Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Genome Biol ; 25(1): 102, 2024 Apr 19.
Article En | MEDLINE | ID: mdl-38641822

BACKGROUND: Splicing factors are vital for the regulation of RNA splicing, but some have also been implicated in regulating transcription. The underlying molecular mechanisms of their involvement in transcriptional processes remain poorly understood. RESULTS: Here, we describe a direct role of splicing factor RBM22 in coordinating multiple steps of RNA Polymerase II (RNAPII) transcription in human cells. The RBM22 protein widely occupies the RNAPII-transcribed gene locus in the nucleus. Loss of RBM22 promotes RNAPII pause release, reduces elongation velocity, and provokes transcriptional readthrough genome-wide, coupled with production of transcripts containing sequences from downstream of the gene. RBM22 preferentially binds to the hyperphosphorylated, transcriptionally engaged RNAPII and coordinates its dynamics by regulating the homeostasis of the 7SK-P-TEFb complex and the association between RNAPII and SPT5 at the chromatin level. CONCLUSIONS: Our results uncover the multifaceted role of RBM22 in orchestrating the transcriptional program of RNAPII and provide evidence implicating a splicing factor in both RNAPII elongation kinetics and termination control.


Positive Transcriptional Elongation Factor B , RNA Polymerase II , Humans , Chromatin , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , RNA Polymerase II/metabolism , RNA Splicing , RNA Splicing Factors/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism
2.
FASEB J ; 34(12): 16662-16675, 2020 12.
Article En | MEDLINE | ID: mdl-33124722

We previously showed that calnexin (Canx)-deficient mice are desensitized to experimental autoimmune encephalomyelitis (EAE) induction, a model that is frequently used to study inflammatory demyelinating diseases, due to increased resistance of the blood-brain barrier to immune cell transmigration. We also discovered that Fabp5, an abundant cytoplasmic lipid-binding protein found in brain endothelial cells, makes protein-protein contact with the cytoplasmic C-tail domain of Canx. Remarkably, both Canx-deficient and Fabp5-deficient mice commonly manifest resistance to EAE induction. Here, we evaluated the importance of Fabp5/Canx interactions on EAE pathogenesis and on the patency of a model blood-brain barrier to T-cell transcellular migration. The results demonstrate that formation of a complex comprised of Fabp5 and the C-tail domain of Canx dictates the permeability of the model blood-brain barrier to immune cells and is also a prerequisite for EAE pathogenesis.


Calnexin/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Fatty Acid-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Line , Cell Movement/physiology , Disease Models, Animal , Endothelial Cells/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Permeability
3.
Sci Rep ; 10(1): 18115, 2020 10 22.
Article En | MEDLINE | ID: mdl-33093545

Calsequestrin is among the most abundant proteins in muscle sarcoplasmic reticulum and displays a high capacity but a low affinity for Ca2+ binding. In mammals, calsequestrin is encoded by two genes, CASQ1 and CASQ2, which are expressed almost exclusively in skeletal and cardiac muscles, respectively. Phylogenetic analysis indicates that calsequestrin is an ancient gene in metazoans, and that the duplication of the ancestral calsequestrin gene took place after the emergence of the lancelet. CASQ2 gene variants associated with catecholaminergic polymorphic ventricular tachycardia (CPVT) in humans are positively correlated with a high degree of evolutionary conservation across all calsequestrin homologues. The mutations are distributed in diverse locations of the calsequestrin protein and impart functional diversity but remarkably manifest in a similar phenotype in humans.


