Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Commun Biol ; 6(1): 447, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185301

ABSTRACT

Efficacy of immune checkpoint inhibitors in cancers can be limited by CD8 T cell dysfunction or HLA-I down-regulation. Tumor control mechanisms independent of CD8/HLA-I axis would overcome these limitations. Here, we report potent CD4 T cell-mediated tumor regression and memory responses in humanized immune system (HIS) mice implanted with HT-29 colorectal tumors. The regressing tumors showed increased CD4 cytotoxic T lymphocyte (CTL) infiltration and enhanced tumor HLA-II expression compared to progressing tumors. The intratumoral CD4 T cell subset associated with tumor regression expressed multiple cytotoxic markers and exhibited clonal expansion. Notably, tumor control was abrogated by depletion of CD4 but not CD8 T cells. CD4 T cells derived from tumor-regressing mice exhibited HLA-II-dependent and tumor-specific killing ex vivo. Taken together, our study demonstrates a critical role of human CD4 CTLs in mediating tumor clearance independent of CD8 T cells and provides a platform to study human anti-tumor immunity in vivo.


Subject(s)
Neoplasms , T-Lymphocytes, Cytotoxic , Humans , Mice , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Neoplasms/metabolism
2.
Mol Cancer Ther ; 18(11): 2051-2062, 2019 11.
Article in English | MEDLINE | ID: mdl-31395688

ABSTRACT

In the tumor microenvironment, multiple inhibitory checkpoint receptors can suppress T-cell function, thereby enabling tumor immune evasion. Blockade of one of these checkpoint receptors, PD-1, with therapeutic antibodies has produced positive clinical responses in various cancers; however, the efficacy of this approach can be further improved. Simultaneously targeting multiple inhibitory checkpoint receptors has emerged as a promising therapeutic strategy. Here, we report the development and characterization of REGN3767, a fully human IgG4 antibody targeting LAG-3, another inhibitory receptor on T cells. REGN3767 binds human and monkey LAG-3 with high affinity and specificity and blocks the interaction of LAG-3 with its ligand, MHC class II. In an engineered T-cell/antigen-presenting cell bioassay, REGN3767 alone, or in combination with cemiplimab (REGN2810, human anti-PD-1 antibody), blocked inhibitory signaling to T cells mediated by hLAG-3/MHCII in the presence of PD-1/PD-L1. To test the in vivo activity of REGN3767 alone or in combination with cemiplimab, we generated human PD-1xLAG-3 knockin mice, in which the extracellular domains of mouse Pdcd1 and Lag3 were replaced with their human counterparts. In these humanized mice, treatment with cemiplimab and REGN3767 showed increased efficacy in a mouse tumor model and enhanced the secretion of proinflammatory cytokines by tumor-specific T cells. The favorable pharmacokinetics and toxicology of REGN3767 in nonhuman primates, together with enhancement of antitumor efficacy of anti-PD-1 antibody in preclinical tumor models, support its clinical development.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antigens, CD/chemistry , Antigens, CD/genetics , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/genetics , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/metabolism , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Gene Knock-In Techniques , Haplorhini , Histocompatibility Antigens Class II/metabolism , Humans , Mice , Neoplasms/genetics , Protein Binding/drug effects , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , Lymphocyte Activation Gene 3 Protein
3.
Mol Cancer Ther ; 16(5): 861-870, 2017 05.
Article in English | MEDLINE | ID: mdl-28265006

ABSTRACT

The Programmed Death-1 (PD-1) receptor delivers inhibitory checkpoint signals to activated T cells upon binding to its ligands PD-L1 and PD-L2 expressed on antigen-presenting cells and cancer cells, resulting in suppression of T-cell effector function and tumor immune evasion. Clinical antibodies blocking the interaction between PD-1 and PD-L1 restore the cytotoxic function of tumor antigen-specific T cells, yielding durable objective responses in multiple cancers. This report describes the preclinical characterization of REGN2810, a fully human hinge-stabilized IgG4(S228P) high-affinity anti-PD-1 antibody that potently blocks PD-1 interactions with PD-L1 and PD-L2. REGN2810 was characterized in a series of binding, blocking, and functional cell-based assays, and preclinical in vivo studies in mice and monkeys. In cell-based assays, REGN2810 reverses PD-1-dependent attenuation of T-cell receptor signaling in engineered T cells and enhances responses of human primary T cells. To test the in vivo activity of REGN2810, which does not cross-react with murine PD-1, knock-in mice were generated to express a hybrid protein containing the extracellular domain of human PD-1, and transmembrane and intracellular domains of mouse PD-1. In these mice, REGN2810 binds the humanized PD-1 receptor and inhibits growth of MC38 murine tumors. As REGN2810 binds to cynomolgus monkey PD-1 with high affinity, pharmacokinetic and toxicologic assessment of REGN2810 was performed in cynomolgus monkeys. High doses of REGN2810 were well tolerated, without adverse immune-related effects. These preclinical studies validate REGN2810 as a potent and promising candidate for cancer immunotherapy. Mol Cancer Ther; 16(5); 861-70. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Cell Proliferation/drug effects , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/genetics , Animals , Antibodies, Monoclonal, Humanized/immunology , Cell Line, Tumor , Gene Knock-In Techniques , Humans , Immunotherapy , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
4.
Cancer Res ; 76(8): 2327-39, 2016 04 15.
Article in English | MEDLINE | ID: mdl-26921327

