretinoin/administration & dosage , Lovastatin/administration & dosage , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Nude , Muscle, Smooth/chemistry , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Time Factors , Transplantation, Homologous , Treatment Outcome , Tumor Burden/drug effects , Tyrosine 3-Monooxygenase/metabolism
6.
Int J Oncol ; 43(6): 1824-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24100469

ABSTRACT

The heat shock protein (HSP) 90 chaperone machine involved in numerous oncogenic signaling pathways is overexpressed in cancer cells and is currently being evaluated for anticancer therapy. Neuroendocrine tumors (NETs) of the gastroenteropancreatic (GEP) system comprise a heterogeneous group of tumors with increasing incidence and poor prognosis. Here, we report the antiproliferative effects of the HSP90 inhibitors AUY922 and HSP990 in neuroendocrine tumor cells. Treatment of human pancreatic BON1, bronchopulmonary NCI-H727 and midgut carcinoid GOT1 neuroendocrine tumor cells with increasing concentrations of AUY922 and HSP990 dose-dependently suppressed cell viability. Significant effects on neuroendocrine cell viability were observed with inhibitor concentrations as low as 5 nM. Inhibition of cell viability was associated with the induction of apoptosis as demonstrated by an increase in sub-G1 events and PARP cleavage. HSP90 inhibition was associated with decreased neuroendocrine ErbB and IGF-I receptor expression, decreased Erk and Akt phospho-rylation and the induction of HSP70 expression. These findings provide evidence that targeted inhibition of upregulated HSP90 activity could be useful for the treatment of aggressive neuroendocrine tumors resistant to conventional therapy.


Subject(s)
Carcinoid Tumor/drug therapy , Carcinoma, Neuroendocrine/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Pyridones/pharmacology , Pyrimidines/pharmacology , Resorcinols/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Carcinoma, Neuroendocrine/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HSP70 Heat-Shock Proteins/biosynthesis , Humans , Lung Neoplasms/drug therapy , M Phase Cell Cycle Checkpoints/drug effects , Pancreatic Neoplasms/drug therapy , Phosphoproteins/metabolism , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/biosynthesis , Receptor, IGF Type 1/biosynthesis , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects
7.
Neuroendocrinology ; 98(2): 128-36, 2013.
Article in English | MEDLINE | ID: mdl-23797089

ABSTRACT

The majority of neuroendocrine tumors (NETs) of the gastroenteropancreatic system coexpress somatostatin receptors (SSTRs) and dopamine type 2 receptors (D2R), thus providing a rationale for the use of novel SSTR2/D2R chimeric compounds in NET disease. Here we investigate the antitumor potential of the SSTR2/D2R chimeric compounds BIM-23A760 and BIM-23A758 in comparison to the selective SSTR2 agonist BIM-23023 and the selective D2R agonist BIM-53097 on human NET cell lines of heterogeneous origin. While having only minor effects on human pancreatic and bronchus carcinoid cells (BON1 and NCI-H727), BIM-23A758 induced significant antitumor effects in human midgut carcinoid cells (GOT1). These effects involved apoptosis induction as well as inhibition of mitogen-activated protein kinase and Akt signaling. Consistent with their antitumor response to BIM-23A758, GOT1 cells showed relatively high expression levels of SSTR2 and D2R mRNA. In particular, GOT1 cells highly express the short transcript variant of D2R. In contrast to BIM-23A758, the SSTR2/D2R chimeric compound BIM-23A760 as well as the individual SSTR2 and D2R agonistic compounds BIM-23023 and BIM-53097 induced no or only minor antitumor responses in the examined NET cell lines. Taken together, our findings suggest that the novel SSTR2/D2R chimeric compound BIM-23A758 might be a promising substance for the treatment of NETs highly expressing SSTR2 and D2R. In particular, a sufficient expression of the short transcript variant of DR2 might play a pivotal role for effective treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoid Tumor/pathology , Dopamine/analogs & derivatives , Intestinal Neoplasms/pathology , Recombinant Fusion Proteins/pharmacology , Somatostatin/analogs & derivatives , Carcinoid Tumor/genetics , Cell Line, Tumor , Cell Survival/drug effects , Dopamine/pharmacology , Humans , Intestinal Neoplasms/genetics , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/genetics , Receptors, Somatostatin/agonists , Receptors, Somatostatin/genetics , Somatostatin/pharmacology , Transcriptome
8.
Endocr Relat Cancer ; 19(3): 423-34, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22499437