Calcium/metabolism , Calsequestrin/genetics , Calsequestrin/metabolism , Heart Diseases/pathology , Mutation , Phylogeny , Amino Acid Sequence , Animals , Calcium Signaling , Calsequestrin/chemistry , Heart Diseases/genetics , Heart Diseases/metabolism , Humans , Phenotype , Protein Conformation , Sequence Homology, Amino Acid
4.
Mol Cancer ; 19(1): 118, 2020 07 29.
Article En | MEDLINE | ID: mdl-32727463

BACKGROUND: Pancreatic cancer is one of the most lethal malignancies and has an extremely poor diagnosis and prognosis. The development of resistance to gemcitabine is still a major challenge. The long noncoding RNA PVT1 was reported to be involved in carcinogenesis and chemoresistance; however, the mechanism by which PVT1 regulates the sensitivity of pancreatic cancer to gemcitabine remains poorly understood. METHODS: The viability of pancreatic cancer cells was assessed by MTT assay in vitro and xenograft tumor formation assay in vivo. The expression levels of PVT1 and miR-619-5p were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Western blotting analysis and qRT-PCR were performed to assess the protein and mRNA levels of Pygo2 and ATG14, respectively. Autophagy was explored via autophagic flux detection under confocal microscopy and autophagic vacuole investigation under transmission electron microscopy (TEM). The functional role and mechanism of PVT1 were further investigated by gain- and loss-of-function assays in vitro. RESULTS: In the present study, we demonstrated that PVT1 was up-regulated in gemcitabine-resistant pancreatic cancer cell lines. Gain- and loss-of-function assays revealed that PVT1 impaired sensitivity to gemcitabine in vitro and in vivo. We further found that PVT1 up-regulated the expression of both Pygo2 and ATG14 and thus regulated Wnt/ß-catenin signaling and autophagic activity to overcome gemcitabine resistance through sponging miR-619-5p. Moreover, we discovered three TCF/LEF binding elements (TBEs) in the promoter region of PVT1, and activation of Wnt/ß-catenin signaling mediated by the up-regulation of Pygo2 increased PVT1 expression by direct binding to the TBE region. Furthermore, PVT1 was discovered to interact with ATG14, thus promoting assembly of the autophagy specific complex I (PtdIns3K-C1) and ATG14-dependent class III PtdIns3K activity. CONCLUSIONS: These data indicate that PVT1 plays a critical role in the sensitivity of pancreatic cancer to gemcitabine and highlight its potential as a valuable target for pancreatic cancer therapy.


Adaptor Proteins, Vesicular Transport/genetics , Autophagy-Related Proteins/genetics , Autophagy/genetics , Drug Resistance, Neoplasm/genetics , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , RNA, Long Noncoding/genetics , Wnt Signaling Pathway , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Humans , Mice , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Binding , RNA Interference , Xenograft Model Antitumor Assays , Gemcitabine
5.
Commun Biol ; 3(1): 94, 2020 03 03.
Article En | MEDLINE | ID: mdl-32127635

Early embryogenesis relies on maternally inherited mRNAs. Although the mechanism of maternal mRNA degradation during maternal-to-zygotic transition (MZT) has been extensively studied in vertebrates, how the embryos maintain maternal mRNA stability remains unclear. Here, we identify Igf2bp3 as an important regulator of maternal mRNA stability in zebrafish. Depletion of maternal igf2bp3 destabilizes maternal mRNAs prior to MZT and leads to severe developmental defects, including abnormal cytoskeleton organization and cell division. However, the process of oogenesis and the expression levels of maternal mRNAs in unfertilized eggs are normal in maternal igf2bp3 mutants. Gene ontology analysis revealed that these functions are largely mediated by Igf2bp3-bound mRNAs. Indeed, Igf2bp3 depletion destabilizes while its overexpression enhances its targeting maternal mRNAs. Interestingly, igf2bp3 overexpression in wild-type embryos also causes a developmental delay. Altogether, these findings highlight an important function of Igf2bp3 in controlling early zebrafish embryogenesis by binding and regulating the stability of maternal mRNAs.


