Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Cancer Cell ; 5(3): 221-30, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15050914

ABSTRACT

Insulin-like growth factors and their receptor (IGF-1R) have been implicated in cancer pathophysiology. We demonstrate that IGF-1R is universally expressed in various hematologic (multiple myeloma, lymphoma, leukemia) and solid tumor (breast, prostate, lung, colon, thyroid, renal, adrenal cancer, retinoblastoma, and sarcoma) cells. Specific IGF-1R inhibition with neutralizing antibody, antagonistic peptide, or the selective kinase inhibitor NVP-ADW742 has in vitro activity against diverse tumor cell types (particularly multiple myeloma), even those resistant to conventional therapies, and triggers pleiotropic antiproliferative/proapoptotic molecular sequelae, delineated by global transcriptional and proteomic profiling. NVP-ADW742 monotherapy or its combination with cytotoxic chemotherapy had significant antitumor activity in an orthotopic xenograft MM model, providing in vivo proof of principle for therapeutic use of selective IGF-1R inhibitors in cancer.


Subject(s)
Hematologic Neoplasms/metabolism , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Antineoplastic Agents , Bone Marrow/metabolism , Enzyme Inhibitors/pharmacology , Flow Cytometry , Gene Expression Profiling , Hematologic Neoplasms/drug therapy , Humans , Multiple Myeloma , Neoplasms/drug therapy , Neoplasms/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Transplantation, Heterologous/pathology , Vascular Endothelial Growth Factor A/metabolism
2.
Mol Vis ; 15: 1418-28, 2009 Jul 25.
Article in English | MEDLINE | ID: mdl-19641635

ABSTRACT

PURPOSE: The pathophysiology of diabetic retinopathy involves leukocyte adhesion to retinal vasculature, early blood-retinal barrier breakdown, capillary nonperfusion, and endothelial cell death. We investigated the involvement of tumor necrosis factor alpha (TNF-alpha) in diabetes-related histopathological changes in two relevant rodent models. METHODS: In short-term studies, Long-Evans rats with streptozotocin-induced diabetes were treated with or without the TNF-alpha inhibitor, etanercept. For long-term studies, tumor necrosis factor receptor I (TNF-RI)-deficient mice and TNF-RII-deficient mice, as well as C57/Bl6 wild-type mice, were fed 30% galactose for up to 20 months. The retinal histopathological alterations of hypergalactosemia were analyzed in trypsin digest preparations. Endothelial cell injury and apoptosis in rat retinas were evaluated by propidium iodide, TUNEL, CytoDeath staining, and DNA fragmentation ELISA. Caspase 3 and 8 activity was evaluated by immunoblotting and quantitative enzymatic activity assay. RESULTS: Etanercept suppressed caspase activation, retinal cell injury, and apoptosis in short-term diabetic rats. Pericyte and endothelial cell loss were also reduced in long-term hypergalactosemic mice. Long-term studies demonstrated that pericyte loss and endothelial cell loss were reduced in comparison to wild-type diabetic controls. CONCLUSIONS: Our study identifies an important role for TNF-alpha in the pathogenesis of signature diabetic retinopathy pathologies and demonstrates that etanercept can inhibit retinal cell death and long-term complication of diabetes. Taken together, our results suggest that etanercept could prove beneficial in preventing both early and late vascular diabetic complications.


Subject(s)
Apoptosis , Diabetic Retinopathy/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/drug effects , Capillaries/drug effects , Capillaries/pathology , Caspase 3/metabolism , Caspase Inhibitors , DNA Fragmentation/drug effects , Diabetic Retinopathy/enzymology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Enzyme Activation/drug effects , Etanercept , Feeding Behavior/drug effects , Galactose/administration & dosage , Galactose/pharmacology , Galactosemias/pathology , Immunoglobulin G/pharmacology , In Situ Nick-End Labeling , Male , Mice , Rats , Rats, Long-Evans , Receptors, Tumor Necrosis Factor , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Retina/drug effects , Retina/enzymology , Retina/pathology , Time Factors
3.
FASEB J ; 21(9): 2113-23, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17400913

ABSTRACT

Heat shock protein (Hsp) 90 inhibitors, such as 17-allylamino-17-demethoxy-geldanamycin (17-AAG), constitute promising novel therapeutic agents. We investigated the anti-inflammatory activity of 17-AAG in endotoxin-induced uveitis (EIU) in rats. After the induction of EIU with a footpad injection of lipopolysaccharide (LPS), female Lewis rats received a single intraperitoneal. (i.p.) injection of 17-AAG or vehicle. Twenty-four hours later, the retinas were extracted and assayed for leukocyte adhesion; blood-retinal barrier breakdown; VEGF, TNF-alpha, IL-1beta, and CD14 protein levels; NF-kappaB and HIF-1alpha activity; hsp90 and 70 levels and expression and phosphorylation of the tight junction proteins ZO-1 and occludin. 17-AAG treatment significantly suppressed the LPS-induced increase in retinal leukocyte adhesion; vascular leakage; NF-kappaB, HIF-1alpha, p38, and PI-3K activity; and VEGF, TNF-alpha, and IL-1beta levels. 17-AAG also suppressed phosphorylation of ZO-1 and occludin by inhibiting their association with p38 and PI-3K. Although 17-AAG treatment did not reduce the LPS-induced increase in total CD14 levels in leukocytes, it significantly decreased membrane CD14 levels. These data suggest that Hsp90 inhibition suppresses several cardinal manifestations of endotoxin-induced uveitis in the rat. 17-AAG has demonstrated a favorable safety profile in clinical trials in cancer patients and represents a promising therapeutic agent for the treatment of inflammatory eye diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Benzoquinones/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lactams, Macrocyclic/therapeutic use , Uveitis, Anterior/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bacterial Toxins/toxicity , Blood-Retinal Barrier/drug effects , Cell Adhesion/drug effects , Cell Membrane/chemistry , Drug Evaluation, Preclinical , Endotoxins/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-1beta/biosynthesis , Interleukin-1beta/blood , Leukocytes/chemistry , Leukostasis/etiology , Leukostasis/prevention & control , Lipopolysaccharide Receptors/blood , Male , Membrane Proteins/metabolism , NF-kappa B/metabolism , Occludin , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Rats , Rats, Long-Evans , Retinal Vasculitis/chemically induced , Retinal Vasculitis/prevention & control , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/drug effects , Uveitis, Anterior/chemically induced , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/blood , Zonula Occludens-1 Protein , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Mol Cancer Ther ; 6(3): 1070-8, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363500