ABSTRACT

The majority of neuroendocrine tumors (NETs) of the gastroenteropancreatic system show aberrant Akt activity. Several inhibitors of the phosphoinositide 3-kinase (PI(3)K)-Akt-mTOR signaling pathway are currently being evaluated in clinical phase II and III studies for the treatment of NETs with promising results. However, the molecular mechanisms and particularly the role of different Akt isoforms in NET signaling are not fully understood. In this study, we examine the effect of Akt inhibition on NET cells of heterogeneous origin. We show that the Akt inhibitor perifosine effectively inhibits Akt phosphorylation and cell viability in human pancreatic (BON1), bronchus (NCI-H727), and midgut (GOT1) NET cells. Perifosine treatment suppressed the phosphorylation of Akt downstream targets such as GSK3α/ß, MDM2, and p70S6K and induced apoptosis. To further investigate the role of individual Akt isoforms for NET cell function, we specifically blocked Akt1, Akt2, and Akt3 via siRNA transfection. In contrast to Akt2 knockdown, knockdown of Akt isoforms 1 and 3 decreased phosphorylation levels of GSK3α/ß, MDM2, and p70S6K and suppressed NET cell viability and colony-forming capacity. The inhibitory effect of simultaneous downregulation of Akt1 and Akt3 on tumor cell viability was significantly stronger than that caused by downregulation of all Akt isoforms, suggesting a particular role for Akt1 and Akt3 in NET signaling. Akt3 siRNA-induced apoptosis while all three isoform-specific siRNAs impaired BON1 cell invasion. Together, our data demonstrate potent antitumor effects of the pan-Akt inhibitor perifosine on NET cells in vitro and suggest that selective targeting of Akt1 and/or Akt3 might improve the therapeutic potential of Akt inhibition in NET disease.


Subject(s)
Neuroendocrine Tumors/metabolism , Phosphorylcholine/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Apoptosis/drug effects , Bronchial Neoplasms/drug therapy , Bronchial Neoplasms/metabolism , Bronchial Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA Fragmentation , Humans , Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Neoplasm Invasiveness , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylcholine/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics
9.
Endocr Relat Cancer ; 19(1): C1-5, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22009798

ABSTRACT

Angiogenic markers in pituitary adenomas remain enigmatic in terms of their function in tumorigenesis, despite being upregulated by the normal physiological trigger of hypoxia. In this issue of Endocrine-Related Cancer, Shan et al. report that the novel RWD domain containing protein, RWD-containing sumoylation enhancer, is expressed in human pituitary adenomas and plays a pivotal role in regulating the hypoxia-inducible factor 1α-vascular endothelial growth factor response to hypoxia.


Subject(s)
Adenoma/metabolism , Hypoxia/metabolism , Pituitary Neoplasms/metabolism , Adenoma/pathology , Animals , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic , Pituitary Neoplasms/pathology , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Endocr Relat Cancer ; 18(6): R197-211, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21917845