Embryonic Development/genetics , RNA, Messenger, Stored/metabolism , RNA-Binding Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , RNA Stability/genetics , RNA, Messenger, Stored/genetics , RNA-Binding Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zygote/metabolism
6.
FASEB J ; 33(8): 8892-8904, 2019 08.
Article En | MEDLINE | ID: mdl-31051095

The endoplasmic reticulum (ER) plays a central role in cellular stress responses via mobilization of ER stress coping responses, such as the unfolded protein response (UPR). The inositol-requiring 1α (IRE1α) is an ER stress sensor and component of the UPR. Muscle cells also have a well-developed and highly subspecialized membrane network of smooth ER called the sarcoplasmic reticulum (SR) surrounding myofibrils and specialized for Ca2+ storage, release, and uptake to control muscle excitation-contraction coupling. Here, we describe 2 distinct pools of IRE1α in cardiac and skeletal muscle cells, one localized at the perinuclear ER and the other at the junctional SR. We discovered that, at the junctional SR, calsequestrin binds to the ER luminal domain of IRE1α, inhibiting its dimerization. This novel interaction of IRE1α with calsequestrin, one of the highly abundant Ca2+ handling proteins at the junctional SR, provides new insights into the regulation of stress coping responses in muscle cells.-Wang, Q., Groenendyk, J., Paskevicius, T., Qin, W., Kor, K. C., Liu, Y., Hiess, F., Knollmann, B. C., Chen, S. R. W., Tang, J., Chen, X.-Z., Agellon, L. B., Michalak, M. Two pools of IRE1α in cardiac and skeletal muscle cells.


Endoribonucleases/metabolism , Muscle Fibers, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Binding Sites , COS Cells , Calcium Signaling , Calsequestrin/metabolism , Cells, Cultured , Chlorocebus aethiops , Endoribonucleases/chemistry , Mice , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Rabbits , Sarcoplasmic Reticulum/metabolism
7.
Oncotarget ; 8(17): 27915-27928, 2017 Apr 25.
Article En | MEDLINE | ID: mdl-28427190

Anti-microtubule drugs, such as paclitaxel (PTX), are extensively used for the treatment of numerous cancers. However, growing evidence has shown that PTX resistance, either intrinsic or acquired, frequently occurs in patients and results in the failure of treatment, contributing to the high cancer mortality rate. Therefore, it is necessary to identify the genes or pathways involved in anti-microtubule drug resistance for future successful treatment of cancers. Pygopus2 (Pygo2), which contains a Zn-coordinated plant homeodomain (PHD) finger domain, is critical for ß-catenin-dependent transcriptional switches in normal and malignant tissues and is over-expressed in various cancers, including human brain glioma. In this study, we report that over-expression of Pygo2 inhibited the efficacy of PTX and contributed to cell multidrug resistance in two different ways. First, over-expression of Pygo2 inhibited the PTX-induced phosphorylation of B-cell lymphoma 2 (Bcl-2), suppressing the proteolytic cleavage of procaspase-8/9 and further inhibiting the activation of caspase-3, which also inhibits the activation of the JNK/SAPK pathway, ultimately inhibiting cell apoptosis. Second, over-expression of Pygo2 facilitated the expression of P-glycoprotein, which acts as a drug efflux pump, by promoting the transcription of Multi-drug resistance 1 (MDR1) at the MDR1 promoter loci, resulting in acceleration of the efflux of PTX.


Antineoplastic Agents, Phytogenic/pharmacology , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioma/genetics , Intracellular Signaling Peptides and Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Glioma/drug therapy , Glioma/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/genetics , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , beta Catenin/metabolism
8.
Oncotarget ; 7(34): 54488-54502, 2016 Aug 23.
Article En | MEDLINE | ID: mdl-27359054

Breast cancer is a heterogeneous disease associated with diverse clinical, biological and molecular features, presenting huge challenges for prognosis and treatment. Here we found that perilipin-1 (PLIN1) mRNA expression is significantly downregulated in human breast cancer. Kaplan-Meier analysis indicated that patients presenting with reduced PLIN1 expression exhibited poorer overall metastatic relapse-free survival (p = 0.03). Further Cox proportional hazard models analysis revealed that the reduced expression of PLIN1 is an independent predictor of overall survival in estrogen receptor positive (p < 0.0001, HR = 0.87, 95% CI = 0.81-0.92, N = 3,600) and luminal A-subtype (p = 0.02, HR = 0.88, 95% CI = 0.78-0.98, N = 1,469) breast cancer patients. We also demonstrated that the exogenous expression of PLIN1 in human breast cancer MCF-7 and MDA-MB-231 cells significantly inhibits cell proliferation, migration, invasion and in vivo tumorigenesis in mice. Together, these data provide novel insights into a prognostic significance of PLIN1 in human breast cancer and reveal a potentially new gene therapy target for breast cancer.


