Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
J Ocul Pharmacol Ther ; 29(6): 530-8, 2013.
Article En | MEDLINE | ID: mdl-23573802

PURPOSE: To evaluate endothelial progenitor cell [late outgrowth endothelial progenitor cells (OECs)], vascular endothelial growth factor (VEGF), and stromal cell-derived factor 1α (SDF-1α) plasma levels as potential biomarkers before and during ranibizumab (Lucentis(®)) treatment for neovascular age-related macular degeneration (nvAMD). METHODS: Thirty-one patients with untreated nvAMD presenting for 3 consecutive intravitreal ranibizumab injections and a follow-up visit at 4 weeks intervals were enrolled. Peripheral blood was collected before each injection and at the follow-up visit and OEC clusters were cultured and evaluated according to previously published protocols. VEGF and SDF-1α plasma levels were measured by enzyme-linked immunosorbent assay and compared to values from healthy young and old control. RESULTS: Patients with a high OEC count before treatment presented significantly more often with a short symptom duration and a smaller choroidal neovascularization size. VEGF plasma levels were significantly higher in nvAMD (282.4±195.2 pg/mL) compared to young (45.5±6.8 pg/mL) and old control (46.1±8.5 pg/mL). OEC levels decreased nonsignificantly during ranibizumab treatment, returning to baseline levels after the third injection. VEGF and SDF-1α plasma levels decreased significantly during treatment toward control values. Patients needing retreatment after 3 ranibizumab injections had significantly higher VEGF plasma levels at pretreatment compared to patients not needing further treatment. CONCLUSIONS: The results presented here suggest that VEGF plasma levels may warrant further evaluation regarding biological, therapeutical, and predictive implications in nvAMD.


Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Chemokine CXCL12/blood , Choroidal Neovascularization/drug therapy , Endothelial Cells/pathology , Macular Degeneration/drug therapy , Vascular Endothelial Growth Factor A/blood , Aged , Aged, 80 and over , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Biomarkers/blood , Cell Count , Choroidal Neovascularization/blood , Choroidal Neovascularization/complications , Drug Administration Schedule , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Intravitreal Injections , Macular Degeneration/blood , Macular Degeneration/etiology , Male , Ranibizumab , Treatment Outcome
2.
Melanoma Res ; 21(5): 405-16, 2011 Oct.
Article En | MEDLINE | ID: mdl-21900792

'Cancer stem cells' (CSCs) are tumor cells with stem cell properties hypothesized to be responsible for tumorigenesis, metastatis, and resistance to treatment, and have been identified in different tumors including cutaneous melanoma, using stem cell markers such as CD133. This study explored expression of CD133 and other putative stem cell markers in uveal melanoma. Eight uveal melanoma cell lines were subjected to flow-cytometric (fluorescence-activated cell sorting) analysis of CD133 and other stem cell markers. Eight paraffin-embedded tumors were analyzed by immunohistochemistry for CD133, Pax6, Musashi, nestin, Sox2, ABCB5, and CD68 expressions. Ocular, uveal melanoma, and hematopoietic stem cell distributions of C-terminal and N-terminal CD133 mRNA splice variants were compared by reverse-transcription PCR. Fluorescence-activated cell sorting analysis revealed a population of CD133-positive/nestin-positive cells in cell lines Mel270, OMM 2.3, and OMM2.5. All cell lines studied were positive for nestin, CXCR-4, CD44, and c-kit. Immunohistochemistry identified cells positive for CD133, Pax6, Musashi, nestin, Sox2, ABCB5, and CD68 predominantly at the invading tumor front. C-terminal primers interacting with CD133 splice variant s2 detected a novel variant lacking exon 27. Differential expression of CD133 splice variants was found in iris, ciliary body, retina, and retinal pigment epithelium/choroid as well as in uveal melanoma cell lines. mRNA for nestin, Sox2, and Musashi was present in all studied cell lines. Uveal melanoma such as cutaneous melanoma may therefore contain CSCs. Further experiments are needed to isolate stem cell marker-positive cells, to evaluate their functional properties and to explore therapeutical approaches to these putative CSCs in uveal melanoma.


