Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Invest Ophthalmol Vis Sci ; 55(4): 2130-9, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24595385

ABSTRACT

PURPOSE: Uveal melanoma (UM) tumors require large doses of radiation therapy (RT) to achieve tumor ablation, which frequently results in damage to adjacent normal tissues, leading to vision-threatening complications. Approximately 50% of UM patients present with activating somatic mutations in the gene encoding for G protein αq-subunit (GNAQ), which lead to constitutive activation of downstream pathways, including protein kinase C (PKC). In this study, we investigated the impact of small-molecule PKC inhibitors bisindolylmaleimide I (BIM) and sotrastaurin (AEB071), combined with ionizing radiation (IR), on survival in melanoma cell lines. METHODS: Cellular radiosensitivity was determined by using a combination of proliferation, viability, and clonogenic assays. Cell-cycle effects were measured by flow cytometry. Transcriptomic and proteomic profiling were performed by quantitative real-time PCR, reverse-phase protein array analysis, and immunofluorescence. RESULTS: We found that the PKC inhibitors combined with IR significantly decreased the viability, proliferation, and clonogenic potential of GNAQ(mt), but not GNAQ(wt)/BRAF(mt) cells, compared with IR alone. Combined treatment increased the antiproliferative and proapoptotic effects of IR in GNAQ(mt) cells through delayed DNA-damage resolution and enhanced induction of proteins involved in cell-cycle arrest, cell-growth arrest, and apoptosis. CONCLUSIONS: Our preclinical results suggest that combined modality treatment may allow for reductions in the total RT dose and/or fraction size, which may lead to better functional organ preservation in the treatment of primary GNAQ(mt) UM. These findings suggest future clinical trials combining PKC inhibitors with RT in GNAQ(mt) UM warrant consideration.


Subject(s)
DNA, Neoplasm/genetics , GTP-Binding Protein alpha Subunits/genetics , Melanoma/enzymology , Mutation , Protein Kinase Inhibitors/pharmacology , Uveal Neoplasms/enzymology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Proliferation , Cell Survival , Combined Modality Therapy , Flow Cytometry , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits/radiation effects , GTP-Binding Protein alpha Subunits, Gq-G11 , Humans , Melanoma/genetics , Melanoma/therapy , Radiation, Ionizing , Real-Time Polymerase Chain Reaction , Uveal Neoplasms/genetics , Uveal Neoplasms/therapy
2.
Invest Ophthalmol Vis Sci ; 52(10): 7248-55, 2011 Sep 14.
Article in English | MEDLINE | ID: mdl-21828154

ABSTRACT

PURPOSE: Inhibitors of B-Raf and MEK kinases hold promise for the management of cutaneous melanomas harboring BRAF mutations. BRAF mutations are rare in uveal melanomas (UMs), but somatic mutations in the G protein α subunits Gαq and Gα11 (encoded by GNAQ and GNA11, respectively) occur in a mutually exclusive pattern in ∼80% of UMs. The impact of B-Raf and MEK inhibitors on Gα-mutant UMs remains unknown. METHODS: The impact of the B-Raf inhibitor PLX4720, the MEK inhibitor AZD6244, and the Akt inhibitor MK2206 on UM cell lines was assessed with the use of cell viability, proliferation, and apoptosis assays and immunoblot analysis. RESULTS: BRAF-mutant UM cells were sensitive to both PLX4720 and AZD6244, undergoing cell cycle arrest but not apoptosis. UM cells with a Gα-protein mutation (GNAQ or GNA11) were mildly sensitive to AZD6244 but completely resistant to PLX4720. In fact, PLX4720 paradoxically increased ERK phosphorylation in Gα-mutant UM cells. The combination of AZD6244 with PLX4720 had synergistic anticancer activity in BRAF-mutant cells but not in Gα-mutant cells. The Akt inhibitor MK2206 sensitized BRAF-mutant cells to both PLX4720 and AZD6244 and sensitized Gα-mutant cells to AZD6244 but did not overcome the resistance of the Gα-mutant cells to PLX4720. CONCLUSIONS: The response of UM cells to inhibition of B-Raf, MEK, and Akt depends on their genotype. Future use of such targeted therapies in clinical trials of UM patients will require careful design and patient selection based on genotype to provide personalized and effective therapy.