ABSTRACT

The role of ErbB family in discreet pituitary functions is reviewed. Several ErbB receptor ligands, EGF, TGFα, and heregulin are differentially expressed in normal gonadotroph and lacto-somatotroph lineages, and other elements of the anterior pituitary. ErbB receptors, i.e. EGFR and ErbB2, are also localized to the anterior pituitary with preferential EGFR lactosomatotroph expression. EGF regulates CRH and ACTH secretion and corticotroph proliferation as well as exhibiting autocrine and paracrine effects on gonadotrophs and on lactosomatotroph proliferation, gene and protein expression, and hormonal secretion. EGF and EGFR are expressed in both functioning and non-functioning pituitary adenomas, with higher expression in more aggressive tumor subtypes. ErbB2 receptor is detected in all tumor subtypes, particularly in invasive tumors. ErbB tyrosine kinase inhibitors regulate hormonal secretion, cell morphology, and proliferation in lacto-somatotroph tumors, reflecting the emerging application of targeted pituitary therapeutics.


Subject(s)
Oncogene Proteins v-erbB/biosynthesis , Pituitary Gland/metabolism , Receptor, ErbB-2/biosynthesis , Animals , Epidermal Growth Factor/metabolism , Humans , Neuregulin-1/metabolism , Oncogene Proteins v-erbB/metabolism , Pituitary Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Transforming Growth Factor alpha/metabolism
11.
Endocrinology ; 150(7): 3252-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19359387

ABSTRACT

To investigate paracrine regulation of pituitary cell growth, we tested fibroblast growth factor (FGF) regulation of TtT/GF folliculostellate (FS) cells. FGF-2, and FGF-4 markedly induced cell proliferation, evidenced by induction of pituitary tumor transforming gene-1 (Pttg1) mRNA expression and percentage of cells in S phase. Signaling for FGF-2-induced FS cell proliferation was explored by specific pharmacological inhibition. A potent inhibitory effect on FGF-2 action was observed by blocking of Src tyrosine kinase with 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d] pyrimidine (>or=0.1 microm), followed by protein kinase C (PKC) inhibition with GF109203X. Treatment with FGF-2 (30 ng/ml; 10 min) activated phosphorylation of signal transducer and activator of transcription-3, ERK, stress-activated protein kinase/c-Jun N-terminal kinase, Akt, and focal adhesion kinase. Src inhibition with 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d] pyrimidine suppressed FGF-2-induced Akt and focal adhesion kinase, indicating effects downstream of FGF-2-induced Src activation. FGF-2 also markedly induced its own mRNA expression, peaking at 2-4 h, and this effect was suppressed by Src tyrosine kinase inhibition. The PKC inhibitor GF109203X abolished FGF-2 autoinduction, indicating PKC as the primary pathway involved in FGF-2 autoregulation in these cells. In addition to pituitary FGF-2 paracrine activity on hormonally active cells, these results show an autofeedback mechanism for FGF-2 in non-hormone-secreting pituitary FS cells, inducing cell growth and its own gene expression, and mediated by Src/PKC signaling.


Subject(s)
Fibroblast Growth Factor 2/physiology , Pituitary Gland/cytology , Signal Transduction/drug effects , Animals , Cell Line , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/physiology , Feedback, Physiological , Fibroblast Growth Factor 2/genetics , Mice , Pituitary Gland/metabolism , Pyrimidines/pharmacology , RNA, Messenger/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
12.
Cancer Res ; 69(10): 4209-16, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19401448