Antigens, CD/biosynthesis , Glycoproteins/biosynthesis , Melanoma/metabolism , Neoplastic Stem Cells/metabolism , Uveal Neoplasms/metabolism , AC133 Antigen , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Female , Glycoproteins/genetics , Humans , Immunohistochemistry , Intermediate Filament Proteins/biosynthesis , Male , Melanoma/genetics , Melanoma/pathology , Mice , Middle Aged , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/biosynthesis , Nestin , Peptides/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology
3.
Biochim Biophys Acta ; 1813(12): 2145-56, 2011 Dec.
Article En | MEDLINE | ID: mdl-21810446

The novel PKCθ isoform is highly expressed in T-cells, brain and skeletal muscle and originally thought to have a restricted distribution. It has been extensively studied in T-cells and shown to be important for apoptosis, T-cell activation and proliferation. Recent studies showed its presence in other tissues and importance in insulin signaling, lung surfactant secretion, intestinal barrier permeability, platelet and mast-cell functions. However, little information is available for PKCθ activation by gastrointestinal (GI) hormones/neurotransmitters and growth factors. In the present study we used rat pancreatic acinar cells to explore their ability to activate PKCθ and the possible interactions with important cellular mediators of their actions. Particular attention was paid to cholecystokinin (CCK), a physiological regulator of pancreatic function and important in pathological processes affecting acinar function, like pancreatitis. PKCθ-protein/mRNA was present in the pancreatic acini, and T538-PKCθ phosphorylation/activation was stimulated only by hormones/neurotransmitters activating phospholipase C. PKCθ was activated in time- and dose-related manner by CCK, mediated 30% by high-affinity CCK(A)-receptor activation. CCK stimulated PKCθ translocation from cytosol to membrane. PKCθ inhibition (by pseudostrate-inhibitor or dominant negative) inhibited CCK- and TPA-stimulation of PKD, Src, RafC, PYK2, p125(FAK) and IKKα/ß, but not basal/stimulated enzyme secretion. Also CCK- and TPA-induced PKCθ activation produced an increment in PKCθ's direct association with AKT, RafA, RafC and Lyn. These results show for the first time the PKCθ presence in pancreatic acinar cells, its activation by some GI hormones/neurotransmitters and involvement in important cell signaling pathways mediating physiological responses (enzyme secretion, proliferation, apoptosis, cytokine expression, and pathological responses like pancreatitis and cancer growth).


Acinar Cells/drug effects , Gastrointestinal Hormones/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Isoenzymes/metabolism , Neurotransmitter Agents/pharmacology , Pancreas, Exocrine/drug effects , Protein Kinase C/metabolism , Acinar Cells/cytology , Acinar Cells/metabolism , Animals , Blotting, Western , Cells, Cultured , Enzyme Activation , Immunoprecipitation , Isoenzymes/genetics , Male , Pancreas, Exocrine/cytology , Pancreas, Exocrine/metabolism , Phosphorylation/drug effects , Protein Kinase C/genetics , Protein Kinase C-theta , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , raf Kinases/genetics , raf Kinases/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
4.
Mol Vis ; 17: 85-98, 2011 Jan 10.
Article En | MEDLINE | ID: mdl-21245959

PURPOSE: We recently demonstrated increased frequency and growth potential of late outgrowth endothelial progenitor cells (OECs) in patients with neovascular age-related macular degeneration (nvAMD). This study investigated the effects of short- and long-term in vitro inhibition of vascular endothelial growth factor (VEGF) Receptor-2 (VEGFR-2) signaling by SU5416 and other inhibitors of the VEGF signaling pathway in OECs. METHODS: OECs, from the peripheral blood of patients with nvAMD, and human umbilical vein endothelial cells were grown in the presence of SU5416, other VEGFR-2 tyrosine kinase inhibitors (TKIs), and inhibitors of phosphatidylinositol 3'-Kinase (PI3K)/protein kinase B (Akt) and protein kinase C (PKC) in complete angiogenic medium. Apotosis was assessed after 48 h using the fluorescein isothiocyanate Annexin V method. Cell counts were performed for 10 days, and features of senescence were analyzed using senescence-associated ß-galactosidase staining, the telomeric repeat amplification protocol for telomerase activity, Southern blot analysis for mean telomere length, flow cytometric analysis for cell-cycle arrest, and western blot for p53 and p21. Control OECs, cells treated for 7 days with inhibitors, as well as naturally senescent OECs were analyzed for expression of different endothelial antigens, including VEGFR-2 and the receptor for stromal cell-derived factor 1, chemokine receptor 4 (CXCR-4). Migration in vitro to VEGF and stromal cell-derived factor 1 of OECs was assessed. RESULTS: SU5416, other VEGFR-2 TKIs, and inhibitors of PI3K, Akt, and PKC induced apoptosis, inhibited long-term proliferation, reduced telomerase activity, and induced premature senescence and cell-cycle arrest in OECs as well as in human umbilical vein endothelial cells. Naturally senescent cells and cells rendered senescent by VEGFR-2 TKIs had reduced VEGFR-2 and CXCR-4 expression and demonstrated reduced migratory ability to VEGF. CONCLUSIONS: This study demonstrates apoptosis upon short-term inhibition and inhibition of long-term survival of OECs from patients with nvAMD by SU5416, presumably via PI3K/Akt and/or PKC-mediated reduction in telomerase activity and subsequent induction of premature senescence, which is accompanied by impaired endothelial activity. Therefore, induction of premature senescence in endothelial cells may represent a potential therapeutic target in nvAMD.


