Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Ocul Immunol Inflamm ; : 1-4, 2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37787936

ABSTRACT

Macrophage-like cells (MLC) have a fundamental role in the maintenance of immunosurveillance, response to inflammation and tissue injury in the retina. MLC can be visualized in vivo with conventional en face optical coherence tomography (OCT). The aim of this study is to describe this population of cells in active toxoplasmosis. We present two cases of active toxoplasma retinochoroiditis imaged at 2 time points, where the MLC were threshold after image processing and averaging for removing background and noise. In both patients the MLC collocated with the area of ischemia at the level of the choriocapillaris and retinal vessels.

3.
Article in English | MEDLINE | ID: mdl-29666605

ABSTRACT

For a long time, the effects of distinct Eph tyrosine kinase receptors and their ligands, ephrins on the structure, immunophenotype, and development of thymus and their main cell components, thymocytes (T) and thymic epithelial cells (TECs), have been studied. In recent years, the thymic phenotype of mutant mice deficient in several Ephs and ephrins B has been determined. Remarkably, thymic stroma in these animals exhibits important defects that appear early in ontogeny but little alterations in the proportions of distinct lymphoid cell populations. In the present manuscript, we summarize and extend these results discussing possible mechanisms governing phenotypical and functional thymocyte maturation in an absence of the critical T-TEC interactions, concluding that some signaling mediated by key molecules, such as MHCII, CD80, ß5t, Aire, etc. could be sufficient to enable a proper maturation of thymocytes, independently of morphological alterations affecting thymic epithelium.

4.
Histochem Cell Biol ; 146(2): 167-82, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27060907

ABSTRACT

Thymus development is a complex process in which cell-to-cell interactions between thymocytes and thymic epithelial cells (TECs) are essential to allow a proper maturation of both thymic cell components. Although signals that control thymocyte development are well known, mechanisms governing TEC maturation are poorly understood, especially those that regulate the maturation of immature TEC populations during early fetal thymus development. In this study, we show that EphB2-deficient, EphB2LacZ and EphB3-deficient fetal thymuses present a lower number of cells and delayed maturation of DN cell subsets compared to WT values. Moreover, deficits in the production of chemokines, known to be involved in the lymphoid seeding into the thymus, contribute in decreased proportions of intrathymic T cell progenitors (PIRA/B(+)) in the mutant thymuses from early stages of development. These features correlate with increased proportions of MTS20(+) cells but fewer MTS20(-) cells from E13.5 onward in the deficient thymuses, suggesting a delayed development of the first epithelial cells. In addition, in vitro the lack of thymocytes or the blockade of Eph/ephrin-B-mediated cell-to-cell interactions between either thymocytes-TECs or TECs-TECs in E13.5 fetal thymic lobes coursed with increased proportions of MTS20(+) TECs. This confirms, for the first time, that the presence of CD45(+) cells, corresponding at these stages to DN1 and DN2 cells, and Eph/ephrin-B-mediated heterotypic or homotypic cell interactions between thymocytes and TECs, or between TECs and themselves, contribute to the early maturation of MTS20(+) TECs.


Subject(s)
Ephrin-B2/metabolism , Ephrin-B3/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Animals , Ephrin-B2/deficiency , Ephrin-B3/deficiency , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Knockout , Tissue Culture Techniques
5.
Regul Pept ; 144(1-3): 101-8, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-17683807