ABSTRACT

B-Raf is an important mediator of cell proliferation and survival signals transduced via the Ras-Raf-MEK-ERK cascade. BRAF mutations have been detected in several tumors, including papillary thyroid carcinoma, but the precise role of B-Raf as a therapeutic target for thyroid carcinoma is still under investigation. We analyzed a panel of 93 specimens and 14 thyroid carcinoma cell lines for the presence of BRAF mutations and activation of the mitogen-activated protein/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway. We also compared the effect of a B-Raf small inhibitory RNA construct and the B-Raf kinase inhibitor AAL881 on both B-Raf wild-type and mutant thyroid carcinoma cell lines. We found a high prevalence of the T1799A (V600E) mutation in papillary and anaplastic carcinoma specimens and cell lines. There was no difference in patient age, B-Raf expression, Ki67 immunostaining, or clinical stage at presentation between wild-type and BRAF(V600E) specimens. Immunodetection of phosphorylated and total forms of MEK and ERK revealed no difference in their phosphorylation between wild-type and BRAF(V600E) patient specimens or cell lines. Furthermore, a small inhibitory RNA construct targeting the expression of both wild-type B-Raf and B-Raf(V600E) induced a comparable reduction of viability in both wild-type and BRAF(V600E) mutant cancer cells. Interestingly, AAL881 inhibited MEK and ERK phosphorylation and induced apoptosis preferentially in BRAF(V600E)-harboring cells than wild-type ones, possibly because of better inhibitory activity against B-Raf(V600E). We conclude that B-Raf is important for the pathophysiology of thyroid carcinomas irrespective of mutational status. Small molecule inhibitors that selectively target B-Raf(V600E) may provide clinical benefit for patients with thyroid cancer.


Subject(s)
Adenocarcinoma, Follicular/genetics , Carcinoma, Papillary/genetics , Isoquinolines/pharmacology , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , Adenocarcinoma, Follicular/pathology , Adenocarcinoma, Follicular/prevention & control , Adult , Aged , Apoptosis/drug effects , Carcinoma/genetics , Carcinoma/pathology , Carcinoma/prevention & control , Carcinoma, Papillary/pathology , Carcinoma, Papillary/prevention & control , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/prevention & control , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Thyroid Neoplasms/pathology , Thyroid Neoplasms/prevention & control , Tumor Cells, Cultured/drug effects
5.
J Clin Endocrinol Metab ; 92(12): 4845-52, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17848408

ABSTRACT

CONTEXT: The Bcl-2 family of proteins regulates apoptosis in various models and may represent a promising therapeutic target in human malignancies. OBJECTIVE/METHODS: We evaluated the sensitivity of thyroid carcinoma cell lines (two papillary, one follicular, two anaplastic, three medullary) in vitro to BH3I-1 and BH3I-2', two cell-permeable inhibitors of the Bcl-2 homology (BH)-3 domain-mediated interaction between proapoptotic and antiapoptotic Bcl-2 family members. The thyroid carcinoma cell line FRO was stably transfected with cDNA for Bcl-2 or constitutively active Akt and evaluated for sensitivity to BH3-domain inhibition. RESULTS: BH3-domain inhibition disrupted the mitochondrial membrane potential in thyroid carcinoma cells, induced caspase-dependent apoptosis, and potently sensitized them to sublethal concentrations of doxorubicin and the proteasome inhibitor bortezomib (Velcade). Overexpression of constitutively active Akt suppressed BH3I-1-induced cell death. Bcl-2-overexpressing FRO cells were more resistant to conventional chemotherapeutic agents (such as doxorubicin) but significantly more sensitive to BH3I-1 than control cells and were found to overexpress caspase-9, caspase-8, Bmf, Bok, and Bik transcripts and express less A1, BRaf, and FLIP transcripts. CONCLUSIONS: Bcl-2 expression protects thyroid carcinomas against chemotherapy-induced apoptosis. Nevertheless, overexpression of Bcl-2 may result in "oncogene addiction" of the cancer cell, which can be exploited by using BH3-domain inhibitors alone or in combination with other agents, including conventional chemotherapeutics (such as doxorubicin) or novel targeted therapies (such as the proteasome inhibitor bortezomib), for the treatment of aggressive thyroid cancer, including the medullary and anaplastic types.