Subject(s)
MAP Kinase Kinase Kinases/antagonists & inhibitors , Melanoma/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Uveal Neoplasms/genetics , Apoptosis/drug effects , Benzimidazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Genotype , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Immunoblotting , In Situ Nick-End Labeling , Indoles/pharmacology , MAP Kinase Kinase Kinases/metabolism , Melanoma/pathology , Precision Medicine , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sulfonamides/pharmacology , Uveal Neoplasms/pathology
3.
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
4.
Invest Ophthalmol Vis Sci ; 50(10): 4898-904, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19553613

ABSTRACT

PURPOSE: The pathophysiology of diabetic retinopathy is mediated by leukocyte adhesion to the vascular endothelium of the diabetic retina, which results in endothelial injury, blood-retina barrier breakdown, and capillary nonperfusion. Leukocyte adhesion is triggered by the interaction of vascular endothelium adhesion molecules, such as ICAM-1, with leukocyte integrins, such as CD18. Inhibition of ICAM-1/CD18 signaling suppresses but does not completely abolish the cardinal manifestations of diabetic retinopathy, suggesting a role for additional adhesion molecules. Integrin alpha 4 (CD49d), in complex with integrin beta1, forms very late antigen-4 (VLA-4), which interacts with vascular cell adhesion molecule-1. The authors have now studied the role of integrin alpha 4/CD49d in the pathogenesis of diabetic retinopathy. METHODS: Diabetes mellitus was induced in Long Evans rats with streptozotocin, and an anti-alpha 4 integrin/CD49d neutralizing antibody was injected 5 and 10 days later. Two weeks after streptozotocin administration, vascular leakage was quantified with the Evans Blue technique. Leukostasis was measured with a static adhesion assay ex vivo and the FITC-lectin perfusion method in vivo. Retinal VEGF and TNF-alpha levels and NF-kappaB activity were measured by ELISA. RESULTS: Blockade of alpha 4 integrin/CD49d attenuated the diabetes-induced upregulation of NF-kappaB activation, VEGF, and TNF-alpha protein levels and reduced significantly diabetes-induced leukocyte adhesion and vascular leakage. CONCLUSIONS: These data identify alpha 4 integrin/CD49d as a mediator of leukocyte adhesion and the resultant early signature abnormalities of diabetic retinopathy. Inhibition of this signaling pathway may hold promise for clinical activity in patients with diabetes.


Subject(s)
Diabetic Retinopathy/etiology , Diabetic Retinopathy/metabolism , Integrin alpha4/physiology , Animals , Antibodies, Blocking , Blood-Retinal Barrier , Capillary Permeability , Cell Adhesion , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Leukocytes/physiology , Leukostasis/etiology , Leukostasis/metabolism , Male , NF-kappa B/metabolism , Rats , Rats, Long-Evans , Retinal Vessels/cytology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
5.
Invest Ophthalmol Vis Sci ; 50(9): 4072-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19387079

ABSTRACT

PURPOSE: To characterize the molecular sequelae induced in retinoblastoma (Rb) cells by histone deacetylase inhibitors (HDACIs). Hydroxamic acid-based HDACIs such as vorinostat (suberoylanilide hydroxamic acid) induce the differentiation and apoptosis of transformed cells. Vorinostat has demonstrated significant anticancer activity against hematologic and solid tumors at doses well tolerated by patients and has been approved for the treatment of patients with cutaneous T-cell lymphoma. METHODS: The authors evaluated the effects of the HDACIs vorinostat and m-carboxycinnamic acid bis-hydroxamide on the Rb cell lines Y79 and WERI-Rb1 with the use of the MTT assay, BrdU incorporation assay, flow cytometry, immunoblotting, gene-expression profiling, quantitative RT-PCR, and NF-kappaB DNA-binding assay. RESULTS: Both HDACIs were effective against both Rb cell lines, inducing growth arrest and apoptosis in vitro. Vorinostat increased p53 expression and activated caspases -8, -9 and -3, whereas caspase inhibition abrogated vorinostat-induced apoptosis. Vorinostat downregulated baseline NF-kappaB activity and potentiated the activity of the DNA-damaging chemotherapeutic doxorubicin. Gene expression profiling and qRT-PCR demonstrated that vorinostat modulated the mRNA levels of genes important for signal transduction, cell cycle, cellular metabolism, stress response, apoptosis, extracellular matrix synthesis, and cell differentiation. Notably, several transcripts involved in the ephrin and Notch signaling pathways were upregulated. CONCLUSIONS: HDACIs, such as vorinostat, induce caspase-dependent apoptosis in Rb cells, downregulate baseline NF-kappaB activity, and potentiate the effectiveness of conventional chemotherapy. The finding that vorinostat augments the effectiveness of doxorubicin provides a rationale for future clinical studies looking at the use of vorinostat in combination with conventional chemotherapy in Rb.


Subject(s)
Apoptosis , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Retinal Neoplasms/pathology , Retinoblastoma/pathology , Acetylation , Bromodeoxyuridine/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation , Cinnamates/pharmacology , Doxorubicin/pharmacology , Drug Synergism , Flow Cytometry , Gene Expression Profiling , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Immunoblotting , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Retinal Neoplasms/enzymology , Retinoblastoma/enzymology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism , Vorinostat
6.
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
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 ; 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
9.
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
10.
Graefes Arch Clin Exp Ophthalmol ; 245(2): 258-66, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16741710

ABSTRACT

BACKGROUND: The benzoquinoid antibiotic 17-allylaminogeldanamycin (17-AAG) inhibits the Ras/Raf/MEK and PI3-Kinase signaling pathways and down-regulates vascular endothelial factor expression. Here we use a mouse model of oxygen-induced retinopathy to investigate the effect of 17-AAG on retinal neovascularization and vascular recovery. MATERIAL AND METHODS: C57BL/6 mice were exposed to 75% oxygen from postnatal day 7 (P7) to P12 and recovered in room air thereafter. Beginning with P12 mice were treated for 5 days by daily IP injection of 17-AAG (12.5 mg/kg body weight) micro dispersed in an emulsion of 4% Lipoid EPC, 5% sucrose, and 0.9% NaCl or Wortmannin (100 microg/kg body weight). On P17, the retinal vascular and avascular area, neovascular blood vessel tufts, and main vessel tortuosity were quantified after perfusion of the mice with FITC-Concanavalin A. The mRNA levels of VEGF, angiopoietin 1 and 2 were quantified by real-time RT-PCR. RESULTS: After 17-AAG treatment, a reduction of the vascular area was measured from 37.8 +/- 5.2% to 30.8 +/- 5.7% (P = 0.005), and an increase of the avascular area from 10.8 +/- 5.6% to 20.3 +/- 6.6% (P = 0.001). No alteration of the vascular pattern, the number of blood vessel tufts and the main vessel tortuosity was achieved by treatment with the PI-3 kinase inhibitor Wortmannin. After treatment with 17-AAG, the numbers of tufts (127.9 33.2) were different from the controls (173.7 +/- 55.2, P = 0.035), but not the main vessel tortuosity. No significant change in VEGF and angiopoietin 1 mRNA expression could be achieved with either of the treatments. Wortmannin treatment also did not change the angiopoietin 2 mRNA level, whereas the level was reduced in 17-AAG treated mice retina from 436-fold (+/- 64) to 200-fold (+/-55) (P = 0.035). CONCLUSION: An IP injection of 17-AAG is able to reduce angioproliferative retinopathy in a mouse model for oxygen-induced retinopathy. Our data indicate that the mechanism does not involve a direct or indirect reduction of the VEGF mRNA level, but acts downstream of the VEGF pathway. Thus, 17-AAG probably does not work by PI-3 kinase inhibition but via the Ras/Raf/MEK pathway. These data underline the potential utility of tyrosine kinase inhibitors in hypoxia induced neovascularization.


Subject(s)
Benzoquinones/therapeutic use , Disease Models, Animal , Lactams, Macrocyclic/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Retinal Neovascularization/drug therapy , Retinopathy of Prematurity/drug therapy , Angiopoietin-1/genetics , Angiopoietin-2/genetics , Animals , Female , Fluorescein Angiography , Humans , Infant, Newborn , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Oxygen/toxicity , RNA, Messenger/metabolism , Retinal Neovascularization/diagnosis , Retinal Vessels/drug effects , Retinal Vessels/pathology , Retinopathy of Prematurity/diagnosis , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics
11.
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
12.
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
13.
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
14.
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
15.
Blood ; 107(3): 1092-100, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16234364