ABSTRACT

Anti-VEGF therapies benefit several cancer types, but drug resistance that limits therapeutic response can emerge. We generated cell lines from anti-VEGF-resistant tumor xenografts to investigate the mechanisms by which resistance develops. Of all tumor cells tested, only A431 (A431-V) epidermoid carcinoma cells developed partial resistance to the VEGF inhibitor aflibercept. Compared with the parental tumors, A431-V tumors secreted greater amounts of IL6 and exhibited higher levels of phospho-STAT3. Notably, combined blockade of IL6 receptor (IL6R) and VEGF resulted in enhanced activity against A431-V tumors. Similarly, inhibition of IL6R enhanced the antitumor effects of aflibercept in DU145 prostate tumor cells that displays high endogenous IL6R activity. In addition, post hoc stratification of data obtained from a clinical trial investigating aflibercept efficacy in ovarian cancer showed poorer survival in patients with high levels of circulating IL6. These results suggest that the activation of the IL6/STAT3 pathway in tumor cells may provide a survival advantage during anti-VEGF treatment, suggesting its utility as a source of response biomarkers and as a therapeutic target to heighten efficacious results. Cancer Res; 76(8); 2327-39. ©2016 AACR.


Subject(s)
Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Heterografts , Humans , Interleukin-6/antagonists & inhibitors , Mice , Receptors, Vascular Endothelial Growth Factor , Recombinant Fusion Proteins/pharmacology
5.
Int J Oncol ; 34(1): 79-87, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19082480

ABSTRACT

Approval of the anti-vascular endothelial growth factor (VEGF) antibody bevacizumab by the FDA in 2004 reflected the success of this vascular targeting strategy in extending survival in patients with advanced cancers. However, consistent with previous reports that experimental tumors can grow or recur during VEGF blockade, it has become clear that many patients treated with VEGF inhibitors will ultimately develop progressive disease. Previous studies have shown that disruption of VEGF signaling in tumors induces remodeling in surviving vessels, and link increased expression of angiopoietin-1 (Ang-1) with this process. However, overexpression of Ang-1 in different tumors has yielded divergent results, restricting angiogenesis in some systems while promoting it in others. These data raise the possibility that effects of Ang-1/Tie-2 may be context-dependent. Expression of an Ang-1 construct (Ang1*) did not significantly change tumor growth in our model prior to treatment, although vessels exhibited changes consistent with increased Tie-2 signaling. During inhibition of VEGF, however, both overexpression of Ang1* and administration of an engineered Ang-1 agonist (Bow-Ang1) strikingly protected tumors and vasculature from regression. In this context, Ang-1/Tie-2 activation limited tumor hypoxia, increased vessel caliber, and promoted recruitment of mural cells. Thus, these studies support a model in which activation of Tie-2 is important for tumor and vessel survival when VEGF-dependent vasculature is stressed. Understanding such mechanisms of adaptation to this validated form of therapy may be important in designing regimens that make the best use of this approach.