Subject(s)
Carcinoma, Medullary/genetics , Carcinoma/genetics , Genes, bcl-2/genetics , Thyroid Neoplasms/genetics , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , BH3 Interacting Domain Death Agonist Protein/genetics , Blotting, Western , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Colorimetry , Doxorubicin/pharmacology , Humans , Membrane Potentials/physiology , Oncogene Protein v-akt/genetics , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Tetrazolium Salts , Thiazoles , Transcription, Genetic
6.
J Clin Invest ; 109(6): 805-15, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11901189

ABSTRACT

Acute intensive insulin therapy is an independent risk factor for diabetic retinopathy. Here we demonstrate that acute intensive insulin therapy markedly increases VEGF mRNA and protein levels in the retinae of diabetic rats. Retinal nuclear extracts from insulin-treated rats contain higher hypoxia-inducible factor-1alpha (HIF-1alpha) levels and demonstrate increased HIF-1alpha-dependent binding to hypoxia-responsive elements in the VEGF promoter. Blood-retinal barrier breakdown is markedly increased with acute intensive insulin therapy but can be reversed by treating animals with a fusion protein containing a soluble form of the VEGF receptor Flt; a control fusion protein has no such protective effect. The insulin-induced retinal HIF-1alpha and VEGF increases and the related blood-retinal barrier breakdown are suppressed by inhibitors of p38 mitogen-activated protein kinase (MAPK) and phosphatidylinositol (PI) 3-kinase, but not inhibitors of p42/p44 MAPK or protein kinase C. Taken together, these findings indicate that acute intensive insulin therapy produces a transient worsening of diabetic blood-retinal barrier breakdown via an HIF-1alpha-mediated increase in retinal VEGF expression. Insulin-induced VEGF expression requires p38 MAPK and PI 3-kinase, whereas hyperglycemia-induced VEGF expression is HIF-1alpha-independent and requires PKC and p42/p44 MAPK. To our knowledge, these data are the first to identify a specific mechanism for the transient worsening of diabetic retinopathy, specifically blood-retinal barrier breakdown, that follows the institution of intensive insulin therapy.


Subject(s)
Blood-Retinal Barrier/drug effects , DNA-Binding Proteins/metabolism , Diabetes Mellitus/physiopathology , Endothelial Growth Factors/metabolism , Insulin/pharmacology , Lymphokines/metabolism , Nuclear Proteins/metabolism , Retina/metabolism , Animals , Blood-Retinal Barrier/physiology , Cell Nucleus/metabolism , Cells, Cultured , Culture Media, Conditioned , Diabetic Retinopathy/physiopathology , Disease Models, Animal , Drug Implants , Endothelial Growth Factors/genetics , Glucose/metabolism , Glucose/pharmacology , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Insulin/therapeutic use , Lymphokines/genetics , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rats , Rats, Long-Evans , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Retina/cytology , Transcription Factors/metabolism , Transcriptional Activation/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
7.
Invest Ophthalmol Vis Sci ; 48(10): 4706-19, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17898295

ABSTRACT

PURPOSE: To evaluate the potential of proteasome inhibitors, a novel class of antitumor agents, for the treatment of retinoblastoma. The proteasome inhibitor bortezomib (PS-341, Velcade; Millennium Pharmaceuticals, Cambridge, MA), approved by the US Food and Drug Administration for the treatment of multiple myeloma, is being studied for the treatment of several other malignancies. Among other effects, it inactivates the transcription factor nuclear factor-kappaB (NF-kappaB) by blocking the degradation of its inhibitor, IkappaB. NF-kappaB, which is constitutively active in human retinoblastoma cells and promotes their survival, represents a therapeutic target for patients with this malignancy. METHODS: The authors evaluated the effect of bortezomib on the retinoblastoma cell lines Y79 and WERI-Rb1 in vitro using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, flow cytometry with propidium iodide, gene expression profiling, RT-PCR, and immunoblotting. RESULTS: Bortezomib induced caspase-dependent apoptosis in both retinoblastoma cell lines at clinically achievable concentrations. Bortezomib upregulated heat-shock proteins, other stress-response proteins, proapoptotic molecules, cell-cycle regulators, transcription factors, cytokines, and several proteasome subunits and solute carrier proteins, whereas it downregulated antiapoptotic and adhesion molecules. Bortezomib also induced cleavage of caspases, Bid and poly(ADP-ribose) polymerase (PARP), and sensitized retinoblastoma cells to doxorubicin. CONCLUSIONS: Bortezomib induces a stress response and triggers caspase-dependent apoptosis in human retinoblastoma cells at clinically achievable concentrations. This study provides insight into the molecular mechanism(s) of the antitumor activity of bortezomib and a basis for future preclinical studies leading to clinical trials of bortezomib, alone or in combination with conventional chemotherapy, to improve patient outcomes in retinoblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Boronic Acids/pharmacology , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bortezomib , Caspases/metabolism , Cell Proliferation/drug effects , Flow Cytometry , Formazans , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Indicators and Reagents , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinoblastoma/genetics , Retinoblastoma/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts , Tumor Cells, Cultured , Up-Regulation
8.
FASEB J ; 20(10): 1689-91, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16807368

ABSTRACT

Choroidal neovascularization (CNV) is responsible for the severe visual loss in age-related macular degeneration. CNV formation is considered to be due to an imbalance between pro- and antiangiogenic factors that lead to neovascular growth from the choriocapillaris into the subretinal space. To define whether FasL overexpression in retinal pigment epithelial cells (RPE) can inhibit choroidal neovascularization through Fas-FasL-mediated apoptosis, we examined the role of this pathway in a mouse model of laser-induced choroidal neovascularization. FasL was expressed in the retinal pigment epithelium of transgenic mice. Polymerase chain reaction (PCR), immunoblot, and immunohistochemistry confirmed that the transgene FasL was specifically expressed in RPE. The established laser model was used to induce choroidal neovascularization (CNV) in wild-type (WT) and transgenic mice. CNV formation was compared with respect to fluorescein angiographic leakage (at days 0 and 14 after laser injury) and histological appearance. The lesions were assessed on RPE-choroidal flatmounts after CD31-labeling and with confocal microscopy after perfusion with rhodamine-labeled concanavalin A (Con A). Apoptosis was quantified by TUNEL positivity and caspase activation. FasL mRNA and protein were highly expressed in the RPE of the transgenic mice before and after laser photocoagulation. In contrast, FasL was only weakly expressed in the RPE layer of WT C57BL/6J mice. While ruptures of Bruch's membrane and CNV formation were observed histologically two weeks after laser photocoagulation in transgenic as well as control eyes, the shape and size of CNV lesions were reduced in the transgenic mice. The area of leakage was decreased by 70% in FasL transgenic mice compared with WT mice (P<0.005). The number of TUNEL-positive cells was greater in FasL-overexpressing mice and correlated with the expression of activated caspases. Th expression of other antiangiogenic factors such as PEDF remained unchanged. The specific overexpression of FasL in RPE layer reduced CNV formation in our laser model. Our results strongly point to the FasL-Fas pathway as a potential therapeutic target in controlling pathological choroidal neovascularization.