ABSTRACT

Previous studies have shown that vasoactive intestinal peptide (VIP) and its receptors (VPAC(1) and VPAC(2) receptors) are involved in promotion and growth of many human tumours including breast cancer. Here we investigated whether VIP regulates the expression of the main angiogenic factor, vascular endothelial cell growth factor (VEGF) in human oestrogen-dependent (T47D) and oestrogen-independent (MDA-MB-4687) breast cancer cells. Semiquantitative and quantitative real-time RT-PCRs were used at mRNA level whereas enzyme immunoanalysis was performed at protein level. Both cancer cell lines expressed VIP and VPAC(1) (but not VPAC(2)) receptors that were functional as shown by VIP stimulation of adenylate cyclase activity. VIP induced VEGF expression at both mRNA and protein levels following a time-dependent pattern. The responses were faster in T47D than in MDA-MB-468 cells. The observed VIP regulation of VEGF expression appears to be modulated at least by the cAMP/protein kinase A (PKA) and the phosphoinositide 3-kinase (PI3-K) signalling systems as shown by studies of adenylate cyclase stimulation and using specific kinase inhibitors such as H89 and wortmannin. These actions suggest a proangiogenic potential of VIP in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vasoactive Intestinal Peptide/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Gene Expression , Humans , Phosphatidylinositol 3-Kinases/metabolism , RNA, Messenger/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vasoactive Intestinal Peptide/genetics
6.
Peptides ; 28(9): 1896-901, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17544169

ABSTRACT

We show that vasoactive intestinal peptide (VIP) exerts trophic and proangiogenic activities in experimental prostate cancer in vivo. Nude mice were subcutaneously injected with Matrigel impregnated with LNCaP prostate cancer cells. Cell treatment with 100 nM VIP for 1h before xenograft resulted in increased tumor growth after 8 and, more remarkably, 15 days of injection. The same occurred with the mRNA expression of the main angiogenic factor, vascular endothelial growth factor (VEGF), as shown by real-time RT-PCR quantification. The proangiogenic activity of VIP was further established by showing increases of hemoglobin levels, Masson trichromic staining, and immunohistochemical CD34 staining in tumors excised 15 days after subcutaneous injection of VIP-treated cells as compared to control conditions. All these parameters indicate that VIP increases vessel formation. This xenograft model is a useful tool to study in vivo the effects of VIP-related peptides in tumor growth and development of blood supply as well as their therapeutical potential in prostate cancer.


Subject(s)
Neovascularization, Pathologic/pathology , Prostatic Neoplasms/pathology , Vasoactive Intestinal Peptide/pharmacology , Animals , Antigens, CD34/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Hemoglobins/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Nude , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden , Vascular Endothelial Growth Factor A/genetics , Xenograft Model Antitumor Assays/methods
7.
Mol Cell Endocrinol ; 270(1-2): 8-16, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17434257

ABSTRACT

The effect of vasoactive intestinal peptide (VIP) on cyclooxygenase-2 (COX-2) expression was analyzed in human prostate non-neoplastic (RWPE-1) as well as cancer androgen-dependent (LNCaP) and independent (PC3) cells. The three cell lines expressed VIP mRNA and VIP peptide, as measured by RT-PCR and immunochemistry, which supports an autocrine/paracrine action of VIP in the prostate gland. VIP levels were progressively higher from non-neoplastic to androgen-dependent and independent cells. Real-time RT-PCR and Western-blotting showed that VIP stimulated both COX-2 mRNA and protein expression in a faster manner as prostate cancer stage progressed (i.e. RWPE1

Subject(s)
Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , NF-kappa B/metabolism , Prostatic Neoplasms/pathology , Vasoactive Intestinal Peptide/pharmacology , Cell Line, Tumor , Curcumin/pharmacology , Disease Progression , Gene Expression Regulation, Enzymologic/drug effects , Humans , Male , Prostatic Neoplasms/genetics , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
8.
Mol Cell Endocrinol ; 249(1-2): 116-22, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16563610