Subject(s)
Angiopoietin-1/genetics , Gene Expression Regulation/physiology , Kidney Neoplasms/blood supply , Neovascularization, Pathologic/pathology , Receptor, TIE-2/genetics , Sarcoma, Ewing/blood supply , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Blotting, Western , Cell Hypoxia , Cell Line, Tumor , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Kidney Neoplasms/pathology , Mice , Mice, Nude , Phosphorylation , Polymerase Chain Reaction , Sarcoma, Ewing/pathology , Transfection , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/metabolism
6.
Kidney Int ; 74(3): 300-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18480750

ABSTRACT

The loss of interstitial capillaries is a feature of several experimental models of renal disease and this contributes to secondary kidney injury. Angiopoietin-1 is a secreted growth factor which binds to Tie-2 present on endothelia to enhance cell survival thereby stabilizing capillary architecture in-vitro. Previous studies showed that angiopoietin-1 prevented renal capillary and interstitial lesions following experimental ureteric obstruction. We tested here the effect of angiopoietin-1 treatment on capillary loss and associated tubulointerstitial damage known to follow recovery from folic acid-induced tubular necrosis and acute renal injury. We found that delivery of angiopoietin-1 by adenoviral vectors stabilized peritubular capillaries in folic acid nephropathy but this was accompanied by profibrotic and inflammatory effects. These results suggest that the use of endothelial growth factor therapy for kidney disease may have varying outcomes that depend on the disease model tested.


Subject(s)
Angiopoietin-1/adverse effects , Fibrosis/chemically induced , Inflammation/chemically induced , Kidney Tubular Necrosis, Acute/drug therapy , Adenoviridae/genetics , Angiopoietin-1/administration & dosage , Angiopoietin-1/therapeutic use , Animals , Disease Models, Animal , Folic Acid/adverse effects , Genetic Vectors , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Kidney Diseases/pathology , Kidney Tubular Necrosis, Acute/chemically induced , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Renal Circulation/drug effects
7.
Proc Natl Acad Sci U S A ; 103(42): 15491-6, 2006 Oct 17.
Article in English | MEDLINE | ID: mdl-17030814

ABSTRACT

Angiopoietin (Ang)-2, a context-dependent agonist/antagonist for the vascular-specific Tie2 receptor, is highly expressed by endothelial cells at sites of normal and pathologic angiogenesis. One prevailing model suggests that in these settings, Ang-2 acts as an autocrine Tie2 blocker, inhibiting the stabilizing influence of the Tie2 activator Ang-1, thereby promoting vascular remodeling. However, the effects of endogenous Ang-2 on cells that are actively producing it have not been studied in detail. Here, we demonstrate that Ang-2 expression is rapidly induced in endothelial cells by the transcription factor FOXO1 after inhibition of the phosphatidylinositol 3-kinase/Akt pathway. We employ RNAi and blocking antibodies to show that in this setting, Ang-2 unexpectedly functions as a Tie2 agonist, bolstering Akt activity so as to provide negative feedback on FOXO1-regulated transcription and apoptosis. In addition, we show that Ang-2, like Ang-1, activates Tie2/Akt signaling in vivo, thereby inhibiting the expression of FOXO1 target genes. Consistent with a role for Ang-2 as a Tie2 activator, we demonstrate that Ang-2 inhibits vascular leak. Our data suggests a model in which Ang-2 expression is induced in stressed endothelial cells, where it acts as an autocrine Tie2 agonist and protective factor.


Subject(s)
Angiopoietin-2/metabolism , Autocrine Communication , Endothelial Cells/physiology , Oxidative Stress , Androstadienes/metabolism , Angiopoietin-2/genetics , Animals , Apoptosis/physiology , Cells, Cultured , Endothelial Cells/cytology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Transcriptional Activation , Wortmannin
8.
J Invest Dermatol ; 126(10): 2316-22, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16741507

ABSTRACT

Hemangioma of infancy is the most common neoplasm of childhood. While hemangiomas are classic examples of angiogenesis, the angiogenic factors responsible for hemangiomas are not fully understood. Previously, we demonstrated that malignant endothelial tumors arise in the setting of autocrine loops involving vascular endothelial growth factor (VEGF) and its major mitogenic receptor vascular endothelial growth factor receptor 2. Hemangiomas of infancy differ from malignant endothelial tumors in that they usually regress, or can be induced to regress by pharmacologic means, suggesting that angiogenesis in hemangiomas differs fundamentally from that of malignant endothelial tumors. Here, we demonstrate constitutive activation of the endothelial tie-2 receptor in human hemangioma of infancy and, using a murine model of hemangioma, bEnd.3 cells; we show that bEnd.3 hemangiomas produce both angiopoietin-2 (ang-2) and its receptor, tie-2, in vivo. We also demonstrate that inhibition of tie-2 signaling with a soluble tie-2 receptor decreases bEnd.3 hemangioma growth in vivo. The efficacy of tie-2 blockade suggests that either tie-2 activation or ang-2 may be required for in vivo growth. To address this issue, we used tie-2-deficient bEnd.3 hemangioma cells, which, surprisingly, were fully proficient in in vivo growth. Previous studies from our laboratory and others have implicated reactive oxygen-generating nox enzymes in the angiogenic switch, so we examined the effect of nox inhibitors on ang-2 production in vitro and on bEnd.3 tumor growth in vivo. We then inhibited ang-2 production pharmacologically using novel inhibitors of nox enzymes and found that this treatment nearly abolished bEnd.3 hemangioma growth in vivo. Signal-transduction blockade targeting ang-2 production may be useful in the treatment of human hemangiomas in vivo.