Subject(s)
Choroid/blood supply , Membrane Glycoproteins/genetics , Membrane Glycoproteins/therapeutic use , Neovascularization, Pathologic/prevention & control , Pigment Epithelium of Eye/cytology , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/therapeutic use , Animals , Apoptosis , Bruch Membrane/pathology , Capillaries , Fas Ligand Protein , Lasers/adverse effects , Macular Degeneration , Membrane Glycoproteins/analysis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pigment Epithelium of Eye/chemistry , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Tumor Necrosis Factors/analysis
9.
Clin Cancer Res ; 12(12): 3705-12, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778096

ABSTRACT

PURPOSE: The death receptor Fas is present in thyroid carcinomas, yet fails to trigger apoptosis. Interestingly, Fas has been reported to be actually overexpressed in papillary thyroid carcinomas, suggesting that it may confer a survival advantage. EXPERIMENTAL DESIGN: We investigated the expression and activation status of Fas pathway mediators in thyroid carcinoma cell lines and tumor specimens. RESULTS: All cell lines tested express Fas-associated death domain, procaspase-8, procaspase-9, and procaspase-3; resistance to Fas-mediated apoptosis could not be attributed to lack of any of these apoptosis mediators. Moreover, Fas death domain mutations were not found in our study. The proteasome inhibitors MG132 and PS-341 (bortezomib, Velcade), which lead to accumulation of the nuclear factor kappaB (NF-kappaB) inhibitor IkappaB, did not sensitize SW579 cells to Fas-mediated apoptosis, suggesting that resistance to Fas-mediated apoptosis is not due to proteasome or NF-kappaB activity. Cross-linking of Fas in vitro induced recruitment of Fas-associated death domain-like interleukin-1beta-converting enzyme inhibitory protein (FLIP) instead of procaspase-8. Inhibition of FLIP expression with a FLIP antisense oligonucleotide resulted in significant sensitization to Fas-mediated apoptosis. Fas cross-linking promoted BrdUrd incorporation; activated the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase, NF-kappaB, and activator protein-1 pathways in thyroid carcinoma cells in vitro; and protected cells from tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. We also found that good prognosis papillary thyroid carcinoma specimens exhibited higher immunoreactivity for cleaved (activated) caspase-8 than poor prognosis tumors. CONCLUSIONS: In thyroid carcinomas, the proteolytic cleavage and activation of caspase-8 depends on the balance between expression levels for procaspase-8 and FLIP and correlates with favorable clinical prognosis. Fas may actually stimulate proliferation and confer a survival advantage to thyroid cancer cells.


Subject(s)
Signal Transduction/physiology , Thyroid Neoplasms/pathology , fas Receptor/physiology , Apoptosis , Base Sequence , CASP8 and FADD-Like Apoptosis Regulating Protein , Cell Division , Humans , Intracellular Signaling Peptides and Proteins/physiology , Mutation , Oligonucleotides, Antisense , Receptors, Tumor Necrosis Factor/physiology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/physiopathology , Transfection
10.
J Clin Endocrinol Metab ; 91(10): 4013-21, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16849420

ABSTRACT

CONTEXT: The ubiquitin-proteasome pathway is a major pathway for degradation of intracellular proteins. Proteasome inhibitors constitute a novel class of antitumor agents with preclinical and clinical evidence of activity against hematological malignancies and solid tumors. The proteasome inhibitor bortezomib (PS-341, Velcade) has been approved by the Food and Drug Administration for the treatment of multiple myeloma and is being studied intensely in several other malignancies. Its mechanism of action is complex but appears to include the inhibition of inhibitory-kappaB degradation, which leads to inactivation of the transcriptional factor nuclear factor-kappaB (NF-kappaB). NF-kappaB has been implicated in the pathophysiology of the most aggressive forms of thyroid carcinoma, i.e. medullary and anaplastic. OBJECTIVE AND METHODS: We evaluated the effect of bortezomib on a panel of thyroid carcinoma cell lines, originating from papillary, follicular, anaplastic, and medullary carcinomas. RESULTS: Bortezomib induced apoptosis in medullary and anaplastic cell lines with IC(50) values well within the range of clinically achievable concentrations and much lower than respective IC(50) values for other solid malignancies. Bortezomib inhibited NF-kappaB activity; increased p53, p21, and jun expression; and induced caspase-dependent apoptosis. Sensitivity of thyroid carcinoma cells to bortezomib was partially decreased by overexpression of Bcl-2 or treatment with IGF-I, whereas the combination of bortezomib with chemotherapy (doxorubicin) was synergistic. CONCLUSIONS: These data provide both insights into the molecular mechanisms of antitumor activity of proteasome inhibitors and the rationale for future clinical trials of bortezomib, alone or in combination with conventional chemotherapy, to improve patient outcome in medullary and anaplastic thyroid carcinomas.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Carcinoma, Medullary/drug therapy , Carcinoma/drug therapy , Enzyme Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Thyroid Neoplasms/drug therapy , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Bortezomib , Caspases/physiology , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/analysis , Doxorubicin/pharmacology , Humans , Insulin-Like Growth Factor I/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Thyroid Neoplasms/pathology , Tumor Suppressor Protein p53/analysis
11.
J Clin Endocrinol Metab ; 91(9): 3662-6, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16822827