ABSTRACT

To investigate the role of p185(her2/neu)/ErbB3 signaling in pituitary tumor function, we examined these receptors in human prolactinomas. Immunofluorescent p185(her2/neu) was detected in almost all (seven of eight), and ErbB3 expression in a subset (four of eight) of tumors (seven adenomas and one carcinoma). Quantitative PCR also showed abundant ErbB3 mRNA in tumor specimens derived from a rarely encountered prolactin-cell carcinoma. Activation of p185(c-neu)/ErbB3 signaling with heregulin, the ErbB3 ligand, in rat lacto-somatotroph (GH4C1) tumor cells specifically induced prolactin (PRL) mRNA expression approximately 5-fold and PRL secretion approximately 4-fold, whereas growth hormone expression was unchanged. Heregulin (6 nmol/L) induced tyrosine phosphorylation and ErbB3 and p185(c-neu) heterodimerization, with subsequent activation of intracellular ERK and Akt. The Akt signal was specific to ErbB3 activation by heregulin, and was not observed in response to epidermal growth factor activation of epidermal growth factor receptor. Gefitinib, the tyrosine kinase inhibitor, suppressed heregulin-mediated p185(c-neu)/ErbB3 signaling to PRL. Heregulin induction of PRL was also abrogated by transfecting cells with short interfering RNA directed against ErbB3. Pharmacologic inhibition of heregulin-induced phosphoinositide-3-kinase/Akt (with LY294002) and ERK (with U0126) signaling, as well as short interfering RNA-mediated mitogen-activated protein kinase-1 down-regulation, showed ERK signaling as the primary transducer of heregulin signaling to PRL. These results show ErbB3 expression in human prolactinomas and a novel ErbB3-mediated mechanism for PRL regulation in experimental lactotroph tumors. Targeted inhibition of up-regulated p185(c-neu)/ErbB3 activity could be useful for the treatment of aggressive prolactinomas resistant to conventional therapy.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Neuregulin-1/pharmacology , Pituitary Neoplasms/genetics , Prolactinoma/genetics , Actins/genetics , Animals , Female , Glucuronidase/genetics , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Growth Hormone/metabolism , Humans , Pituitary Gland/drug effects , Pituitary Gland/physiology , Pituitary Neoplasms/enzymology , Polymerase Chain Reaction , Prolactin/metabolism , Prolactinoma/enzymology , RNA, Small Interfering/genetics , Rats , Rats, Inbred WF , Transfection , beta 2-Microglobulin/genetics
13.
Cancer Res ; 68(15): 6377-86, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676863

ABSTRACT

Epidermal growth factor (EGF) regulates pituitary development, hormone synthesis, and cell proliferation. Although ErbB receptor family members are expressed in pituitary tumors, the effects of EGF signaling on pituitary tumors are not known. Immunoprecipitation and Western blot confirmed EGF receptor (EGFR) and p185(c-neu) protein expression in GH3 lacto-somatotroph but not in adrenocorticotropic hormone-secreting AtT20 pituitary tumor cells. EGF (5 nmol/L) selectively enhanced baseline ( approximately 4-fold) and serum-induced (>6-fold) prolactin (PRL) mRNA levels, whereas gefitinib, an EGFR antagonist, suppressed serum-induced cell proliferation and Pttg1 expression, blocked PRL gene expression, and reversed EGF-mediated somatotroph-lactotroph phenotype switching. Downstream EGFR signaling by ERK, but not phosphoinositide-3-kinase or protein kinase C, mediated the gefitinib response. Tumors in athymic mice implanted s.c. with GH3 cells resulted in weight gain accompanied by increased serum PRL, growth hormone, and insulin growth factor 1. Gefitinib decreased tumor volumes and peripheral hormone levels by approximately 30% and restored normal mouse body weight patterns. Mice treated with gefitinib exhibited decreased tumor tissue ERK1/2 phosphorylation and down-regulated tumor PRL and Pttg1 mRNA abundance. These results show that EGFR inhibition controls tumor growth and PRL secretion in experimental lacto-somatotroph tumors. EGFR inhibitors could therefore be useful for the control of PRL secretion and tumor load in prolactinomas resistant to dopaminergic treatment, or for those prolactinomas undergoing rare malignant transformation.