Breast Neoplasms/mortality , Perilipin-1/analysis , Animals , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Genes, p53 , Humans , Mice , Mutation , Neoplasm Invasiveness , Prognosis , Proportional Hazards Models , Receptors, Estrogen/analysis
9.
Article En | MEDLINE | ID: mdl-22691797

Successfully forming ligand-protein complexes with specific compounds can be a significant challenge in supporting structure-based drug design for a given protein target. In this respect, an on-column ligand- and detergent-exchange method was developed to obtain ligand-protein complexes of an adamantane series of compounds with 11ß-hydroxysteroid dehydrogenase type 1 (11ß-HSD1) after a variety of other complexation methods had failed. This report describes the on-column exchange method and an unexpected byproduct of the method in which artificial trimers were observed in the structures.


Crystallography, X-Ray/methods , Drug Design , Enzyme Inhibitors/chemistry , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , 11-beta-Hydroxysteroid Dehydrogenase Type 1/chemistry , Crystallography, X-Ray/instrumentation , Humans , Ligands , Models, Molecular , Protein Structure, Quaternary
10.
Bioorg Med Chem Lett ; 17(3): 750-5, 2007 Feb 01.
Article En | MEDLINE | ID: mdl-17110106

A novel class of adamantane ethers 11beta-hydroxysteroid hydrogenase type I inhibitors has been discovered. These compounds have excellent HSD-1 potency and selectivity against HSD-2. The structure-activity relationships, selectivity, metabolism, PK, ex vivo pharmacodynamic data, and an X-ray crystal structure of one of these inhibitors bound to h-HSD-1 are discussed.


11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Adamantane/analogs & derivatives , Adamantane/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , Adamantane/chemical synthesis , Alkylation , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Ethers/chemical synthesis , Ethers/pharmacology , Half-Life , Humans , Indicators and Reagents , Mice , Mice, Knockout , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Models, Molecular
12.
Anticancer Res ; 24(6): 3907-10, 2004.
Article En | MEDLINE | ID: mdl-15736430

Chk1 (checkpoint kinase 1) is a serine-threonine kinase that is critical for G2/M arrest in response to DNA damage. Chk1 phosphorylates Cdc25C at serine-216, a major regulatory site, in response to DNA damage. Furthermore, Chk1 also phosphorylates Cdc25A on serine 123 which accelerates its degradation through the ubiquitin-proteasome pathway and arrests cells in late G2-phase after DNA damage. In the present study, we demonstrated that Chk1 phosphorylates pro-apoptotic protein BAD (Bcl-2/Bcl-XL-Antagonist, causing cell Death) in vitro. In vitro phosphorylation analysis with various mouse BAD peptides has revealed two phosphorylation sites for Chk1 at serine-155 and serine-170. When wild-type and mutant BAD (S155A) constructs were transfected into 293T cells, an association between BAD and Chk1 was observed by co-immunoprecipitation. In addition, there was an increase in the phosphorylation of serine-155 following DNA damage by adriamycin treatment. Our results suggest that Chk1 associates with BAD and phosphorylates the BAD protein at serine-155. Taken together, our results suggest that Chk1 may inactivate BAD by associating with and phosphorylating residues critical for BAD function in response to DNA damage.