Subject(s)
Angiopoietin-2/antagonists & inhibitors , Hemangioma/drug therapy , Angiopoietin-2/biosynthesis , Angiopoietin-2/genetics , Animals , Gentian Violet/pharmacology , Hemangioma/etiology , Hemangioma/pathology , Humans , Mice , NADPH Oxidases/antagonists & inhibitors , Quaternary Ammonium Compounds/pharmacology , RNA, Messenger/analysis , Receptor, TIE-2/antagonists & inhibitors , Receptor, TIE-2/physiology , Signal Transduction
10.
Genes Dev ; 18(9): 1060-71, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15132996

ABSTRACT

Despite genetic evidence establishing angiopoietin-1 (Ang-1) as an essential regulator of vascular development, the molecular mechanisms underlying Ang-1 function are almost completely uncharacterized. In this report, we demonstrate that Ang-1, via Akt activation, is a potent inhibitor of the forkhead transcription factor FKHR (FOXO1), identifying for the first time a nuclear signaling pathway through which Ang-1 modulates gene expression. We use microarray analysis to show that FKHR, whose function in endothelial cells has not previously been elucidated, regulates many genes associated with vascular destabilization and remodeling (including angiopoietin-2, an Ang-1 antagonist) and endothelial cell apoptosis (e.g., survivin, TRAIL). Ang-1 inhibits FKHR-mediated changes in gene expression and FKHR-induced apoptosis. Analysis of gene expression changes induced by an activated version of Akt confirms that FKHR is a major target through which Akt regulates transcription in endothelial cells. We use RNA interference to demonstrate that FKHR is required for the expression of genes (including Ang-2) that have important vascular functions. Our data suggest a novel, tissue-specific role for the Akt/FKHR pathway in the vasculature and suggest a mechanistic basis for the previously described actions of Ang-1 as a regulator of endothelial cell survival and blood vessel stability.


Subject(s)
Angiopoietin-1/pharmacology , DNA-Binding Proteins/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Protein Serine-Threonine Kinases , Transcription Factors/physiology , Angiopoietin-1/physiology , Animals , Apoptosis/drug effects , Base Sequence , Cattle , Cells, Cultured , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , Endothelium, Vascular/cytology , Forkhead Box Protein O1 , Forkhead Transcription Factors , Gene Expression/drug effects , Gene Expression Profiling , Humans , Models, Biological , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Recombinant Proteins/pharmacology , Signal Transduction , Transcription Factors/genetics
11.
Nat Struct Biol ; 10(1): 38-44, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12469114

ABSTRACT

Angiopoietins are a recently discovered family of angiogenic factors that interact with the endothelial receptor tyrosine kinase Tie2, either as agonists (angiopoietin-1) or as context-dependent agonists/antagonists (angiopoietin-2). Here we show that angiopoietin-1 has a modular structure unlike any previously characterized growth factor. This modular structure consists of a receptor-binding domain, a dimerization motif and a superclustering motif that forms variable-sized multimers. Genetic engineering of precise multimers of the receptor-binding domain of angiopoietin-1, using surrogate multimerization motifs, reveals that tetramers are the minimal size required for activating endothelial Tie2 receptors. In contrast, engineered dimers can antagonize endothelial Tie2 receptors. Surprisingly, angiopoietin-2 has a modular structure and multimerization state similar to that of angiopoietin-1, and its antagonist activity seems to be a subtle property encoded in its receptor-binding domain.


