Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Signal Transduct ; 2012: 282050, 2012.
Article in English | MEDLINE | ID: mdl-22523681

ABSTRACT

Recently, we reported the combination of multitargeted ErbB1 inhibitor-DNA damage combi-molecules with OCT in order to downregulate ErbB1 and activate SSTRs. Absence of translation to cell kill was believed to be partially due to insufficient ErbB1 blockage and DNA damage. In this study, we evaluated cell response to molecules that damage DNA more aggressively and induce stronger attenuation of ErbB1 phosphorylation. We used three cell lines expressing low levels (U87MG) or transfected to overexpress wildtype (U87/EGFR) or a variant (U87/EGFRvIII) of ErbB1. The results showed that Iressa ± HN2 and the combi-molecules, ZRBA4 and ZR2003, significantly blocked ErbB1 phosphorylation in U87MG cells. Addition of OCT significantly altered cell cycle distribution. Analysis of the DNA damage response pathway revealed strong upregulation of p53 by HN2 and the combi-molecules. Apoptosis was only induced by a 48 h exposure to HN2. All other treatments resulted in cell necrosis. This is in agreement with Akt-Bad pathway activation and survivin upregulation. Despite strong DNA damaging properties and downregulation of ErbB1 phosphorylation by these molecules, the strongest effect of SSTR activation was on cell cycle distribution. Therefore, any enhanced antiproliferative effects of combining ErbB1 inhibition with SSTR activation must be addressed in the context of cell cycle arrest.

2.
Clin Nutr ; 31(1): 85-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21855185

ABSTRACT

BACKGROUND & AIMS: Despite the descriptive presence of cancer cachexia (CC) in clinical practice, the underlying mechanisms and diagnostic definition have not been clearly identified. Recent work, attempting to establish diagnostic and staging criteria for CC, has identified IL-6 as a biomarker. This study aimed to investigate the clinical relevance of plasma levels of four pro-inflammatory cytokines (IL-6, IL-1ß, IL-8 and TNF-α) in advanced cancer patients (ACP) to further establish their potential in the diagnostic definition of CC. METHODS: Blood was obtained from 83 ACP (47 male and 36 female, aged 34-85 years) and analyzed for white blood cells, lymphocytes, C-reactive protein, albumin and cytokines. Subjects completed questionnaires to establish weakness, loss of appetite, fatigue, quality of life and weight loss; completed tests to determine strength, body composition and sarcopenia; and consented to chart review to calculate survival and total days admitted to hospital. RESULTS: This study shows that, in ACP, IL-1ß is better associated with clinical features of the cachectic condition, such as weakness, loss of appetite, weight loss and sarcopenia, than IL-6. CONCLUSION: IL-6 may not best represent the clinical correlates of CC in ACP. Additional cytokines should be considered in the definition of this condition.


Subject(s)
Biomarkers/blood , Cachexia/blood , Interleukin-6/blood , Neoplasms/blood , Adult , Aged , Aged, 80 and over , Albumins/metabolism , Appetite , C-Reactive Protein/metabolism , Cachexia/etiology , Female , Humans , Inflammation/blood , Inflammation/complications , Interleukin-1beta/blood , Interleukin-8/blood , Lymphocytes/metabolism , Male , Middle Aged , Neoplasms/complications , Quality of Life , Sarcopenia/complications , Sarcopenia/metabolism , Surveys and Questionnaires , Tumor Necrosis Factor-alpha/blood , Weight Loss
3.
Biochim Biophys Acta ; 1813(6): 1172-89, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21419811

ABSTRACT

Epidermal growth factor (EGF) regulates normal and tumor cell proliferation via epidermal growth factor receptor (EGFR) phosphorylation, homo- or heterodimerization and activation of mitogen-activated protein kinases (MAPKs) and PI3K/AKT cell survival pathways. In contrast, SST via activation of five different receptor subtypes inhibits cell proliferation and has been potential target in tumor treatment. To gain further insight for the effect of SSTRs on EGFR activated signaling, we determine the role of SSTR1 and SSTR1/5 in human embryonic kidney (HEK) 293 cells. We here demonstrate that cells transfected with SSTR1 or SSTR1/5 negatively regulates EGF mediated effects attributed to the inhibition of EGFR phosphorylation, MAPKs as well as the cell survival signaling. Furthermore, SSTR effects were significantly enhanced in cells when EGFR was knock down using siRNA or treated with selective antagonist (AG1478). Most importantly, the presence of SSTR in addition to modulating signaling pathways leads to the dissociation of the constitutive and EGF induced heteromeric complex of EGFR/ErbB2. Furthermore, cells cotransfected with SSTR1/5 display pronounced effect of SST on the signaling and dissociation of the EGFR/ErbB2 heteromeric complex than the cells expressing SSTR1 alone. Taken together this study provides the first evidence that the presence of SSTR controls EGF mediated cell survival pathway via dissociation of ErbB heteromeric complex. We propose that the activation of SSTR and blockade of EGFR might serve novel therapeutic approach in inhibition of tumor proliferation.


Subject(s)
ErbB Receptors/metabolism , Receptors, Somatostatin/metabolism , Signal Transduction/physiology , Blotting, Western , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , HEK293 Cells , Humans , Immunoprecipitation , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Binding , Protein Multimerization , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines , RNA Interference , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Somatostatin/genetics , Signal Transduction/drug effects , Somatostatin/pharmacology , Tyrphostins/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Endocrinology ; 152(3): 931-45, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21190959

ABSTRACT

Epidermal growth factor through the stimulation of epidermal growth factor receptor (EGFR) plays a critical role in the activation of MAPKs and phosphatidylinositol-3-protein kinase/AKT cell survival pathways attributed in many pathological conditions. At the cellular level, such functions involve EGFR overactivation and phosphorylation. In the present study, we describe that human embryonic kidney-293 cells transfected with somatostatin (SST) receptor 5 (SSTR5) exhibit inhibition of EGFR phosphorylation and modulate MAPK and phosphatidylinositol-3-protein kinase/AKT cell survival signaling. Furthermore, suppression of EGFR by using small interference RNA and an antagonist (AG1478) potentiates the SST effect via activation of SSTR5 on signaling molecules. In wild-type human embryonic kidney-293 cells, EGFR/ErbB2 exists as constitutive heterodimers. The presence of SSTR5 leads to the dissociation of the heteromeric complex of EGFR/ErbB2 and display preferential heterodimerization between SSTR5 and EGFR in an agonist-dependent manner. These findings highlight a new undiscovered mechanism and potential role of SSTR5 to attenuate the EGFR-mediated signaling pathways involved in tumorigenesis. Our data indicate that the activation and/or overexpression of SST receptors along with the inhibition of EGFR will serve as an important therapeutic approach in the treatment of ErbB-positive tumors.


Subject(s)
ErbB Receptors/metabolism , Receptor, ErbB-2/metabolism , Receptors, Somatostatin/metabolism , Signal Transduction/physiology , Apoptosis , Cell Line , Cell Proliferation , ErbB Receptors/chemistry , ErbB Receptors/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/physiology , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Receptors, Somatostatin/genetics
5.
Cell Signal ; 21(3): 428-39, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19070659

ABSTRACT

The role of somatostatin (SST) and epidermal growth factor (EGF) in breast cancer is undisputed; however, the molecular mechanisms underlying their antiproliferative or proliferative effects are not well understood. We initially confirmed that breast tumour tissues express all five somatostatin receptors (SSTR1-5) and four epidermal growth factor receptors (ErbB1-4). Subsequently, to gain insight into the function of SSTRs and ErbBs in oestrogen receptor (ER)-positive (MCF-7) or ERalpha-negative (MDA-MB-231) breast cancer cells, we defined SSTR1, SSTR5 and ErbB1 mRNA and protein expression in these two tumour cell lines. Consistent with previous studies showing SSTR1/SSTR5 heterodimerization and having seen cell-specific and ligand-selective alterations in receptor expression, we next elucidated whether SSTR1 and SSTR5 functionally interact with ErbB1 using pbFRET analysis. We subsequently determined the effects of SST and EGF either alone, or in combination, on selected downstream signalling molecules such as erk1/2, p38 and JNK. Here, we showed that both SST and EGF influenced erk1/2 phosphorylation and that SST modulated the effects of EGF in a cell-specific manner. We also demonstrated agonist-, time and cell-dependent regulation of p38 phosphorylation. We further investigated modulation of Grb2, SOS, Shc, SH-PTP1 and SH-PTP2. ErbB1 adaptor proteins known to play a role in MAPK activation, Shc, Grb2 and SOS, changed in an agonist- and cell-specific manner whereas, SH-PTP1 and SH-PTP2, adaptor proteins reported to interact with SSTRs, translocated from the cytosol to membrane in a cell-specific manner following SST and/or EGF treatment. Although several previous studies have shown crosstalk between RTKs and GPCRs, there are no reports describing SSTR (GPCR) modulation of ErbBs (RTK) in breast cancer. To the best of our knowledge, this is the first report describing crosstalk/interactions between SSTRs and ErbBs.


Subject(s)
ErbB Receptors/metabolism , MAP Kinase Signaling System/physiology , Receptors, Somatostatin/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , ErbB Receptors/drug effects , Female , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Plakins/drug effects , Plakins/metabolism , Protein Transport/drug effects , Protein Transport/physiology , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Somatostatin/drug effects
6.
Mol Cell Endocrinol ; 286(1-2): 251-61, 2008 May 14.
Article in English | MEDLINE | ID: mdl-18308465

ABSTRACT

The biological effects of the neuropeptide somatostatin (SST) are mediated via a family of five somatostatin receptors (SSTRs) belonging to a family of G-protein-coupled receptors (GPCRs). SSTR regulate the secretion of hormones, growth factors, neurotransmission and cell growth in receptor-specific manner. In addition, SST plays an inhibitory role in several mammary cancer models. These effects are mediated both indirectly through inhibition of hormones and growth factors which promote tumor growth as well as directly via SSTRs present on tumor cells to inhibit mitogenic signaling of growth factor receptor kinases leading to growth arrest and induction of apoptosis. Here, we present an overview on the role of SST and its analogs in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Somatostatin/physiology , Somatostatin/physiology , Amino Acid Sequence , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Molecular Sequence Data , Receptors, Somatostatin/agonists , Signal Transduction , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use
7.
Cell Signal ; 19(11): 2304-16, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17706924

ABSTRACT

Somatostatin and dopamine receptors are well expressed and co-localized in several brain regions, suggesting the possibility of functional interactions. In the present study we used a combination of pharmacological, biochemical and photobleaching fluorescence resonance energy transfer (pbFRET) to determine the functional interactions between human somatostatin receptor 2 (hSSTR2) and human dopamine receptor 2 (hD2R) in both co-transfected CHO-K1 or HEK-293 cells as well as in cultured neuronal cells which express both the receptors endogenously. In monotransfected CHO-K1 or HEK-293 cells, D2R exists as a preformed dimer which is insensitive to agonist or antagonist treatment. In control CHO-K1 cells stably co-transfected with hD2R and hSSTR2, relatively low FRET efficiency and weak expression in co-immunoprecipitate from HEK-293 cells suggest the absence of preformed heterooligomers. However, upon treatment with selective ligands, hD2R and hSSTR2 exhibit heterodimerization. Agonist-induced heterodimerization was accompanied by increased affinity for dopamine and augmented hD2R signalling as well as prolonged hSSTR2 internalization. In contrast, cultured striatal neurons display constitutive heterodimerization between D2R and SSTR2, which were agonist-independent. However, heterodimerization in neurons was completely abolished in the presence of the D2R antagonist eticlopride. These findings suggest that hD2R and hSSTR2 operate as functional heterodimers modulated by ligands in situ, which may prove to be a useful model in designing new therapeutic drugs.


Subject(s)
Fluorescence Resonance Energy Transfer , Receptors, Dopamine D2/metabolism , Receptors, Somatostatin/metabolism , Animals , Brain/cytology , Brain/metabolism , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Cyclic AMP/antagonists & inhibitors , Dimerization , Endocytosis , Fluorescence , Humans , Immunoprecipitation , Male , Microscopy, Confocal , Neurons/cytology , Neurons/metabolism , Protein Binding , Protein Structure, Quaternary , Protein Transport , Rats , Rats, Sprague-Dawley
8.
Cancer Cell Int ; 6: 5, 2006 Mar 06.
Article in English | MEDLINE | ID: mdl-16519802

ABSTRACT

BACKGROUND: Somatostatin receptor (SSTR) expression is positively correlated with tumor size and inversely correlated with epidermal growth factor receptor (ErbB) levels and tumor differentiation. In the present study, we compared SSTR1-5 and ErbB1-4 mRNA and protein expression in two breast cancer cell lines: MCF-7 (ER+) and MDA-MB-231 (ERalpha-). RESULTS: All five SSTRs and four ErbBs were variably expressed as both cell surface and cytoplasmic proteins. In both cell lines, SSTR4 and SSTR1 were highly expressed, followed by SSTR2 and SSTR5 with SSTR3 being the least expressed subtype, at the protein level. ErbBs were variably expressed with ErbB1 as the predominant subtype in both cell lines. ErbB1 is followed by ErbB3, ErbB2 and ErbB4 in MCF-7 at both the protein and mRNA levels. In MDA-MB-231 cells, ErbB1 is followed by ErbB2, ErbB4 and ErbB3. Our results indicate significant correlations at the level of mRNA and protein expression in a cell and receptor-specific manner. Using indirect immunofluorescence, we found that, in MCF-7 cells, SSTR5 was the most prominent subtype coexpressed with ErbBs followed by SSTR3, SSTR4, SSTR1 and SSTR2, respectively. In MDA-MB-231 cells, SSTR1 colocalized strongly with ErbBs followed by SSTR5, SSTR4, SSTR3 and SSTR2. ErbBs displayed higher levels of colocalization amongst themselves in MCF-7 cells than in MDA-MB-231 cells. CONCLUSION: These findings may explain the poor response to endocrine therapy in ER-cancer. Differential distribution of SSTR subtypes with ErbBs in breast cancer cells in a receptor-specific manner may be considered as a novel diagnosis for breast tumors.

9.
Biochim Biophys Acta ; 1669(2): 182-92, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15893521

ABSTRACT

We have previously shown that the human somatostatin receptor type 1 (hSSTR1) does not undergo agonist-induced internalization, but is instead up-regulated at the membrane upon prolonged somatostatin (SST) exposure. The deletion of the carboxyterminal C-tail of the receptor completely abolishes up-regulation. To identify molecular signals that mediate hSSTR1 up-regulation, we created mutant receptors with progressive C-tail deletions. Up-regulation was found to be absent in mutants lacking residues Lys359-Ser360-Arg361. Moreover, point mutation of Ser360 to Ala completely abolished up-regulation. The coexpression of wild type hSSTR1 with V53D, a dominant negative mutant of beta-arrestin-1, completely blocked hSSTR1 up-regulation. Further analysis demonstrated that calcium-calmodulin (CaM) dependent kinases were essential for the SST-induced up-regulation response. Like wild type receptors, all mutants failed to internalize after agonist exposure and were able to inhibit forskolin-stimulated cAMP accumulation. Taking these data together, we suggest that SST-induced hSSTR1 up-regulation is critically dependent upon a specific Lys-Ser-Arg sequence in the C-tail of the receptor, with Ser360 being essential. Up-regulation also requires the participation of CaM protein kinases and interactions with beta-arrestins. In contrast, coupling to adenyl cyclase (AC) and internalization occur independently of molecular signals in the receptor's C-tail.


Subject(s)
Arrestins/metabolism , Receptors, Somatostatin/agonists , Serine/metabolism , Up-Regulation , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Arrestins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Humans , Molecular Sequence Data , Mutation , Protein Structure, Secondary , RNA, Messenger/metabolism , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , beta-Arrestin 1 , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...