Carrier Proteins/metabolism , Protein Kinases/metabolism , Amino Acid Sequence , Cell Line , Checkpoint Kinase 1 , Embryo, Mammalian , Humans , Kidney/cytology , Molecular Sequence Data , Polyisoprenyl Phosphates , Protein Binding , bcl-Associated Death Protein
13.
Bioorg Med Chem ; 10(8): 2803-6, 2002 Aug.
Article En | MEDLINE | ID: mdl-12057670

The crystal structure of ABT-378 (lopinavir), bound to the active site of HIV-1 protease is described. A comparison with crystal structures of ritonavir, A-78791, and BILA-2450 shows some analogous features with previous reported compounds. A cyclic urea unit in the P(2) position of ABT-378 is novel and makes two bidentate hydrogen bonds with Asp 29 of HIV-1 protease. In addition, a previously unreported shift in the Gly 48 carbonyl position is observed. A discussion of the structural features responsible for its high potency against wild-type HIV protease is given along with an analysis of the effect of active site mutations on potency in in vitro assays.


HIV Protease Inhibitors/chemistry , HIV Protease/chemistry , Pyrimidinones/chemistry , Binding Sites/genetics , Crystallization , Crystallography, X-Ray , Humans , Hydrogen Bonding , Lopinavir , Molecular Structure , Mutation , Protein Binding
14.
Am J Pathol ; 160(5): 1683-93, 2002 May.
Article En | MEDLINE | ID: mdl-12000720

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.


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
15.
J Clin Invest ; 109(6): 805-15, 2002 Mar.
Article En | MEDLINE | ID: mdl-11901189

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.


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
16.
Am J Pathol ; 160(2): 501-9, 2002 Feb.
Article En | MEDLINE | ID: mdl-11839570

Leukocyte adhesion to the diabetic retinal vasculature results in early blood-retinal barrier breakdown, capillary nonperfusion, and endothelial cell injury and death. Previous work has shown that intercellular adhesion molecule-1 (ICAM-1) and CD18 are required for these processes. However the relevant in vivo stimuli for ICAM-1 and CD18 expression in diabetes remain unknown. The current study investigated the causal role of endogenous vascular endothelial growth factor (VEGF) and nitric oxide in initiating these events. Diabetes was induced in Long-Evans rats with streptozotocin, resulting in a two- to threefold increase in retinal leukocyte adhesion. Confirmed diabetic animals were treated with a highly specific VEGF-neutralizing Flt-Fc construct (VEGF TrapA(40)). Retinal ICAM-1 mRNA levels in VEGF TrapA(40)-treated diabetic animals were reduced by 83.5% compared to diabetic controls (n = 5, P < 0.0001). VEGF TrapA(40) also potently suppressed diabetic leukocyte adhesion in retinal arterioles (47%, n = 11, P < 0.0001), venules (36%, n = 11, P < 0.0005), and capillaries (36%, n = 11, P < 0.001). The expression of endothelial nitric oxide synthase (eNOS), a downstream mediator of VEGF activity, was increased in diabetic retina, and was potently suppressed with VEGF TrapA(40) treatment (n = 8, P < 0.005). Further, VEGF TrapA(40) reduced the diabetes-related nitric oxide increases in the retinae of diabetic animals. The inhibition of eNOS with N-omega-nitro-L-arginine methyl ester also potently reduced retinal leukocyte adhesion. Although neutrophil CD11a, CD11b, and CD18 levels were increased in 1-week diabetic animals, VEGF TrapA(40) did not alter the expression of these integrin adhesion molecules. Taken together, these data demonstrate that VEGF induces retinal ICAM-1 and eNOS expression and initiates early diabetic retinal leukocyte adhesion in vivo. The inhibition of VEGF bioactivity may prove useful in the treatment of the early diabetic retinopathy.


Cell Adhesion , Diabetic Retinopathy/physiopathology , Endothelial Growth Factors/metabolism , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/physiology , Lymphokines/metabolism , Nitric Oxide Synthase/metabolism , Retina/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Retinopathy/metabolism , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Integrins/metabolism , Intercellular Adhesion Molecule-1/genetics , Leukocytes/metabolism , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type III , Nitrites/metabolism , Rats , Rats, Long-Evans , Retina/enzymology , Retinal Vessels/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
...