Subject(s)
ErbB Receptors/metabolism , Pituitary Neoplasms/pathology , Prolactinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Base Sequence , Blotting, Western , Cell Division , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Primers , Female , Flow Cytometry , Gefitinib , Gene Expression/drug effects , Immunoprecipitation , Ligands , Mice , Mice, Nude , Pituitary Neoplasms/metabolism , Polymerase Chain Reaction , Prolactinoma/metabolism , Quinazolines/pharmacology , Rats
14.
Clin Gastroenterol Hepatol ; 6(3): 360-3, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18255351

ABSTRACT

BACKGROUND & AIMS: The nonapproved use of human growth hormone (HGH) for anti-aging has been increasing. Theoretical concerns for neoplastic potentiation by HGH have been raised, but not proven clinically. METHODS: We report the case of a 68-year-old man with colonic Crohn's disease who was found to have aggressive metastatic colon cancer. The patient had been receiving HGH therapy for anti-aging purposes for 7 years before presentation. Normal and malignant colonic tissue was examined for qualitative and quantitative molecular profiles of growth hormone (GH) and its signaling molecules, using immunohistochemistry and RNA extraction with polymerase chain reaction amplification. RESULTS: Immunoreactivity was more robust in tumor tissue than in normal colon for insulin-like growth factor-1 receptor (IGF-1R) but not for IGF, GH, or GH receptor. RNA extraction with quantitative polymerase chain reaction showed that IGF-1R and vascular endothelial growth factor expression, but not IGF-1, GH receptor, or suppressor of cytokine signaling-2, were higher in tumor than in normal colonic tissue. CONCLUSIONS: Colorectal cancer development concurrent with administration of HGH for anti-aging purposes occurred in an individual already at increased risk for colon cancer. This underscores the need for further investigation of the proneoplastic potential of GH supplementation for anti-aging.


Subject(s)
Aging/drug effects , Colonic Neoplasms/complications , Crohn Disease/complications , Human Growth Hormone/therapeutic use , Aged , Colonic Neoplasms/secondary , Disease Progression , Follow-Up Studies , Humans , Male , Neoplasm Metastasis , Recombinant Proteins
15.
Expert Rev Endocrinol Metab ; 3(2): 207-222, 2008 Mar.
Article in English | MEDLINE | ID: mdl-30764093

ABSTRACT

Constitutive activation of PI(3)K-Akt-mTOR signaling is a frequently occurring event in human cancer and has also been detected in the majority of neuroendocrine tumors (NETs) of the gastroenteropancreatic system. Molecular analysis of NETs suggests, that in addition to mutations in certain tumor-suppressor genes (e.g., PTEN), multiple autocrine growth factor loops contribute to hyperactive PI(3)K-Akt-mTOR signaling, thus promoting unrestricted proliferation and resistance to apoptosis. These insights opened new perspectives for targeted therapy in NETs. In particular, several novel small-molecule inhibitors of tyrosine and serine/threonine kinases have demonstrated potent anti-tumor activity. This review will summarize current knowledge on PI(3)K-Akt-mTOR signaling, its role in proliferation and apoptosis, as well as novel therapeutic approaches targeting PI(3)K-Akt-mTOR pathway components in NET disease.

16.
Am J Physiol Cell Physiol ; 293(3): C1082-92, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17626243

ABSTRACT

The mammalian securin, pituitary tumor-transforming gene (Pttg), regulates sister chromatid separation during mitosis. Mice deficient in Pttg expression exhibit organ-specific hypoplasia of the testis, spleen, pituitary, and postmaturity pancreatic beta-cells, pointing to a possible adult stem cell defect. Bone marrow stem cells (BMSCs) contribute to bone, cartilage, and fat tissue repair and regeneration, and multipotent adult progenitor cells (MAPCs) have broader differentiation ability. Bone marrow cells derived under MAPC conditions are involved in a spectrum of tissue repair. We therefore tested whether Pttg deletion affects stem cell proliferation and differentiation. BMSCs were isolated under MAPC conditions, although unlike MAPCs, wild-type (WT) and Pttg(-/-) BMSCs do not express octamer-binding transcription factor 4 and are stem cell antigen-I positive. WT and Pttg(-/-) cells did not differ in their ability to differentiate into adipogenic, osteogenic, or hepatocyte-like cells or in phenotypic markers. Cells underwent >100 population doublings, with no observed transforming events. Pttg-null BMSCs replicated 27% slower than WT BMSCs, and under hypoxic conditions, this difference widened. Although apoptosis was not enhanced in Pttg(-/-) cells, Pttg(-/-) BMSC senescence-associated beta-galactosidase activity was elevated, consistent with enhanced p21 protein levels. Using gene array assays, DNA repair genes were shown to be upregulated in Pttg(-/-) BMSCs, whereas genes involved in cell cycle progression, including cyclin D(1), were decreased. Separase, the protease regulated by Pttg, has been implicated in DNA damage repair and was downregulated in Pttg(-/-) BMSCs. Separase was constitutively phosphorylated in Pttg(-/-) cells, a modification likely serving as a compensatory mechanism for Pttg deletion. The results indicate that Pttg deletion reduces BMSC proliferation, renders cells more sensitive to hypoxia, and enhances senescent features, thus pointing to a role for Pttg in the maintenance and proliferation of BMSCs.