ABSTRACT

We show that multiple myeloma (MM), the second most commonly diagnosed hematologic malignancy, is responsive to hsp90 inhibitors in vitro and in a clinically relevant orthotopic in vivo model, even though this disease does not depend on HER2/neu, bcr/abl, androgen or estrogen receptors, or other hsp90 chaperoning clients which are hallmarks of tumor types traditionally viewed as attractive clinical settings for use of hsp90 inhibitors, such as the geldanamycin analog 17-AAG. This class of agents simultaneously suppresses in MM cells the expression and/or function of multiple levels of insulin-like growth factor receptor (IGF-1R) and interleukin-6 receptor (IL-6R) signaling (eg, IKK/NF-kappaB, PI-3K/Akt, and Raf/MAPK) and downstream effectors (eg, proteasome, telomerase, and HIF-1alpha activities). These pleiotropic proapoptotic effects allow hsp90 inhibitors to abrogate bone marrow stromal cell-derived protection on MM tumor cells, and sensitize them to other anticancer agents, including cytotoxic chemotherapy and the proteasome inhibitor bortezomib. These results indicate that hsp90 can be targeted therapeutically in neoplasias that may not express or depend on molecules previously considered to be the main hsp90 client proteins. This suggests a more general role for hsp90 in chaperoning tumor- or tissue-type-specific constellations of client proteins with critical involvement in proliferative and antiapoptotic cellular responses, and paves the way for more extensive future therapeutic applications of hsp90 inhibition in diverse neoplasias, including MM.


Subject(s)
Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Multiple Myeloma/metabolism , Rifabutin/analogs & derivatives , Signal Transduction/drug effects , Androgens/metabolism , Apoptosis/drug effects , Benzoquinones , Boronic Acids/pharmacology , Bortezomib , Drug Synergism , Estrogens/metabolism , Fusion Proteins, bcr-abl/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , I-kappa B Kinase/metabolism , Lactams, Macrocyclic , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Pyrazines/pharmacology , Receptor, ErbB-2/metabolism , Receptor, IGF Type 1/biosynthesis , Receptors, Interleukin-6/biosynthesis , Rifabutin/pharmacology , Tumor Cells, Cultured , raf Kinases/metabolism
16.
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
17.
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
18.
Am J Pathol ; 165(2): 457-69, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277220

ABSTRACT

Diabetic retinopathy is a leading cause of blindness in the Western world. Aberrant intercellular adhesion molecule-1 expression and leukocyte adhesion have been implicated in its pathogenesis, raising the possibility of an underlying chronic inflammatory mechanism. In the current study, the role of insulin-like growth factor (IGF)-I in these processes was investigated. We found that systemic inhibition of IGF-I signaling with a receptor-neutralizing antibody, or with inhibitors of PI-3 kinase (PI-3K), c-Jun kinase (JNK), or Akt, suppressed retinal Akt, JNK, HIF-1alpha, nuclear factor (NF)-kappaB, and AP-1 activity, vascular endothelial growth factor (VEGF) expression, as well as intercellular adhesion molecule-1 levels, leukostasis, and blood-retinal barrier breakdown, in a relevant animal model. Intravitreous administration of IGF-I increased retinal Akt, JNK, HIF-1alpha, NF-kappaB, and AP-1 activity, and VEGF levels. IGF-I stimulated VEGF promoter activity in vitro, mainly via HIF-1alpha, and secondarily via NF-kappaB and AP-1. In conclusion, IGF-I participates in the pathophysiology of diabetic retinopathy by inducing retinal VEGF expression via PI-3K/Akt, HIF-1alpha, NF-kappaB, and secondarily, JNK/AP-1 activation. Taken together, these in vitro and in vivo signaling studies thus identify potential targets for pharmacological intervention to preserve vision in patients with diabetes.


Subject(s)
Diabetic Retinopathy/metabolism , Insulin-Like Growth Factor I/physiology , JNK Mitogen-Activated Protein Kinases , Pigment Epithelium of Eye/metabolism , Animals , Blood-Retinal Barrier/drug effects , Blood-Retinal Barrier/physiology , Diabetic Retinopathy/pathology , Electrophoretic Mobility Shift Assay , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , Intercellular Adhesion Molecule-1/metabolism , MAP Kinase Kinase 4 , Male , Mitogen-Activated Protein Kinase Kinases/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Long-Evans , Signal Transduction , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...