ABSTRACT

Vascular endothelial growth factor (VEGF) is a main factor promoting neovascularization (angiogenesis) of solid tumours as prostate carcinoma. Hypoxia stimulates VEGF gene expression by activating the hypoxia-inducible factor-1 (HIF-1alpha). In the present study, the hypoxia-mimicking agent Ni(2+) induced vasoactive intestinal peptide (VIP) expression at both mRNA and peptide levels but it did not modify the expression of VIP receptors (VPAC(1), VPAC(2) and PAC(1) receptors) in androgen-dependent human LNCaP prostate cancer cells. VIP increased the mRNA levels of VPAC(1) and PAC(1) receptors whereas it decreased VPAC(2) receptor mRNA level. These features support that hypoxia up-regulation of VIP gene expression in prostatic carcinoma may lead to VIP regulation of the expression of its receptors by means of autocrine/paracrine mechanisms. Either VIP or hypoxia mimetics with Ni(2+) increased VEGF expression whereas both conditions together resulted in an additive response. It suggests two independent mechanisms for the observed pro-angiogenic activities of VIP and hypoxia. VIP did not stimulate HIF-1alpha mRNA expression but increased the translocation of HIF-1alpha from the cytosolic compartment to the cell nucleus. Moreover, VIP was unable to modify the expression of the HIF-1alpha inhibitor FIH-1 discarding the possibility of an indirect effect of VIP on HIF-1 transactivation.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vasoactive Intestinal Peptide/metabolism , Cell Hypoxia , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mixed Function Oxygenases , Nickel/pharmacology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/genetics , RNA, Messenger/metabolism , Receptors, Vasoactive Intestinal Peptide/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/genetics , Vasoactive Intestinal Peptide/genetics
9.
Gynecol Endocrinol ; 20(6): 327-33, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16019382

ABSTRACT

Vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating peptide (PACAP) bind similarly to VPAC1 and VPAC2 receptors, whereas PACAP binds with higher affinity than VIP to PAC1 receptors. Here we demonstrate by different approaches the expression of the three subclasses of PACAP/VIP receptors in human normal and malignant breast tissue. At the mRNA level, reverse transcription-polymerase chain reaction experiments showed VPAC1 and VPAC2 receptors as well as various isoforms (null, hip/hop) of PAC1 receptors due to alternative splicing. At the protein level, Western blot experiments revealed the three subclasses of receptor although no conclusive differences could be established when comparing control, peritumoral and tumoral tissue samples. Immunohistochemistry showed the distribution of these receptors: they were located at epithelial cells in normal and cancer conditions but also in leukocytes at the stromal level in carcinomatous tissue. A weaker immunostaining of PAC1 receptors in normal tissue and a strong density of the three PACAP/VIP receptor subclasses in cancer tissue may be related to differential expression patterns during breast tumor progression but more samples need to be studied to validate this hypothesis. PAC1, VPAC1 and VPAC2 receptors were functional, as shown by their coupling to adenylate cyclase stimulation: VIP, PACAP-27 and PACAP-38 behaved similarly at this level, whereas both VPAC receptors acted alike as shown by means of specific peptide agonists and antagonists. The present results together with the known presence of PACAP and VIP in the mammary gland support a paracrine/autocrine involvement of both peptides at this level in physiological and pathological conditions, i.e. during malignant transformation.


Subject(s)
Breast Neoplasms/metabolism , Mammary Glands, Human/metabolism , Receptors, Cell Surface/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Transformation, Neoplastic , Female , Humans , Immunohistochemistry , Middle Aged , RNA, Messenger/analysis , Receptors, Cell Surface/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Reverse Transcriptase Polymerase Chain Reaction
10.
Biochim Biophys Acta ; 1744(2): 224-33, 2005 Jun 30.
Article in English | MEDLINE | ID: mdl-15921770