Subject(s)
Bone Marrow Cells/pathology , Hematopoietic Stem Cells/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Animals , Apoptosis/physiology , Bone Marrow Cells/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Cell Lineage/physiology , Cellular Senescence/physiology , Cyclin D , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/metabolism , DNA-Binding Proteins , Endopeptidases/genetics , Endopeptidases/metabolism , G1 Phase/physiology , G2 Phase/physiology , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , RNA Interference , Resting Phase, Cell Cycle/physiology , Securin , Separase
17.
Hormones (Athens) ; 6(1): 52-61, 2007.
Article in English | MEDLINE | ID: mdl-17324918

ABSTRACT

OBJECTIVE: Modulatory effects of estrogens on both the immune and the coagulation system are only partially understood. In severe infections high estrogen levels have been observed both in men and postmenopausal women and are associated with increased mortality. Monocyte-derived tissue factor (TF) expression can activate the coagulation system and worsen the course of severe infection. T he aim of the current study was to evaluate the in vitro effect of estrogens on differentiation, TF expression and Tumor Necrosis Factor alpha (TNFalpha) release in human monocytes. DESIGN: Isolated peripheral blood monocytes, MM6- and T HP-1 cells were cultured and stimulated by lipopolysaccharides (LPS) in the presence of 17beta-estradiol (E2) and/or calcitriol. Proliferative responses were evaluated by determining the proliferation rate and by cell cycle analysis. Cell surface expression of C D14 and T F was determined by flow cytometry. TNFalpha was determined by ELISA. RESULTS: Although calcitriol induced the expression of the differentiation marker C D14 and decreased the expression of T F in both immature monocytic cell lines and primary monocytes, the LPS stimulation of T F expression was not significantly increased in immature monocytic cells and was decreased in mature monocytes. Calcitriol-treatment increased LPS-induced TNFalpha release in MM6 cells but inhibited TNFalpha release from peripheral blood monocytes. Treatment with E2 did not alter the phenotype or cell proliferation of resting monocytic cells. However, E2-treated monocytic cells and monocytes responded to LPS by increased TF expression and decreased TNFalpha. CONCLUSIONS: The results suggest that estrogens may modulate T F expression and cytokine production by monocytes and may thus be involved, at least in part, in the pathophysiology of acute inflammatory processes associated with high estrogen levels.


Subject(s)
Calcitriol/pharmacology , Estradiol/pharmacology , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Thromboplastin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Lipopolysaccharide Receptors/metabolism , Male , Monocytes/drug effects
18.
Endocr Rev ; 28(2): 165-86, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17325339

ABSTRACT

Pituitary tumor-transforming gene-1 (PTTG1) is overexpressed in a variety of endocrine-related tumors, especially pituitary, thyroid, breast, ovarian, and uterine tumors, as well as nonendocrine-related cancers involving the central nervous, pulmonary, and gastrointestinal systems. Forced PTTG1 expression induces cell transformation in vitro and tumor formation in nude mice. In some tumors, high PTTG1 levels correlate with invasiveness, and PTTG1 has been identified as a key signature gene associated with tumor metastasis. Increasing evidence supports a multifunctional role of PTTG1 in cell physiology and tumorigenesis. Physiological PTTG1 properties include securin activity, DNA damage/repair regulation and involvement in organ development and metabolism. Tumorigenic mechanisms for PTTG1 action involve cell transformation and aneuploidy, apoptosis, and tumorigenic microenvironment feedback. This paper reviews recent advances in our understanding of PTTG1 structure and regulation and addresses known mechanisms of PTTG1 action. Recent knowledge gained from PTTG1-null mouse models and transgenic animals and their potential application to subcellular therapeutic targeting PTTG1 are discussed.


Subject(s)
Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms/genetics , Amino Acid Sequence , Animals , Base Sequence , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Models, Biological , Molecular Sequence Data , Multigene Family/physiology , Securin , Sequence Homology
19.
Pituitary ; 10(1): 1-15, 2007.
Article in English | MEDLINE | ID: mdl-17265188

ABSTRACT

This clinical review summarizes current approaches to diagnosis and treatment of anterior pituitary hormone deficiency. The diagnostic value of endocrine function tests and replacement strategies for hydrocortisone, thyroxine, sex steroids, and growth hormone replacement are reviewed. Female androgen deficiency syndrome and the current role of DHEA and testosterone replacement in women are also discussed.


Subject(s)
Hormone Replacement Therapy/methods , Hypopituitarism/drug therapy , Pituitary Hormones, Anterior/therapeutic use , Adrenal Insufficiency/diagnosis , Adult , Diagnostic Techniques, Endocrine , Female , Human Growth Hormone/deficiency , Human Growth Hormone/therapeutic use , Humans , Hydrocortisone/blood , Hypogonadism/drug therapy , Male , Pituitary Hormones, Anterior/administration & dosage , Testosterone/blood , Testosterone/therapeutic use
20.
Biochem Biophys Res Commun ; 331(2): 543-8, 2005 Jun 03.
Article in English | MEDLINE | ID: mdl-15850793

ABSTRACT

Recently, we have shown that SOCS-1/3 overexpression in hepatic cells abrogates signaling of type I interferons (IFN) which may contribute to the frequently observed IFN resistance of hepatitis C virus (HCV). IFN-lambdas (IL-28A/B and IL-29), a novel group of IFNs, also efficiently inhibit HCV replication in vitro with potentially less hematopoietic side effects than IFN-alpha because of limited receptor expression in hematopoietic cells. To further evaluate the potential of IFN-lambdas in chronic viral hepatitis, we examined the influence of SOCS protein expression on IFN-lambda signaling. First, we show that hepatic cell lines express the IFN-lambda receptor complex consisting of IFN-lambdaR1 (IL-28R1) and IL-10R2. Whereas in mock-transfected HepG2 cells, IL-28A and IL-29 induced STAT1 and STAT3 phosphorylation, overexpression of SOCS-1 completely abrogated IL-28A and IL-29-induced STAT1/3 phosphorylation. Similarly, IL-28A and IL-29 induced mRNA expression of the antiviral proteins 2',5'-OAS and MxA was abolished by overexpression of SOCS-1. In conclusion, we assume that despite antiviral properties of IFN-lambdas, their efficacy as antiviral agents may have similar limitations as IFN-alpha due to inhibition by SOCS proteins.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Interferons/metabolism , Interleukins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Repressor Proteins/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , Antiviral Agents/metabolism , Cell Line, Tumor , Cytokines , DNA-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , Gene Expression , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Humans , Interferons/chemistry , Interferons/genetics , Interferons/immunology , Interleukins/genetics , Interleukins/immunology , Intracellular Signaling Peptides and Proteins/genetics , Myxovirus Resistance Proteins , Phosphorylation , Phosphotyrosine/metabolism , Repressor Proteins/genetics , STAT1 Transcription Factor , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...