ABSTRACT

CONTEXT: The epidermal growth factor receptor (EGFR), a transmembrane tyrosine kinase (TK) receptor that mediates proliferation and survival signaling, is expressed in a wide variety of normal and neoplastic tissues. EGFR inhibitors have produced objective responses in patients with non-small-cell lung carcinomas harboring activating EGFR TK domain somatic mutations. OBJECTIVE AND METHODS: Because the EGFR pathway has been reported to be important for the pathophysiology of thyroid carcinoma, we investigated the expression and mutational status of EGFR in 14 thyroid carcinoma cell lines as well as its functional role by evaluating their in vitro sensitivity to AEE788, a new dual-family EGFR/ErbB2 and vascular endothelial growth factor receptor TK inhibitor. We also evaluated the mutational status, mRNA and protein expression, as well as phosphorylation status of EGFR in a panel of thyroid carcinoma specimens. RESULTS: EGFR expression and phosphorylation in the thyroid carcinoma cell lines and tissue specimens were present but not stronger than in noncancerous thyroid tissue. EGFR TK domain mutations were detected in two of 62 histological specimens (3.2%) but not in cell lines. All thyroid carcinoma cell lines were significantly less sensitive (IC(50) at least 25-fold higher) in vitro to AEE788 than a primary culture of EGFR-mutant lung carcinoma cells. CONCLUSIONS: Thyroid carcinoma cells overall are poorly responsive to clinically relevant concentrations of AEE788 in vitro. The presence of EGFR-activating TK domain mutations may identify a small minority of thyroid cancer patients that may benefit from EGFR inhibitors, but additional preclinical evidence of efficacy is needed.


Subject(s)
Carcinoma/drug therapy , ErbB Receptors/antagonists & inhibitors , Purines/pharmacology , Thyroid Neoplasms/drug therapy , Adolescent , Adult , Aged , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Mutation , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism
12.
Clin Cancer Res ; 11(10): 3958-65, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15897598

ABSTRACT

Histone deacetylases (HDAC) and histone acetyltransferases exert opposing enzymatic activities that modulate the degree of acetylation of histones and other intracellular molecular targets, thereby regulating gene expression, cellular differentiation, and survival. HDAC inhibition results in accumulation of acetylated histones and induces differentiation and/or apoptosis in transformed cells. In this study, we characterized the effect of two HDAC inhibitors, suberoylanilide hydroxamic acid (SAHA) and m-carboxycinnamic acid bis-hydroxamide, on thyroid carcinoma cell lines, including lines originating from anaplastic and medullary carcinomas. In these models, both SAHA and m-carboxycinnamic acid bis-hydroxamide induced growth arrest and caspase-mediated apoptosis and increased p21 protein levels, retinoblastoma hypophosphorylation, BH3-interacting domain death agonist cleavage, Bax up-regulation, down-regulation of Bcl-2, A1, and Bcl-x(L) expression, and cleavage of poly(ADP-ribose) polymerase and caspase-8, -9, -3, -7, and -2. Transfection of Bcl-2 cDNA partially suppressed SAHA-induced cell death. SAHA down-regulated the expression of the apoptosis inhibitors FLIP and cIAP-2 and sensitized tumor cells to cytotoxic chemotherapy and death receptor activation. Our studies provide insight into the tumor type-specific mechanisms of antitumor effects of HDAC inhibitors and a framework for future clinical applications of HDAC inhibitors in patients with thyroid cancer, including histologic subtypes (e.g., anaplastic and medullary thyroid carcinomas) for which limited, if any, therapeutic options are available.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/pathology , Cinnamates/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Thyroid Neoplasms/pathology , Apoptosis , Cell Death , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Tumor Cells, Cultured , Vorinostat
13.
Oncogene ; 21(37): 5673-83, 2002 Aug 22.
Article in English | MEDLINE | ID: mdl-12173037

ABSTRACT

Interleukin-6 (IL-6) and insulin-like growth factor-1 (IGF-1) promote the proliferation of multiple myeloma (MM) cells and protect them against dexamethasone (Dex)-induced apoptosis. We have previously shown that Apo2 ligand/TNF-Related apoptosis inducing ligand (Apo2L/TRAIL) induces apoptosis of MM cells, including cells either sensitive or resistant to Dex and cytotoxic drugs, and overcomes the growth and survival effect of IL-6; conversely, NF-kappaB transcriptional activity attenuates their Apo2L/TRAIL-sensitivity. In the current study, we demonstrate that IGF-1 stimulates sustained activation of NF-kappaB and Akt; induces phosphorylation of the FKHRL-1 Forkhead transcription factor; upregulates a series of intracellular anti-apoptotic proteins including FLIP, survivin, cIAP-2, A1/Bfl-1, and XIAP; and decreases Apo2L/TRAIL-sensitivity of MM cells. In contrast, IL-6 does not cause sustained NF-kappaB activation, induces less pronounced Akt activation and FKHRL-1 phosphorylation than IGF-1, and increases the expression of only survivin. Forced overexpression of constitutively active Akt in MM-1S cells reduced their sensitivity to Apo2L/TRAIL and to doxorubicin (Doxo). In contrast, the Akt inhibitor IL-6-Hydroxymethyl-chiro-inositol 2-(R)-2-O-methyl-3-O-octadecylcarbonate induced cell death of both Dex- and Doxo-sensitive and -resistant cells; opposed the protective effect of constitutive Akt activity against Apo2L/TRAIL; and abrogated the NF-kappaB activation, increase of anti-apoptotic proteins and protection against Apo2L/TRAIL induced by IGF-1. These findings therefore define an important role of the Akt pathway in modulating tumor cell responsiveness to Apo2L/TRAIL, delineate molecular mechanisms for the survival effects of IGF-1, and characterize differential pathophysiologic sequelae of IGF-1 vs IL-6 on MM cells. Importantly, they provide the basis for future clinical trials in MM combining conventional or novel agents with strategies designed to neutralize IGF-1.