Cellular Senescence/drug effects , Endothelial Cells/drug effects , Indoles/pharmacology , Macular Degeneration/drug therapy , Pyrroles/pharmacology , Stem Cells/drug effects , Apoptosis , Endothelial Cells/cytology , Humans , Macular Degeneration/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, CXCR4/biosynthesis , Signal Transduction , Stem Cells/cytology , Treatment Outcome , Umbilical Veins/cytology , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Lung Cancer ; 68(2): 154-60, 2010 May.
Article En | MEDLINE | ID: mdl-19628293

The effects of dithiolethione modified valproate, diclofenac and sulindac on non-small cell lung cancer (NSCLC) cells were investigated. Sulfur(S)-valproate and S-diclofenac at 1 microg/ml concentrations significantly reduced prostaglandin (PG)E(2) levels in NSCLC cell lines A549 and NCI-H1299 as did the COX-2 inhibitor DuP-697. In vitro, S-valproate, S-diclofenac and S-sulindac half-maximally inhibited the clonal growth of NCI-H1299 cells at 6, 6 and 15 microg/ml, respectively. Using the MTT assay, 10 microg/ml S-valproate, NO-aspirin and Cay10404, a selective COX-2 inhibitor, but not SC-560, a selective COX-1 inhibitor, inhibited the growth of A549 cells. In vivo, 18mg/kg i.p. of S-valproate and S-diclofenac, but not S-sulindac, significantly inhibited A549 or NCI-H1299 xenograft proliferation in nude mice, but had no effect on the nude mouse body weight. The mechanism by which S-valproate and S-diclofenac inhibited the growth of NSCLC cells was investigated. Nitric oxide-aspirin but not S-valproate caused apoptosis of NSCLC cells. By Western blot, S-valproate and S-diclofenac increased E-cadherin but reduced vimentin and ZEB1 (a transcriptional suppressor of E-cadherin) protein expression in NSCLC cells. Because S-valproate and S-diclofenac inhibit the growth of NSCLC cells and reduce PGE(2) levels, they may prove beneficial in the chemoprevention and/or therapy of NSCLC.


Cadherins/biosynthesis , Carcinoma, Non-Small-Cell Lung/drug therapy , Diclofenac/pharmacology , Lung Neoplasms/drug therapy , Valproic Acid/pharmacology , Anethole Trithione/metabolism , Animals , Cadherins/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase Inhibitors/pharmacology , Diclofenac/chemistry , Dinoprostone/biosynthesis , Dinoprostone/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoxazoles/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Pyrazoles/pharmacology , Sulfones/pharmacology , Valproic Acid/chemistry
7.
Cell Signal ; 21(4): 622-38, 2009 Apr.
Article En | MEDLINE | ID: mdl-19166928

Akt is a central regulator of apoptosis, cell growth and survival. Growth factors and some G-protein-coupled receptors (GPCR) regulate Akt. Whereas growth-factor activation of Akt has been extensively studied, the regulation of Akt by GPCR's, especially gastrointestinal hormones/neurotransmitters, remains unclear. To address this area, in this study the effects of GI growth factors and hormones/neurotransmitters were investigated in rat pancreatic acinar cells which are high responsive to these agents. Pancreatic acini expressed Akt and 5 of 7 known pancreatic growth-factors stimulate Akt phosphorylation (T308, S473) and translocation. These effects are mediated by p85 phosphorylation and activation of PI3K. GI hormones increasing intracellular cAMP had similar effects. However, GI-hormones/neurotransmitters [CCK, bombesin, carbachol] activating phospholipase C (PLC) inhibited basal and growth-factor-stimulated Akt activation. Detailed studies with CCK, which has both physiological and pathophysiological effects on pancreatic acinar cells at different concentrations, demonstrated CCK has a biphasic effect: at low concentrations (pM) stimulating Akt by a Src-dependent mechanism and at higher concentrations (nM) inhibited basal and stimulated Akt translocation, phosphorylation and activation, by de-phosphorylating p85 resulting in decreasing PI3K activity. This effect required activation of both limbs of the PLC-pathway and a protein tyrosine phosphatase, but was not mediated by p44/42 MAPK, Src or activation of a serine phosphatase. Akt inhibition by CCK was also found in vivo and in Panc-1 cancer cells where it inhibited serum-mediated rescue from apoptosis. These results demonstrate that GI growth factors as well as gastrointestinal hormones/neurotransmitters with different cellular basis of action can all regulate Akt phosphorylation in pancreatic acinar cells. This regulation is complex with phospholipase C agents such as CCK, because both stimulatory and inhibitory effects can be seen, which are mediated by different mechanisms.


Gastrointestinal Hormones/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Neurotransmitter Agents/pharmacology , Proto-Oncogene Proteins c-akt/drug effects , Animals , Calcium/pharmacology , Cell Line, Tumor/drug effects , Cyclic AMP/physiology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Male , Pancreas/cytology , Pancreas/drug effects , Pancreas/metabolism , Pancreatic Neoplasms/pathology , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase C/physiology , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/drug effects , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/physiology , Signal Transduction/drug effects , Sincalide/pharmacology , Type C Phospholipases/physiology , src-Family Kinases/physiology
8.
Invest Ophthalmol Vis Sci ; 49(6): 2696-708, 2008 Jun.
Article En | MEDLINE | ID: mdl-18515596

PURPOSE: To evaluate the feasibility of isolating and expanding endothelial progenitor cells (EPCs), in the form of late outgrowth endothelial progenitor cells (OECs), from the peripheral blood of an aged population, particularly patients affected by different forms of AMD. METHODS: Peripheral blood mononuclear cells were collected from young control subjects (n = 18) and from elderly subjects with non-AMD/low-risk dry AMD (n = 15), high-risk dry AMD (n = 6), or neovascular AMD (nvAMD; n = 32); cultured in established conditions; and observed for appearance of OEC clusters and growth characteristics on expansion. Expression of VEGF receptor-2 (KDR) in OECs after expansion was determined by Western blot. Plasma samples of study subjects were analyzed for CRP and VEGF levels. RESULTS: OEC cultures were successfully generated from a similar number of subjects in each group. After adjustment for all other variables, subjects with high-risk dry AMD had a 5.6-fold higher number of OEC clusters per 20 mL blood, and subjects with nvAMD had a 5.1-fold high number than did subjects with non-AMD/low-risk dry AMD (P < 0.05). High-risk dry AMD generated 63 times more (NS) and nvAMD 32-times more (P < 0.05) OECs on expansion of clusters than did non-AMD/low-risk dry AMD. Population doubling occurred significantly faster in cultures from nvAMD eyes compared to non-AMD/low-risk dry AMD eyes. In addition, a significant correlation between the number of OEC clusters, expanded OECs and levels of KDR was demonstrated. CONCLUSIONS: An OEC population was isolated and expanded from the blood of elderly control and AMD-affected patients and demonstrated significantly higher number of initial OEC clusters and expansion potential of OECs in patients at risk for or already affected by nvAMD. OECs may be used for further phenotypic, genetic, and functional analyses in patients with nvAMD.


Endothelium, Vascular/pathology , Macular Degeneration/blood , Stem Cells/pathology , Adult , Aged , Aged, 80 and over , Antigens, CD/metabolism , Biomarkers/metabolism , Blotting, Western , C-Reactive Protein/analysis , Cell Proliferation , Cell Separation , Cells, Cultured , Choroidal Neovascularization/blood , Endothelium, Vascular/metabolism , Female , Flow Cytometry , Humans , Immunophenotyping , Male , Middle Aged , Retinal Vessels , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
Invest Ophthalmol Vis Sci ; 49(5): 2025-9, 2008 May.
Article En | MEDLINE | ID: mdl-18436836

PURPOSE: The neonatal Fc receptor (FcRn) has been known to modulate IgG transport and protect against IgG catabolism, resulting in extension of the serum half-life of IgG. The goal of this study was to localize FcRn receptor expression in the rat's eye. METHODS: The cornea, retina, conjunctiva, ciliary body and iris, retinal pigment epithelium and choroid, and lens were dissected from each rat's eye, and total RNA was purified. The first-strand cDNAs were synthesized and subjected to PCR reaction. For control samples, reverse transcriptase was omitted. A monoclonal antibody against the FcRn heavy chain was used to localize the distribution of the FcRn receptor in ocular tissues. Lymphatic vessels and blood vessels were stained with a rabbit anti-mouse lymphatic vessel endothelial receptor-1 polyclonal antibody and a rabbit anti-human von Willebrand factor polyclonal antibody, respectively. RESULTS: RT-PCR demonstrated expression of FcRn RNA in cornea, retina, conjunctiva, ciliary body and iris, and lens but absence of expression in the retinal pigment epithelium and choroid. Immunohistochemistry and double staining confirmed the expression of FcRn receptor to the conjunctival lymphatic vessels but not in the conjunctival blood vessels. In the ciliary body, the FcRn receptor was found to be expressed in both the nonpigmented ciliary epithelium and the ciliary blood vessels. The expression of FcRn receptor was confirmed in the retinal blood vessels, iris blood vessels, optic nerve vascular structures, corneal epithelium and endothelium, and lens epithelium. CONCLUSIONS: The FcRn receptor is expressed in multiple ocular tissues. The blood-ocular barrier showed FcRn receptor expression, indicating that IgG transport from ocular tissues to the blood system may use this receptor. The role of the FcRn receptor in the anterior segment and the conjunctiva remains unclear.


Eye/metabolism , Histocompatibility Antigens Class I/genetics , RNA, Messenger/metabolism , Receptors, Fc/genetics , Animals , Blood Vessels/metabolism , Gene Expression , Histocompatibility Antigens Class I/metabolism , Lymphatic Vessels/metabolism , Rats , Receptors, Fc/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Stem Cells Dev ; 16(5): 733-45, 2007 Oct.
Article En | MEDLINE | ID: mdl-17999595

The transmembrane protein CD133 is expressed on somatic stem cells of various adult human tissues. To investigate whether human corneal stroma also contains CD133-expressing cells and to analyze their functional features, stromal cells were isolated by collagenase digestion, immunophenotyped, and transferred to different culture systems to determine their stem cell properties as well as their differentiation potentials. For comparison, the embryonic keratocyte cell line EK1.Br, the dermal stromal cell line NHDF, and stromal cells of diseased corneas were studied. On average, 5.3% of the normal stromal cells expressed the stem cell marker CD133 and 3.6% co-expressed CD34. Expression of CD133 but not CD34 was also demonstrated for EK1.Br cells, whereas NHDF cells were negative for both markers. Further analysis of CD133(+) normal corneal cells revealed that a significant proportion displayed a monocytic phenotype with co-expression of CD45 and CD14. In diseased corneas, up to 26.8% of the stromal cells showed expression of CD133, and virtually all CD133(+) cells co-expressed CD14 but not CD45. Moreover, using a standard clonogenic assay, normal stromal cells had the capacity to form colonies of the macrophage lineage. These colonies could be further differentiated into lumican-expressing keratocytes. Our data suggest that the human corneal stroma harbors CD133(+) monocytic progenitor cells, which possess the potential to differentiate into the fibrocytic lineage. Thus, CD133(+) /CD45(+) /CD14(+) cells might represent stromal repair cells that differentiate into keratocytes via a CD133(+)/CD45()/CD14(+) intermediate stage. The findings from our study may shed new light on regenerative processes of the human corneal stroma.


Corneal Stroma/cytology , Wound Healing , AC133 Antigen , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Differentiation , Cell Line , Cell Separation , Clone Cells , Collagenases/metabolism , Colony-Forming Units Assay , Corneal Diseases/pathology , Dermis/cytology , Embryo, Mammalian/cytology , Fibroblasts/cytology , Flow Cytometry , Glycoproteins/metabolism , Hematopoietic System/cytology , Humans , Immunohistochemistry , Immunophenotyping , Limbus Corneae/cytology , Neoplasm Proteins/metabolism , Peptides/metabolism , Phenotype
11.
Invest Ophthalmol Vis Sci ; 48(11): 5184-90, 2007 Nov.
Article En | MEDLINE | ID: mdl-17962472

PURPOSE: To evaluate the angiogenic inhibitory effects of an alpha(v)beta(3)/alpha(v)beta(5) integrin antagonist, EMD478761, released from a polymeric implant in a chick chorioallantoic membrane (CAM) assay and laser-induced experimental choroidal neovascularization (CNV) in rats. METHODS: Polyvinyl alcohol-based reservoir implants releasing EMD478761 were designed for placement onto a CAM or intravitreally in rats. In vitro release rates of the implants were measured using HPLC. Angiogenesis was induced on 10-day-old chick embryos by basic fibroblast growth factor (bFGF), and areas of neovascularization were measured. Experimental CNV was induced in the Brown-Norway rat with a diode laser. EMD478761 or sham microimplants were placed within the vitreous chamber of Brown-Norway rats. Two weeks later, areas of CNV were determined by FITC-dextran staining of choroidal flatmounts. RESULTS: Sustained delivery of EMD478761 significantly inhibited bFGF-induced angiogenesis in CAM, as determined by a reduction in angiogenesis areas, without drug toxicity to the normal CAM vasculature. In an experimental rat model, intravitreal EMD478761 implants significantly suppressed laser-induced CNV compared with intravitreal sham implants, with the mean area reduced by 63% (P < 0.05). CONCLUSIONS: Sustained delivery of EMD478761demonstrates potent antiangiogenic properties in vivo. These results suggest that an EMD478761 implant may be beneficial in the treatment of neovascular ocular diseases.


Angiogenesis Inhibitors/administration & dosage , Benzoxazines/administration & dosage , Chorioallantoic Membrane/blood supply , Choroidal Neovascularization/prevention & control , Integrin alphaVbeta3/antagonists & inhibitors , Integrins/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Receptors, Vitronectin/antagonists & inhibitors , Animals , Chick Embryo , Choroidal Neovascularization/pathology , Chromatography, High Pressure Liquid , Disease Models, Animal , Drug Delivery Systems , Drug Implants , Fibroblast Growth Factor 2/toxicity , Injections , Laser Therapy , Male , Neovascularization, Pathologic/chemically induced , Rats , Rats, Inbred BN , Vitreous Body/drug effects
12.
J Clin Endocrinol Metab ; 92(11): 4394-402, 2007 Nov.
Article En | MEDLINE | ID: mdl-17711922

CONTEXT/OBJECTIVES: The diagnosis of Zollinger-Ellison syndrome requires secretin testing in 60% of patients. Even with secretin, the diagnosis may be difficult because variable responses occur, and 6-30% have negative testing. The basis for variability or negative responses is unclear. It is unknown whether the tumor density of secretin receptors or the presence of a secretin-receptor-variant, which can act as a dominant negative, is important. The aim of this study was to investigate these possibilities. PATIENTS/METHODS: Secretin-receptor and variant mRNA expression was determined in gastrinomas using real-time PCR from 54 Zollinger-Ellison syndrome patients. Results were correlated with Western blotting, secretin-receptor immunohistochemistry, with gastrin-provocative test results and tumoral/clinical/laboratory features. RESULTS: Secretin-receptor mRNA was detectible in all gastrinomas but varied 132-fold with a mean of 0.89 +/- 0.12 molecules per beta-actin. Secretin-receptor PCR results correlated closely with Western blotting (r = 0.95; P < 0.0001) and receptor immunohistochemistry (P = 0.0015; r = 0.71). The variant was detected in all gastrinomas, but levels varied 102-fold and were 72-fold lower than the total. Secretin-receptor levels correlated with variant levels, Deltasecretin, but not Deltacalcium and with tumor location, but not growth, extent, or clinical responses. Variant levels did not correlate with the Deltasecretin. Detailed analysis provides no evidence that variant expression modified the secretin-receptor response or accounted for negative tests. CONCLUSIONS: Secretin-receptor and secretin-receptor-variant expressions occur in all gastrinomas. Because the expression of the total, but not variant, correlated with the secretin results and no evidence for dominant negative activity of the variant was found, our results suggest that the total secretin-receptor density is an important determinant of the secretin test response.


Calcium/metabolism , Gastrinoma/genetics , Gene Expression Regulation/physiology , Pancreatic Neoplasms/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Zollinger-Ellison Syndrome/genetics , Zollinger-Ellison Syndrome/metabolism , Blotting, Western , DNA, Complementary/genetics , Female , Humans , Immunohistochemistry , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Biochim Biophys Acta ; 1773(4): 483-501, 2007 Apr.
Article En | MEDLINE | ID: mdl-17306383

Protein kinase D1 (PKD1) is involved in cellular processes including protein secretion, proliferation and apoptosis. Studies suggest PKD1 is activated by various stimulants including gastrointestinal (GI) hormones/neurotransmitters and growth factors in a protein kinase C (PKC)-dependent pathway. However, little is known about the mechanisms of PKD1 activation in physiologic GI tissues. We explored PKD1 activation by GI hormones/neurotransmitters and growth factors and the mediators involved in rat pancreatic acini. Only hormones/neurotransmitters activating phospholipase C caused PKD1 phosphorylation (S916, S744/748). CCK activated PKD1 and caused a time- and dose-dependent increase in serine phosphorylation by activation of high- and low-affinity CCK(A) receptor states. Inhibition of CCK-stimulated increases in phospholipase C, PKC activity or intracellular calcium decreased PKD1 S916 phosphorylation by 56%, 62% and 96%, respectively. PKC inhibitors GF109203X/Go6976/Go6983/PKC-zeta pseudosubstrate caused a 62/43/49/0% inhibition of PKD1 S916 phosphorylation and an 87/13/82/0% inhibition of PKD1 S744/748 phosphorylation. Expression of dominant negative PKC-delta, but not PKC-epsilon, or treatment with PKC-delta translocation inhibitor caused marked inhibition of PKD phosphorylation. Inhibition of Src/PI3K/MAPK/tyrosine phosphorylation had no effect. In unstimulated cells, PKD1 was mostly located in the cytoplasm. CCK stimulated translocation of total and phosphorylated PKD1 to the membrane. These results demonstrate that CCK(A) receptor activation leads to PKD activation by signaling through PKC-dependent and PKC-independent pathways.


Cholecystokinin/pharmacology , Pancreas, Exocrine/drug effects , Pancreas, Exocrine/enzymology , Protein Kinase C-delta/metabolism , Protein Kinases/metabolism , Signal Transduction/drug effects , Animals , Bombesin/pharmacology , Calcium/deficiency , Carbachol/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Growth Substances/pharmacology , Isoenzymes/antagonists & inhibitors , Male , Mitogen-Activated Protein Kinases/metabolism , Pancreas, Exocrine/cytology , Phosphoserine/metabolism , Phosphotyrosine/metabolism , Protein Kinase C , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Type C Phospholipases/metabolism
14.
J Biol Chem ; 281(49): 37705-19, 2006 Dec 08.
Article En | MEDLINE | ID: mdl-17035232

The activated c-Met receptor has potent effects on normal tissues and tumors. c-Met levels are regulated by hepatocyte growth factor (HGF); however, it is unknown if they can be regulated by gastrointestinal (GI) hormones. c-Met is found in many GI tissues/tumors that possess GI hormone receptors. We studied the effect of GI hormones on c-Met in rat pancreatic acini, which possess both receptors. CCK-8, carbachol, and bombesin, but not VIP/secretin, decreased c-Met. CCK-8 caused rapid and potent c-Met down-regulation and abolished HGF-induced c-Met and Gab1 tyrosine phosphorylation, while stimulating c-Met serine phosphorylation. The effect of cholecystokinin (CCK) was also seen in intact acini using immunofluorescence, in a biotinylated fraction representing membrane proteins, in single acinar cells, in Panc-1 tumor cells, and in vivo in rats injected with CCK. CCK-8 did not decrease cell viability or overall responsiveness. GF109203X, thapsigargin, or their combination partially reversed the effect of CCK-8. In contrast to HGF-induced c-Met down-regulation, the effect of CCK was decreased by a lysosome inhibitor (concanamycin) but not the proteasome inhibitor lactacystin. Inhibitors of clathrin-mediated endocytosis blocked the effect of CCK. HGF but not CCK-8 caused c-Met ubiquitination. These results show CCK and other GI hormones can cause rapid c-Met down-regulation, which occurs by a novel mechanism. These results could be important for c-Met regulation in normal as well as in neoplastic tissue in the GI tract.


Clathrin/metabolism , Gastrointestinal Hormones/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Animals , Bombesin/pharmacology , Calcium/metabolism , Carbachol/pharmacology , Cell Line , Down-Regulation/drug effects , Endocytosis , Hepatocyte Growth Factor/pharmacology , In Vitro Techniques , Lysosomes/metabolism , Male , Pancreas/drug effects , Pancreas/metabolism , Protein Kinase C/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Sincalide/pharmacology
15.
J Hepatol ; 43(5): 845-53, 2005 Nov.
Article En | MEDLINE | ID: mdl-16139387

BACKGROUND/AIMS: Recent studies indicate that after transplantation, circulating bone marrow-derived stem cells migrate into the liver and contribute to liver regeneration. Whether such cells are actively recruited from the bone marrow for liver repair remains to be determined. In this regard, we investigated whether liver resection leads to a release of stem cell marker-positive (+) cells into the peripheral circulation. METHODS: Peripheral blood samples from 11 living liver donors were analyzed by flow cytometry one day before and 12h after partial hepatectomy (PH) using antibodies against CD133, CD34, CD45, CD14, c-kit, bcrp-1. Immunomagnetic separation was performed to select CD133+ cells for functional assays in vitro. RESULTS: A significant increase in the percentage of CD133+ cells could be demonstrated in all donors studied. Unexpectedly, virtually all CD133+ cells coexpressed CD45 and CD14, whereas only a small subset expressed CD34. No significant staining was observed for c-kit and bcrp-1. In culture, immunoselected CD133+ cells displayed characteristics of myelomonocytic precursors. In addition, enriched CD133+ generated an adherent cell population that expressed CK8, alpha-fetoprotein, and human albumin. CONCLUSIONS: PH induces mobilization of a distinct population of myelomonocytic progenitor cells, which have hepatic differentiation potential in vitro, and might play a role in liver regeneration after PH in humans.


Hematopoietic Stem Cells/physiology , Hepatectomy , Liver Regeneration , Liver Transplantation , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Shape , Hematopoietic Stem Cells/cytology , Hepatocytes/cytology , Humans , Immunomagnetic Separation , Living Donors
17.
Ophthalmologica ; 219(3): 167-70, 2005.
Article En | MEDLINE | ID: mdl-15947502

PURPOSE: To assess the safety and efficacy of two topical anesthesia regimes for cataract surgery. METHODS: 21 patients received a combination of 4 times bupivacaine 0.5, oxybuprocaine and diclofenac eyedrops, 18 patients were given a single topical application of lidocaine gel 2%. A single intracameral injection of lidocaine 1% was administered to all subjects. RESULTS: The extent to which the surgeon was bothered by patient motility was graded as low in about two thirds of all procedures. Patients reported lower intraoperative pain levels with a single application of lidocaine gel supplemented with intracameral lidocaine than with a fourfold application of the combination topical anesthesia plus intracameral anesthesia. CONCLUSIONS: A single application of lidocaine gel 2% combined with intracameral anesthesia provides at least as good analgesia than multiple administration of combined topical anesthesia supplemented with intracameral anesthesia and is equally safe.


Anesthesia, Local/methods , Anesthetics, Combined/administration & dosage , Anesthetics, Local/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Diclofenac/administration & dosage , Lidocaine/administration & dosage , Phacoemulsification , Procaine/analogs & derivatives , Aged , Analgesia , Bupivacaine/administration & dosage , Double-Blind Method , Eye Movements , Female , Gels , Humans , Injections , Lens Implantation, Intraocular , Male , Ophthalmic Solutions , Procaine/administration & dosage , Safety , Treatment Outcome
...