Subject(s)
Angiopoietins/chemistry , Angiopoietins/metabolism , Receptor, TIE-2/metabolism , Amino Acid Motifs , Amino Acid Sequence , Angiopoietin-1/chemistry , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Angiopoietin-2/chemistry , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Angiopoietins/genetics , Animals , CHO Cells , Cricetinae , Cricetulus , Dimerization , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Models, Molecular , Phosphorylation , Protein Binding , Protein Engineering , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 99(17): 11393-8, 2002 Aug 20.
Article in English | MEDLINE | ID: mdl-12177445

ABSTRACT

Vascular endothelial growth factor (VEGF) plays a critical role during normal embryonic angiogenesis and also in the pathological angiogenesis that occurs in a number of diseases, including cancer. Initial attempts to block VEGF by using a humanized monoclonal antibody are beginning to show promise in human cancer patients, underscoring the importance of optimizing VEGF blockade. Previous studies have found that one of the most effective ways to block the VEGF-signaling pathway is to prevent VEGF from binding to its normal receptors by administering decoy-soluble receptors. The highest-affinity VEGF blocker described to date is a soluble decoy receptor created by fusing the first three Ig domains of VEGF receptor 1 to an Ig constant region; however, this fusion protein has very poor in vivo pharmacokinetic properties. By determining the requirements to maintain high affinity while extending in vivo half life, we were able to engineer a very potent high-affinity VEGF blocker that has markedly enhanced pharmacokinetic properties. This VEGF-Trap effectively suppresses tumor growth and vascularization in vivo, resulting in stunted and almost completely avascular tumors. VEGF-Trap-mediated blockade may be superior to that achieved by other agents, such as monoclonal antibodies targeted against the VEGF receptor.


Subject(s)
Antineoplastic Agents/pharmacology , Endothelial Growth Factors/antagonists & inhibitors , Endothelial Growth Factors/immunology , Endothelium, Vascular/physiology , Lymphokines/antagonists & inhibitors , Lymphokines/immunology , Melanoma, Experimental/drug therapy , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , 3T3 Cells , Animals , Antineoplastic Agents/therapeutic use , Bone Neoplasms/blood supply , Bone Neoplasms/drug therapy , Cell Division , Drug Design , Endothelial Growth Factors/pharmacology , Extracellular Matrix/physiology , Humans , Immunoglobulin Constant Regions/genetics , Immunoglobulin G/genetics , Lymphokines/pharmacology , Melanoma, Experimental/blood supply , Mice , Mice, Inbred BALB C , Phosphorylation , Protein Engineering , Rhabdomyosarcoma/blood supply , Rhabdomyosarcoma/drug therapy , Umbilical Veins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
13.
Am J Pathol ; 160(5): 1683-93, 2002 May.
Article in English | MEDLINE | ID: mdl-12000720

ABSTRACT

Diabetic retinopathy remains a leading cause of irreversible blindness. A critical early pathology in the disease is the adhesion of leukocytes to the retinal vasculature, a process that occurs, in part, via intercellular adhesion molecule-1. Once leukocyte adhesion occurs, endothelial cell injury ensues, as does blood-retinal barrier breakdown. Here we show that angiopoietin-1 can prevent and reverse these diabetic retinal vascular changes in both new and established diabetes. Angiopoietin-1, when given intravitreally to newly diabetic rats, normalized retinal vascular endothelial growth factor (VEGF) and intercellular adhesion molecule-1 mRNA and protein levels, leading to reductions in leukocyte adhesion, endothelial cell injury, and blood-retinal barrier breakdown. When an adenovirus coding for angiopoietin-1 was given systemically to mice with established diabetes, it similarly inhibited leukocyte adhesion and endothelial cell injury and blood-retinal barrier breakdown. These changes coincided with reductions in retinal eNOS, nitric oxide, Akt (protein kinase B), and MAP kinase activity, known mediators of VEGF bioactivity and leukocyte adhesion. When endogenous VEGF bioactivity was inhibited with a soluble Flt-1/Fc chimera, retinal Akt kinase activity was significantly reduced in vivo. Taken together, these data document new vascular and anti-inflammatory bioactivities for angiopoietin-1 and identify it as the first naturally occurring protein that directly protects the retinal vasculature in diabetes.


Subject(s)
Diabetic Retinopathy/drug therapy , Membrane Glycoproteins/therapeutic use , Protein Serine-Threonine Kinases , Angiopoietin-1 , Animals , Blood-Retinal Barrier/drug effects , Cattle , Cell Adhesion/drug effects , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Dose-Response Relationship, Drug , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Enzyme Activation/drug effects , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Lymphokines/genetics , Lymphokines/metabolism , Male , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Retina/drug effects , Retina/metabolism , Retina/pathology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...