Subject(s)
Apoptosis , Insulin-Like Growth Factor I/pharmacology , Multiple Myeloma/therapy , NF-kappa B/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/physiology , Apoptosis Regulatory Proteins , DNA-Binding Proteins/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors , Humans , Interleukin-6/pharmacology , Membrane Glycoproteins/pharmacology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt , TNF-Related Apoptosis-Inducing Ligand , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
14.
Invest Ophthalmol Vis Sci ; 46(1): 358-66, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15623796

ABSTRACT

PURPOSE: Fas ligand (FasL) and TNF-related apoptosis-inducing ligand (TRAIL)/Apo2L are members of the TNFalpha family that can trigger apoptosis in susceptible cells via respective death receptors (DRs). FasL cross-links its receptor Fas, resulting in recruitment and proteolytic activation of caspase-8, which initiates the downstream apoptotic cascade. TRAIL signals through its receptors DR4 and DR5, which can activate caspase-8 as well. This study was undertaken to investigate the functional status of the FasL and TRAIL apoptotic pathways in retinoblastoma (Rb) cells. METHODS: The human Rb cell lines Y79 and WERI-Rb1 were evaluated for their response to the Fas cross-linking antibody CH11 and recombinant TRAIL, as well as for cell surface presence and mutational status of Fas, DR4, and DR5 by flow cytometry and genomic DNA sequencing, respectively. The expression of caspase-8 and its inhibitor FLIP, as well as their recruitment to the DR signaling complex were studied by immunoblot analysis. RESULTS: Rb cells express Fas, DR4, and DR5 on their surfaces, yet were resistant to DR-mediated apoptosis. This was not due to DR mutations or secretion of the soluble decoy Fas, antiapoptotic NF-kappaB activity, or FLIP overexpression, but to the absence of caspase-8 expression. The demethylating agent 5-aza-2'-deoxycytidine restored caspase-8 expression and sensitivity to DR-mediated apoptosis. CONCLUSIONS: Rb cells are resistant to DR-mediated apoptosis because of a deficiency in caspase-8 expression secondary to epigenetic gene silencing by overmethylation. The data help delineate the apoptotic pathways in Rb cells and suggest that the combination of demethylating agents with DR-activating modalities, such as TRAIL receptor monoclonal antibodies, may benefit patients with retinoblastoma.


Subject(s)
Apoptosis , Caspases/genetics , Gene Silencing/physiology , Receptors, Tumor Necrosis Factor/metabolism , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Azacitidine/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase 8 , Caspase Inhibitors , Cell Survival , Enzyme Inhibitors/pharmacology , Fas Ligand Protein , Flow Cytometry , Humans , Immunoblotting , Intracellular Signaling Peptides and Proteins/pharmacology , Membrane Glycoproteins/metabolism , NF-kappa B/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand , Retinal Neoplasms/enzymology , Retinoblastoma/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , fas Receptor/metabolism
15.
FASEB J ; 16(3): 438-40, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11821258

ABSTRACT

Leukocyte adhesion to the diabetic retinal vasculature results in blood-retinal barrier breakdown, capillary nonperfusion, and endothelial cell injury and death. Intercellular adhesion molecule-1 (ICAM-1) and the leukocyte integrin CD18 are required for these processes. Diabetes was induced in Long Evans rats, resulting in a two- to threefold increase in retinal leukocyte adhesion. Following one week of diabetes, neutrophil CD11a, CD11b, and CD18 expression was increased significantly, as were retinal ICAM-1 levels. Animals were treated with aspirin, a cyclooxygenase 2 (COX-2) inhibitor (meloxicam), or a soluble tumor necrosis factor alpha (TNF-alpha) receptor/Fc construct (TNFR-Fc, etanercept). High-dose aspirin, etanercept, and high-dose meloxicam each reduced leukocyte adhesion and suppressed blood-retinal barrier breakdown. High-dose aspirin also reduced the expression of CD11a, CD11b, and CD18, whereas meloxicam and etanercept did not. High-dose aspirin, etanercept, and high-dose meloxicam each reduced retinal ICAM-1 expression. Aspirin and meloxicam both lowered retinal TNF-alpha levels. Notably, aspirin, meloxicam, and etanercept did not change retinal vascular endothelial growth factor levels. High-dose aspirin, etanercept and high-dose meloxicam, each suppressed the retinal expression of eNOS and the DNA-binding capacity of retinal nuclear factor-kappaB. High-dose aspirin also suppressed Erk kinase activity, which is involved in CD18 up-regulation. Taken together, these data identify COX-2 and TNF-alpha as operative in the early signature pathologies of diabetic retinopathy, a newly recognized inflammatory disease.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Diabetic Retinopathy/prevention & control , Tumor Necrosis Factor-alpha/metabolism , Animals , Aspirin/pharmacology , Blood-Retinal Barrier/drug effects , Cell Adhesion/drug effects , Cyclooxygenase Inhibitors/pharmacology , Diabetic Retinopathy/blood , Diabetic Retinopathy/immunology , Etanercept , Immunoglobulin G/pharmacology , Intercellular Adhesion Molecule-1/metabolism , Kinetics , Meloxicam , Models, Biological , NF-kappa B/metabolism , Neutrophils/drug effects , Neutrophils/immunology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Rats , Rats, Long-Evans , Receptors, Leukocyte-Adhesion/metabolism , Receptors, Tumor Necrosis Factor , Retina/drug effects , Retina/immunology , Thiazines/administration & dosage , Thiazines/pharmacology , Thiazoles/administration & dosage , Thiazoles/pharmacology
16.
FASEB J ; 18(12): 1450-2, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15231732