ABSTRACT

The effect of vasoactive intestinal peptide (VIP) on intracellular Ca(2+) levels and its relationship with the expression of c-fos and vascular endothelial growth factor (VEGF) as well as with neuroendocrine (NE) differentiation were investigated in human prostate LNCaP cells. VIP induced the expression of c-fos mRNA as studied by reverse transcription polymerase chain reaction (RT-PCR). It was accompanied by VIP stimulation of c-fos protein synthesis, as measured by Western blot analysis. VIP enhanced intracellular Ca(2+) levels as evaluated using the calcium probe fura-2. VIP regulation of c-fos expression depended on [Ca(2+)](i) concentration since the intracellular calcium chelator BAPTA/AM decreased c-fos expression (both mRNA and protein) to basal levels. As shown by means of real-time RT-PCR, VIP stimulated VEGF mRNA expression: the effect was inhibited by 40% in the presence of curcumin (an inhibitor of AP-1 binding), and it was dependent on Ca(2+) since BAPTA/AM inhibited this VIP action by 43%. Similar observations were made on the effects of BAPTA/AM and curcumin on VIP stimulation of VEGF protein expression. Simultaneous treatment of cells with the protein kinase A inhibitor H89 and BAPTA/AM completely blocked this VIP effect, whereas each agent alone led only to a partial inhibition. In addition, the calcium chelator blocked by 37% the ability of VIP to induce NE cell differentiation as estimated by the observation of neurite development. These features support a VIP signalling pathway that could be mediated through both cAMP and [Ca(2+)](i) increase in prostate LNCaP cancer cells. Moreover, our data suggest the implication of c-Fos on the induction of the main angiogenic factor VEGF since the promoter region of the VEGF gene possesses AP-1 (i.e., c-Fos/c-Jun heterodimer) response elements. This feature represents a link between the nuclear oncogene c-fos, angiogenesis and NE differentiation by means of an initiating signal upon VIP receptors.


Subject(s)
Calcium Signaling/drug effects , Egtazic Acid/analogs & derivatives , Gene Expression Regulation/drug effects , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/genetics , Vasoactive Intestinal Peptide/pharmacology , Calcium/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Chelating Agents/pharmacology , Cyclic AMP/metabolism , Drug Interactions , Egtazic Acid/pharmacology , Humans , Isoquinolines/pharmacology , Male , Neuropeptides/metabolism , Prostatic Neoplasms , RNA, Messenger/drug effects , Sulfonamides/pharmacology , Vascular Endothelial Growth Factor A/metabolism
11.
Int J Oncol ; 26(6): 1629-35, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15870879

ABSTRACT

Vasoactive intestinal peptide (VIP) functions as a mitogenic agent in the human prostate gland acting by autocrine/paracrine mechanisms. Here we extend knowledge on the VIP system (expression of VIP and VIP receptors, functionality of VIP receptors) at this level by analyzing the differences between human normal prostate and prostate carcinoma specimens. RT-PCR showed the expression of mRNA for VIP in normal and malignant tissues, whereas VIP levels, as measured by enzyme immuno-analysis, were about two times higher in adenocarcinoma samples. Real-time RT-PCR indicated a minor expression of VPAC2 receptors in the prostate gland, as well as the overexpression of VPAC1 and PAC1 receptors in malignant tissue specimens. Radio-labeled binding experiments with [125I]VIP showed an increased number of VIP binding sites (2.5 times for the high- and 1.7 times for the low-affinity sites) during malignant transformation, whereas the affinity values were unaffected. The receptors were functional in control and cancer tissues as shown by the ability of increasing VIP doses to stimulate adenylate cyclase activity. Interestingly, JV-1-53 (a GHRH-related peptide analog) (at 0.1 microM) behaved as a potent VIP antagonist since it inhibited by 60% the maximal VIP effect upon the enzyme activity. The results further explain the mechanisms of the autocrine/paracrine actions of VIP in human prostate and prostatic carcinoma through the observation of differences between healthy tissue and malignant transformation. Moreover, present data support the potential usefulness of VIP and/or synthetic peptide analogs for diagnostic or radiotherapeutic purposes as well as for long-term peptide therapy in this malignancy.


Subject(s)
Growth Hormone-Releasing Hormone/analogs & derivatives , Growth Hormone-Releasing Hormone/pharmacology , Prostatic Neoplasms/chemistry , Receptors, Vasoactive Intestinal Peptide/analysis , Vasoactive Intestinal Peptide/antagonists & inhibitors , Vasoactive Intestinal Peptide/analysis , Aged , Humans , Immunoenzyme Techniques , Male , Middle Aged , Prostatic Neoplasms/therapy , RNA, Messenger/analysis , Receptors, Cell Surface/analysis , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide , Receptors, Vasoactive Intestinal Peptide/genetics , Receptors, Vasoactive Intestinal Peptide, Type II , Receptors, Vasoactive Intestinal Polypeptide, Type I , Reverse Transcriptase Polymerase Chain Reaction
12.
Eur J Pharmacol ; 515(1-3): 20-7, 2005 May 16.
Article in English | MEDLINE | ID: mdl-15913603

ABSTRACT

Vanilloid receptor subtype-1 (TRPV1), the founding member of the vanilloid receptor-like transient receptor potential channel family, is a non-selective cation channel that responds to noxious stimuli such as low pH, painful heat and irritants. In the present study, we show, as means of reverse transcriptase-polymerase chain reaction and Western blot analysis, that the vanilloid TRPV1 receptor is expressed in the prostate epithelial cell lines PC-3 and LNCaP as well as in human prostate tissue. The kinetic parameters inferred from [(125)I]-resiniferatoxin binding were in concordance with data of TRPV1 receptors expressed in other tissues. The contribution of the endogenously expressed TRPV1 channel to intracellular calcium concentration increase in the prostate cells was studied by measuring changes in Fura-2 fluorescence by fluorescence microscopy. Addition of capsaicin, (R)-methanandamide and resiniferatoxin to prostate cells induced a dose-dependent increase in the intracellular calcium concentration that was reversed by the vanilloid TRPV1 receptor antagonist capsazepine. These results indicate that the vanilloid TRPV1 receptor is expressed and functionally active in human prostate cells.


Subject(s)
Gene Expression/genetics , Ion Channels/genetics , Prostate/metabolism , Arachidonic Acids/metabolism , Arachidonic Acids/pharmacology , Binding, Competitive/drug effects , Blotting, Western , Calcium/metabolism , Capsaicin/metabolism , Capsaicin/pharmacology , Cell Line, Tumor , Diterpenes/metabolism , Dose-Response Relationship, Drug , Humans , Iodine Radioisotopes , Ion Channels/agonists , Ion Channels/metabolism , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Radioligand Assay , Reverse Transcriptase Polymerase Chain Reaction , TRPV Cation Channels
13.
Prostate ; 63(1): 44-55, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15468165

ABSTRACT

BACKGROUND: Neuroendocrine (NE) differentiation in prostate cancer has been correlated with unfavorable clinical outcome. The mechanisms by which prostate cancer acquires NE properties are poorly understood, but several signaling pathways have been proposed. We have previously observed that vasoactive intestinal peptide (VIP) stimulates cAMP production mainly through VPAC(1) receptor, inducing NE differentiation in LNCaP cells. The aim of this study was to analyze the mechanisms involved in this process. METHODS: Reverse transcriptase (RT)-polymerase chain reaction (PCR), quantitative real-time RT-PCR, Western blotting, and immunocytochemistry were performed. RESULTS: LNCaP cells produce VIP, as demonstrated by RT-PCR and immunocytochemistry. VIP induced NE differentiation of LNCaP cells at a time as short as 1 hr of treatment, and the same occurred with the expression and secretion of neuronal-specific enolase (NSE, a NE differentiation marker). These effects were faster than those exerted by serum-deprivation. VIP induced extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation and NE differentiation by PKA-dependent and independent pathways, since the PKA inhibitor H89 partially blocked VIP-induced NE differentiation and did not affect ERK1/2 phosphorylation. mitogen-activated protein kinase kinase (MEK) and phosphoinositide 3-kinase (PI3K) appear to be also involved since the inhibitors PD98059 and wortmannin abolished ERK1/2 phosphorylation and decreased NE differentiation induced by VIP. Moreover, VIP activated Ras suggesting the involvement of a Ras-dependent pathway. CONCLUSIONS: VIP behaves as autocrine/paracrine factor in LNCaP cells by inducing NE differentiation through PKA, ERK1/2, and PI3K.


Subject(s)
Carcinoma, Neuroendocrine , MAP Kinase Signaling System/drug effects , Neuroprotective Agents/pharmacology , Prostatic Neoplasms , Vasoactive Intestinal Peptide/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism
14.
Regul Pept ; 119(1-2): 69-75, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15093699

ABSTRACT

Vasoactive intestinal peptide (VIP) upregulates the expression of vascular endothelial cell growth factor (VEGF(189), VEGF(165) and VEGF(121)) mRNAs in human prostate cancer LNCaP cells, as shown by reverse transcriptase-polymerase chain reaction (RT-PCR). Real-time RT-PCR indicated that the effect was maximal by 1-2 h and must be accounted for increased transcription since VIP decreased VEGF(165) mRNA stability. VIP stimulated VEGF(165) protein synthesis as measured by ELISA. VIP regulation of VEGF expression was mediated by VPAC(1) receptor and was cAMP/protein kinase A (PKA) dependent. Phosphoinositide 3-kinase (PI3-K) and mitogen-activated protein kinase MEK1/2 systems may also be involved as shown with specific kinase inhibitors. These actions together with the observation of VIP-induced neuroendocrine differentiation in LNCaP cells suggest a proangiogenic potential of VIP in prostate cancer.


Subject(s)
Neurosecretory Systems/pathology , Prostatic Neoplasms/pathology , Vascular Endothelial Growth Factor A/biosynthesis , Vasoactive Intestinal Peptide/metabolism , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Neovascularization, Pathologic , Phosphatidylinositol 3-Kinases/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Vascular Endothelial Growth Factor A/metabolism
15.
Int J Oncol ; 24(3): 725-30, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14767559

ABSTRACT

The mechanisms by which luteinizing hormone-releasing hormone (LH-RH) antagonists act on extra-pituitary tissues are poorly understood. In view of extensive use of Cetrorelix in gynecology and oncology, we investigated its effects on signal transduction pathways of G-protein coupled receptors and adenylate cyclase which are involved in a huge array of cellular events including normal and pathological cell proliferation. Thirty days after a single i.m. injection of 3 mg Cetrorelix pamoate depot to female rats, normal or ovariectomized, we evaluated the effects of this chronic treatment on the expression of alphas and alphai G-protein subunits in the ovary, breast and pituitary, as well as the adenylate cyclase response in vitro to LH-RH, vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating peptide (PACAP). Varied patterns of response to Cetrorelix, depending on the gland and estrogenic status were observed. Western blot analysis showed a modest decrease of alphas and a modest increase of alphai G-protein subunit levels in ovary, a marked increase of alphas and alphai levels in breast, and a lack of effect on alphas/alphai levels in pituitary. In the ovary, adenylate cyclase activity was not changed by in vitro addition of LH-RH, but the responses to VIP and PACAP increased after Cetrorelix treatment. In the breast, chronic administration of the LH-RH antagonist decreased the adenylate cyclase response to PACAP, which returned to normal after ovariectomy. In the pituitary, Cetrorelix abolished the stimulatory effect of VIP upon adenylate cyclase activity. Thus, the LH-RH antagonist Cetrorelix exerted selective modifications at different steps of the G-protein coupled receptors/adenylate cyclase system of signal transduction in the rat ovary, breast and pituitary.


Subject(s)
Adenylyl Cyclases/metabolism , Breast/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropin-Releasing Hormone/pharmacology , Hormone Antagonists/pharmacology , Ovary/metabolism , Pituitary Gland/metabolism , Animals , Blotting, Western , Cell Division , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Female , Gonadotropin-Releasing Hormone/metabolism , Neuropeptides/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide , Protein Structure, Tertiary , Rats , Rats, Wistar , Signal Transduction , Vasoactive Intestinal Peptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...