ABSTRACT

Diabetic retinopathy is a leading cause of adult vision loss and blindness. Much of the retinal damage that characterizes the disease results from retinal vascular leakage and nonperfusion. Diabetic retinal vascular leakage, capillary nonperfusion, and endothelial cell damage are temporary and spatially associated with retinal leukocyte stasis in early experimental diabetes. Retinal leukostasis increases within days of developing diabetes and correlates with the increased expression of retinal intercellular adhesion molecule-1 (ICAM-1) and CD18. Mice deficient in the genes encoding for the leukocyte adhesion molecules CD18 and ICAM-1 were studied in two models of diabetic retinopathy with respect to the long-term development of retinal vascular lesions. CD18-/- and ICAM-1-/- mice demonstrate significantly fewer adherent leukocytes in the retinal vasculature at 11 and 15 months after induction of diabetes with STZ. This condition is associated with fewer damaged endothelial cells and lesser vascular leakage. Galactosemia of up to 24 months causes pericyte and endothelial cell loss and formation of acellular capillaries. These changes are significantly reduced in CD18- and ICAM-1-deficient mice. Basement membrane thickening of the retinal vessels is increased in long-term galactosemic animals independent of the genetic strain. Here we show that chronic, low-grade subclinical inflammation is responsible for many of the signature vascular lesions of diabetic retinopathy. These data highlight the central and causal role of adherent leukocytes in the pathogenesis of diabetic retinopathy. They also underscore the potential utility of anti-inflammatory treatment in diabetic retinopathy.


Subject(s)
Diabetic Retinopathy/etiology , Diabetic Retinopathy/pathology , Inflammation/complications , Inflammation/pathology , Animals , CD18 Antigens/genetics , CD18 Antigens/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Galactose/blood , Galactose/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Retina/metabolism , Retina/pathology , Time Factors
17.
FASEB J ; 17(1): 76-8, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12475915

ABSTRACT

Diabetic macular edema, resulting from increased microvascular permeability, is the most prevalent cause of vision loss in diabetes. The mechanisms underlying this complication remain poorly understood. In the current study, diabetic vascular permeability (blood-retinal barrier breakdown) is demonstrated to result from a leukocyte-mediated Fas-FasL-dependent apoptosis of the retinal vasculature. Following the onset of streptozotocin-induced diabetes, FasL expression was increased in rat neutrophils (P<0.005) and was accompanied by a simultaneous increase in Fas expression in the retinal vasculature. Static adhesion assays demonstrated that neutrophils from diabetic, but not control, rats induced endothelial cell apoptosis in vitro (P<0.005). The latter was inhibited via an antibody-based FasL blockade (P<0.005). In vivo, the inhibition of FasL potently reduced retinal vascular endothelial cell injury, apoptosis, and blood-retinal barrier breakdown (P<0.0001) but did not diminish leukocyte adhesion to the diabetic retinal vasculature. Taken together, these data are the first to identify leukocyte-mediated Fas-FasL-dependent retinal endothelial cell apoptosis as a major cause of blood-retinal barrier breakdown in early diabetes. These data imply that the targeting of the Fas-FasL pathway may prove beneficial in the treatment of diabetic retinopathy.


Subject(s)
Apoptosis , Blood-Retinal Barrier , Diabetes Mellitus, Experimental/immunology , Diabetic Retinopathy/immunology , Membrane Glycoproteins/antagonists & inhibitors , fas Receptor/physiology , Animals , Cell Adhesion , Cell Death , Coculture Techniques , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Endothelium, Vascular/cytology , Fas Ligand Protein , Leukocytes/immunology , Membrane Glycoproteins/physiology , Models, Immunological , Neutrophils/immunology , Rats , Retina/cytology
18.
J Clin Endocrinol Metab ; 88(11): 5392-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14602779

ABSTRACT

Vascular endothelial growth factor (VEGF) produced by tumor cells potently stimulates endothelial cell proliferation and angiogenesis and plays a key role in the pathophysiology of several neoplasias. Hypoxia activates the VEGF promoter via response elements that bind the transcription factors hypoxia-inducible factor-1 alpha (HIF-1 alpha) and activator protein-1 (AP-1). Yet, the paracrine signaling pathways regulating VEGF production and angiogenesis in thyroid cancer have not been fully elucidated. In this study, we, therefore, investigated the regulation of VEGF production by the thyroid carcinoma cell line SW579. We found that IGF-I up-regulated VEGF mRNA expression and protein secretion. Furthermore, transfection of SW579 cells with vector expressing a constitutively active form of Akt, a major mediator of IGF-I signaling, also stimulated VEGF expression. The IGF-I-induced up-regulation of VEGF production was associated with activation of AP-1 and HIF-1 alpha and was abrogated by phosphatidylinositol 3-kinase inhibitors (wortmannin and LY294002); Jun kinase inhibitor (SP600125); HIF-1 alpha antisense oligonucleotide; or geldanamycin, an inhibitor of the heat shock protein 90 molecular chaperone, which regulates the three-dimensional conformation and function of IGF-I-receptor and Akt. These data indicate that IGF-I stimulates VEGF synthesis in thyroid carcinomas in an Akt-dependent pathway via AP-1 and HIF-1 alpha and provide the framework for clinical use of small-molecule inhibitors, including geldanamycin analogs, to abrogate proangiogenic cascades in thyroid cancer.


Subject(s)
Adenocarcinoma , Insulin-Like Growth Factor I/pharmacology , Protein Serine-Threonine Kinases , Thyroid Neoplasms , Vascular Endothelial Growth Factor A/genetics , Benzoquinones , Cell Line, Tumor/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , JNK Mitogen-Activated Protein Kinases , Lactams, Macrocyclic , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Quinones/pharmacology , Receptor, IGF Type 1/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
19.
Invest Ophthalmol Vis Sci ; 44(5): 2184-91, 2003 May.
Article in English | MEDLINE | ID: mdl-12714660

ABSTRACT

PURPOSE: To investigate the effect of TNF-alpha on leukocyte adhesion, vascular leakage, and apoptotic cell death in endotoxin-induced uveitis (EIU) in the rat. METHODS: EIU was induced in Long-Evans rats by a single footpad injection of lipopolysaccharide (LPS; 350 microg/kg) from Salmonella typhimurium. A single injection of recombinant TNF receptor P75 (etanercept) was given subcutaneously 24 hours before the administration of LPS. Twenty-four hours after administration of LPS, leukocyte adhesion was evaluated in vivo with SLO-acridine orange angiography and ex vivo with concanavalin A lectin staining of retinal flatmounts. Neutrophil activation was quantified by a myeloperoxidase activity assay. Vascular leakage was assessed by Evans blue extravasation. Retinal cell death was assessed with TUNEL staining and quantified with a modified ELISA protocol. Involvement of caspase-3 and -8 was determined by M30 antibody staining, Western blot analysis, and a test for enzymatic activity. RESULTS: Twenty-four hours after the LPS injection, significant increases in leukocyte rolling, adhesion, and activation were observed. In addition, increased levels of apoptosis in the vascular endothelium and the ganglion cell and inner nuclear layers and activation of caspase-8 and -3 were observed. After administration of the TNF-alpha inhibitor, significant reduction in the leukocyte rolling, adhesion, activation, and apoptosis in all the affected layers was observed. The quantitative analysis of vascular leakage revealed a significant decrease after treatment with etanercept. Retinal cell death quantification showed a significant decrease after treatment with the TNF-alpha inhibitor. CONCLUSIONS: Anti-TNF-alpha treatment reduces the LPS-induced increases in leukocyte rolling, adhesion, and vascular leakage in this rat model of inflammatory uveitis. These results suggest the involvement of TNF-alpha in inflammatory uveitis and its potential use as a therapeutic agent in the reduction of ocular inflammation.


Subject(s)
Apoptosis , Leukocytes/physiology , Retina/pathology , Retinal Vessels/metabolism , Tumor Necrosis Factor-alpha/physiology , Uveitis, Posterior/metabolism , Animals , Blood-Retinal Barrier/physiology , Blotting, Western , Capillary Permeability/physiology , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Adhesion/physiology , Endothelium, Vascular/pathology , Endotoxins/toxicity , Enzyme-Linked Immunosorbent Assay , Etanercept , Immunoglobulin G/pharmacology , Immunosuppressive Agents/pharmacology , In Situ Nick-End Labeling , Male , Neutrophil Activation/physiology , Rats , Rats, Long-Evans , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins/pharmacology , Retinal Vessels/pathology , Salmonella typhimurium , Uveitis, Posterior/chemically induced , Uveitis, Posterior/pathology
20.
Invest Ophthalmol Vis Sci ; 44(1): 117-23, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12506063

ABSTRACT

PURPOSE: To investigate the mechanisms governing corneal neovascularization and the appearance of goblet cells in a murine model of limbal insufficiency. METHODS: The spatial and time-dependent relationship between corneal neovascularization and goblet cell density was analyzed in corneal flatmounts. Immunohistochemical detection of the vascular endothelial growth factor (VEGF) receptor Flt-1 (VEGFR1) was performed in paraffin-embedded sections. A transgenic mouse that expresses the reporter gene lacZ targeted to the Flt-1 locus through homologous recombination was used to analyze corneal expression of Flt-1. The presence of soluble and membranous goblet cell Flt-1 mRNA and protein content was assessed with Northern and Western blot analyses, respectively. Finally, systemic adenoviral expression of a soluble Flt-1/Fc construct was used to study the effect of inhibition of VEGF bioactivity on the appearance of goblet cells and neovascularization. RESULTS: Corneal neovascularization preceded the appearance of goblet cells, although both processes overlapped temporally. Flt-1 was abundant in the conjunctiva-like epithelium covering the cornea, as well as in the goblet cells, invading leukocytes, and vasculature. A similar expression pattern was observed in the transgenic mice expressing the lacZ gene downstream from the Flt-1 promoter. Isolated human and rat goblet cells in culture expressed Flt-1 mRNA and protein, as did freshly isolated human conjunctiva. The systemic inhibition of VEGF bioactivity potently suppressed both corneal neovascularization (8.3% +/- 8.1% vs. 41.1% +/- 15.3% corneal area; P < 0.001) and corneal goblet cell density (1.6% +/- 2.5% vs. 12.2% +/- 2.4% corneal area; P < 0.001). CONCLUSIONS: Two important features of corneal conjunctivalization, the appearance of goblet cells and neovascularization, are regulated by VEGF. Both processes are probably mediated, in part, through the Flt-1 receptor. Taken together, these data indicate that an anti-VEGF therapeutic approach may limit the visual loss associated with conjunctivalization of the corneal surface.


Subject(s)
Conjunctiva/metabolism , Corneal Neovascularization/metabolism , Endothelial Growth Factors/metabolism , Goblet Cells/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Count , Conjunctiva/pathology , Corneal Neovascularization/pathology , Epithelial Cells/pathology , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factors , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL