Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Biomedicines ; 11(1)2022 Dec 23.
Article in English | MEDLINE | ID: mdl-36672545

ABSTRACT

We asked whether hyperoxia might induce hypomyelination of the corpus callosum, clinically described as periventricular leukomalacia (PVL) of the severely preterm infant. Mouse pups and their nursing dams were placed in 80% oxygen from P4-P8, then removed to room air until P11. Corpus callosal sections were probed myelin immunofluorescence, tested for myelin basic protein concentration by Western blot, and both glial fibrillary acidic protein levels and apoptosis quantified. Density of corpus callosal capillaries were measured after lectin staining and hypoxia measured by Hypoxyprobe. Numbers of oligodendrocytes were quantified by immunohistochemistry. We next used hypoxiamimesis as a surrogate to hypoxia by comparing cerebral hypoxia inducible factor (HIF) stabilization to hepatic HIF stabilization. Hyperoxia induced hypomyelination and a reduction of corpus callosal capillaries. Hyperoxia decreased numbers of oligodendrocytes with an increase in corpus callosal fibrosis and apoptosis. Cerebral hypoxiamimesis induced hypomyelination whereas hepatic hypoxiamimesis alone increased myelination, oligodendrocyte numbers, and corpus callosal capillary density. Hepatic HIF-1 dependence on myelination was confirmed using the cre/lox hepatic HIF-1 knockout. These findings suggest that hyperoxia can induce hypomyelination through vasoobliteration and subsequent ischemia, adding a potential oxygen induced mechanism to the diverse causes of periventricular leukomalacia of the severely preterm infant. Targeting hepatic HIF-1 alone led to increased myelination.

2.
Front Cell Dev Biol ; 8: 488, 2020.
Article in English | MEDLINE | ID: mdl-32656210

ABSTRACT

Here we rank order small molecule inhibitors of hypoxia inducible factor (HIF) prolyl hydroxylases (PHDs) using severity of oxygen induced retinopathy (OIR) as an outcome measure. Dose response analyses in cell cultures of hepatoma (Hep3B), retinal Müller cells (MIO-M1) and primary retinal endothelial cells were conducted to evaluate potency by comparing dose to HIF-1,2 protein levels by western blotting. In vivo dose response was determined using the luciferase-transgene HIF reporter (luc-ODD). Each compound was placed in rank order by their ability to reduce neovascularization and capillary drop out in the OIR mouse model. An Epas1 KO confined to retinal Müller cells was used to determine whether successful protection by HIF stabilization requires HIF-2. Two candidate small molecules can prevent OIR by stabilizing HIF-1 to prevent oxygen induced growth attenuation and vascular obliteration. Müller cell HIF-2, the mediator of pathologic retinal angiogenesis, is not required for protection. The lack of dependence on Müller cell HIF-2 predicts that inhibition of HIF PHD will not drive pathological angiogenesis.

3.
Nat Commun ; 11(1): 1277, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32152301

ABSTRACT

Although supplemental oxygen is required to promote survival of severely premature infants, hyperoxia is simultaneously harmful to premature developing tissues such as in the retina. Here we report the effect of hyperoxia on central carbon metabolism in primary mouse Müller glial cells and a human Müller glia cell line (M10-M1 cells). We found decreased flux from glycolysis entering the tricarboxylic acid cycle in Müller cells accompanied by increased glutamine consumption in response to hyperoxia. In hyperoxia, anaplerotic catabolism of glutamine by Müller cells increased ammonium release two-fold. Hyperoxia induces glutamine-fueled anaplerosis that reverses basal Müller cell metabolism from production to consumption of glutamine.


Subject(s)
Ependymoglial Cells/metabolism , Glutamine/metabolism , Hyperoxia/metabolism , Animals , Astrocytes/metabolism , Carbon Isotopes , Cells, Cultured , Endothelial Cells/metabolism , Glucose/metabolism , Glutaminase/metabolism , Glycolysis , Humans , Metabolome , Mice , Mitochondria/metabolism , Models, Biological , Oxidation-Reduction , Phosphorylation , Pyruvate Dehydrogenase Complex/metabolism
4.
JCI Insight ; 4(14)2019 07 25.
Article in English | MEDLINE | ID: mdl-31341109

ABSTRACT

We determined which metabolic pathways are activated by hypoxia-inducible factor 1-mediated (HIF-1-mediated) protection against oxygen-induced retinopathy (OIR) in newborn mice, the experimental correlate to retinopathy of prematurity, a leading cause of infant blindness. HIF-1 coordinates the change from oxidative to glycolytic metabolism and mediates flux through serine and 1-carbon metabolism (1CM) in hypoxic and cancer cells. We used untargeted metabolite profiling in vivo to demonstrate that hypoxia mimesis activates serine/1CM. Both [13C6] glucose labeling of metabolites in ex vivo retinal explants as well as in vivo [13C3] serine labeling of metabolites followed in liver lysates strongly suggest that retinal serine is primarily derived from hepatic glycolytic carbon and not from retinal glycolytic carbon in newborn pups. In HIF-1α2lox/2lox albumin-Cre-knockout mice, reduced or near-0 levels of serine/glycine further demonstrate the hepatic origin of retinal serine. Furthermore, inhibition of 1CM by methotrexate blocked HIF-mediated protection against OIR. This demonstrated that 1CM participates in protection induced by HIF-1 stabilization. The urea cycle also dominated pathway enrichment analyses of plasma samples. The dependence of retinal serine on hepatic HIF-1 and the upregulation of the urea cycle emphasize the importance of the liver to remote protection of the retina.


Subject(s)
Carbon/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Retina/pathology , Retinopathy of Prematurity/pathology , Serine/metabolism , Animals , Disease Models, Animal , Glycine/administration & dosage , Glycine/analogs & derivatives , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Isoquinolines/administration & dosage , Liver/metabolism , Metabolic Networks and Pathways/drug effects , Methotrexate/administration & dosage , Mice , Mice, Knockout , Oxygen/toxicity , Protein Stability/drug effects , Retinopathy of Prematurity/etiology , Tissue Culture Techniques , Up-Regulation
5.
JAMA Ophthalmol ; 137(4): 417-423, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30763441

ABSTRACT

Importance: The Surfactant, Positive Pressure, and Pulse Oximetry Randomized Trial (SUPPORT) demonstrated that static low oxygen saturation decreased retinopathy of prematurity (ROP) but increased mortality compared with static high oxygen saturation cohorts. Objective: To compare outcomes of a biphasic oxygen protocol with static targets recommended by SUPPORT. Design, Setting, and Participants: Retrospective cohort study comparing biphasic vs static standards 41 months prior to and 42 months after a change from biphasic to static SUPPORT standards at a level III neonatal intensive care unit (Fairview Hospital, Cleveland, Ohio). The study included infants born at a corrected gestational age (CGA) of 31 weeks or younger or birth weight 1500 g or less. Data were analyzed between August 2010 and July 2017. Interventions: The pre-SUPPORT group underwent biphasic protocol target saturations of 85% to 92% at younger than 34 weeks' CGA and greater than 95% at 34 weeks' CGA or older. The post-SUPPORT group underwent a constant 91% to 95% target. Main Outcomes and Measures: Primary outcome was incidence of type 1 ROP. Secondary outcomes were incidence of any ROP, time to full vascularization, and mortality. Results: Of 596 eligible infants, 562 were included in ophthalmic analysis. Three hundred three patients were boys (54%); 399 were white (71%), 87 were black (15%), and 76 were of other or unknown race/ethnicity (14%). Mean (SD) CGA and birth weight were 29 (2) weeks and 1151 (346) g, respectively. Any ROP overall increased (53 [20%] pre-SUPPORT vs n = 86 [28%] post-SUPPORT; absolute difference, 8%; 95% CI, 1%-15%; odds ratio, 1.6; 95% CI, 1.05-2.3; P = .03). Type 1 ROP increased in the post-SUPPORT era (n = 6 [2%] pre-SUPPORT vs n = 18 [6%] post-SUPPORT; absolute difference, 4%; 95% CI, 0.4%-7%; odds ratio, 2.7; 95% CI, 1.05-6.9; P = .03). There was a delay in vascularization in the post-SUPPORT group (n = 6 [2%] pre-SUPPORT vs n = 18 [6%] post-SUPPORT; absolute difference, 4%; 95% CI, 0.4%-7%; P = .03). Conclusions and Relevance: Compared with static oxygen standards, biphasic oxygen targets are associated with decreased incidence and severity of ROP without increasing mortality.


Subject(s)
Oxygen Inhalation Therapy/methods , Retinopathy of Prematurity , Female , Hospital Mortality , Humans , Incidence , Infant, Newborn , Infant, Very Low Birth Weight , Male , Odds Ratio , Oxygen Consumption/physiology , Retinopathy of Prematurity/epidemiology , Retinopathy of Prematurity/physiopathology , Retinopathy of Prematurity/therapy , Retrospective Studies
6.
Invest Ophthalmol Vis Sci ; 59(8): 3440-3448, 2018 07 02.
Article in English | MEDLINE | ID: mdl-30025089

ABSTRACT

Purpose: Transcriptional analysis of retina protected by hypoxia-inducible factor (HIF) stabilization demonstrates an increase in genes associated with aerobic glycolysis. We hypothesized that since protection is associated with a change in metabolism, oxygen-induced metabolites might transduce oxygen toxicity. We used global metabolic profiling to identify retinal metabolites increased in hyperoxia compared to normoxia. Methods: Untargeted gas chromatography mass spectroscopy (GC-MS) was performed on both mouse retina samples collected in hyperoxia and on primary human retinal endothelial cells, each with and without HIF stabilization. After identifying 3-hydropxypyruvate (3OH-pyruvate) as a unique hyperoxic metabolite, endothelial cells in culture and choroidal explants were challenged with 3OH-pyruvate in order to determine how this glycolytic intermediate was metabolized, and whether it had an effect on angiogenesis. Results: 3OH-pyruvate was one of five metabolites at least 2.0-fold elevated in hyperoxia with a P value < 0.1. Once metabolized by endothelial cells, 3OH-pyruvate led to a 20-fold increase in 3-phosphoglycerate and a 4-fold increase in serine when cells were treated with Roxadustat to induce HIF stabilization. 3OH-pyruvate, but not pyruvate, destabilized HIF in endothelial cells with an increase in proline hydroxylation. 3OH-pyruvate was angiostatic in choroidal explant assays. Conclusions: 3OH-pyruvate is a unique metabolite induced by hyperoxia that destabilizes HIF at least in part by a canonical pathway. 3OH-pyruvate induces angiostasis in vitro. HIF stabilization increases serine biosynthesis in vitro and in vivo.


Subject(s)
Angiogenesis Inhibitors/metabolism , Endothelial Cells/metabolism , Hyperoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pyruvates/metabolism , Retinopathy of Prematurity/metabolism , Animals , Blotting, Western , Cells, Cultured , Gas Chromatography-Mass Spectrometry , Gene Expression Regulation/physiology , Humans , Ketoglutaric Acids/metabolism , Mice , Mice, Inbred C57BL , Oxygen/toxicity , Real-Time Polymerase Chain Reaction , Retinal Vessels/cytology , Transaminases/metabolism
7.
Proc Natl Acad Sci U S A ; 113(18): E2516-25, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27091985

ABSTRACT

Retinopathy of prematurity (ROP) causes 100,000 new cases of childhood blindness each year. ROP is initiated by oxygen supplementation necessary to prevent neonatal death. We used organ systems pharmacology to define the transcriptomes of mice that were cured of oxygen-induced retinopathy (OIR, ROP model) by hypoxia-inducible factor (HIF) stabilization via HIF prolyl hydroxylase inhibition using the isoquinolone Roxadustat or the 2-oxoglutarate analog dimethyloxalylglycine (DMOG). Although both molecules conferred a protective phenotype, gene expression analysis by RNA sequencing found that Roxadustat can prevent OIR by two pathways: direct retinal HIF stabilization and induction of aerobic glycolysis or indirect hepatic HIF-1 stabilization and increased serum angiokines. As predicted by pathway analysis, Roxadustat rescued the hepatic HIF-1 knockout mouse from retinal oxygen toxicity, whereas DMOG could not. The simplicity of systemic treatment that targets both the liver and the eye provides a rationale for protecting the severely premature infant from oxygen toxicity.


Subject(s)
Glycine/analogs & derivatives , Hypoxia-Inducible Factor 1/metabolism , Isoquinolines/administration & dosage , Liver/metabolism , Retina/metabolism , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/prevention & control , Transcriptome/drug effects , Animals , Dose-Response Relationship, Drug , Glycine/administration & dosage , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Liver/drug effects , Mice , Mice, Inbred C57BL , Retina/drug effects , Treatment Outcome
8.
Arterioscler Thromb Vasc Biol ; 35(9): 1975-86, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26139464

ABSTRACT

OBJECTIVE: Thrombospondin-4 (TSP-4) is 1 of the 5 members of the thrombospondin protein family. TSP-1 and TSP-2 are potent antiangiogenic proteins. However, angiogenic properties of the 3 other TSPs, which do not contain the domains associated with the antiangiogeneic activity of TSP-1 and TSP-2, have not been explored. In our previous studies, we found that TSP-4 is expressed in the vascular matrix of blood vessels of various sizes and is especially abundant in capillaries. We sought to identify the function of TSP-4 in the regulation of angiogenesis. APPROACH AND RESULTS: The effect of TSP-4 in in vivo angiogenesis models and its effect on angiogenesis-related properties in cultured cells were assessed using Thbs4(-/-) mice, endothelial cells (EC) derived from these mice, and recombinant TSP-4. Angiogenesis was decreased in Thbs4(-/-) mice compared with wild-type mice. TSP-4 was detected in the lumen of the growing blood vessels. Mice expressing the P387 TSP-4 variant, which was previously associated with coronary artery disease and found to be more active in its cellular interactions, displayed greater angiogenesis compared with A387 form. Lung EC from Thbs4(-/-) mice exhibited decreased adhesion, migration, and proliferation capacities compared with EC from wild-type mice. Recombinant TSP-4 promoted proliferation and the migration of EC. Integrin α2 and gabapentin receptor α2δ-1 were identified as receptors involved in regulation of EC adhesion, migration, and proliferation by TSP-4. CONCLUSION: TSP-4, an extracellular matrix protein previously associated with tissue remodeling, is now demonstrated to possess proangiogenic activity.


Subject(s)
Apoptosis , DNA/genetics , Neovascularization, Pathologic/genetics , Thrombospondins/genetics , Animals , Cell Adhesion , Cells, Cultured , DNA Mutational Analysis , Disease Models, Animal , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Thrombospondins/metabolism
9.
Am J Pathol ; 184(6): 1890-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24731446

ABSTRACT

Activation of hypoxia-inducible factor (HIF) can prevent oxygen-induced retinopathy in rodents. Here we demonstrate that dimethyloxaloylglycine (DMOG)-induced retinovascular protection is dependent on hepatic HIF-1 because mice deficient in liver-specific HIF-1α experience hyperoxia-induced damage even with DMOG treatment, whereas DMOG-treated wild-type mice have 50% less avascular retina (P < 0.0001). Hepatic HIF stabilization protects retinal function because DMOG normalizes the b-wave on electroretinography in wild-type mice. The localization of DMOG action to the liver is further supported by evidence that i) mRNA and protein erythropoietin levels within liver and serum increased in DMOG-treated wild-type animals but are reduced by 60% in liver-specific HIF-1α knockout mice treated with DMOG, ii) triple-positive (Sca1/cKit/VEGFR2), bone-marrow-derived endothelial precursor cells increased twofold in DMOG-treated wild-type mice (P < 0.001) but are unchanged in hepatic HIF-1α knockout mice in response to DMOG, and iii) hepatic luminescence in the luciferase oxygen-dependent degradation domain mouse was induced by subcutaneous and intraperitoneal DMOG. These findings uncover a novel endocrine mechanism for retinovascular protection. Activating HIF in visceral organs such as the liver may be a simple strategy to protect capillary beds in the retina and in other peripheral tissues.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver/metabolism , Oxygen/toxicity , Retinal Diseases/metabolism , Amino Acids, Dicarboxylic/pharmacology , Animals , Erythropoietin/genetics , Erythropoietin/metabolism , Hyperoxia/drug therapy , Hyperoxia/genetics , Hyperoxia/metabolism , Hyperoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Liver/pathology , Mice , Mice, Knockout , Retinal Diseases/chemically induced , Retinal Diseases/drug therapy , Retinal Diseases/genetics , Retinal Diseases/pathology
10.
Invest Ophthalmol Vis Sci ; 54(7): 4919-26, 2013 Jul 22.
Article in English | MEDLINE | ID: mdl-23761085

ABSTRACT

PURPOSE: To study the effect of systemic hypoxia-inducible factor prolyl hydroxylase inhibition (HIF PHDi) in the rat 50/10 oxygen-induced retinopathy (OIR) model. METHODS: Oxygen-induced retinopathy was created with the rat 50/10 OIR model. OIR animals received intraperitoneal injections of dimethyloxalylglycine (DMOG, 200 µg/g), an antagonist of α-ketoglutarate cofactor and inhibitor for HIF PHD, on postnatal day (P)3, P5, and P7. Control animals received intraperitoneal injections of PBS. On P14 and P21, animals were humanely killed and the effect on vascular obliteration, tortuosity, and neovascularization quantified. To analyze HIF and erythropoietin, rats at P5 were injected with DMOG (200 µg/g). Western blot or ELISA measured the levels of HIF-1 and Epo protein. Epo mRNA was measured by quantitative PCR. RESULTS: Alternating hyperoxia and hypoxia in untreated rats led to peripheral vascular obliteration on day P14 and P21. Rats that were treated with systemic DMOG by intraperitoneal injections had 3 times less ischemia and greater peripheral vascularity (P = 0.001) than control animals treated with PBS injections. Neovascularization similarly decreased by a factor of 3 (P = 0.0002). Intraperitoneal DMOG administration increased the levels of HIF and Epo in the liver and brain. Serum Epo also increased 6-fold (P = 0.0016). Systemic DMOG had no adverse effect on growth of rats treated with oxygen. CONCLUSIONS: One of the many controversies in the study of retinopathy of prematurity is whether hyperoxia or alternating hyperoxia and hypoxia creates the disease phenotype in humans. We have previously demonstrated that PHDi prevents OIR in mice exposed to 5 days of sustained 75% oxygen followed by 5 days of 21% oxygen. The 50/10 rat experiments demonstrate that PHDi is also effective in a 24-hour alternating hyperoxia-hypoxia model. The rat OIR model further validates the therapeutic value of HIF PHDi to prevent retinopathy of prematurity because it reduces oxygen-induced vascular obliteration and retinovascular growth attenuation in prolonged and/or alternating hyperoxia.


Subject(s)
Hyperoxia/enzymology , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Retinal Diseases/prevention & control , Amino Acids, Dicarboxylic , Animals , Animals, Newborn , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Erythropoietin/metabolism , Hypoxia-Inducible Factor 1/metabolism , Injections, Intraperitoneal , Neovascularization, Pathologic/drug therapy , Oxygen/pharmacology , Rats , Retinal Diseases/chemically induced , Retinal Diseases/drug therapy , Retinal Vessels/drug effects
11.
Trans Am Ophthalmol Soc ; 111: 169-79, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24385673

ABSTRACT

PURPOSE: To discover novel small molecules that inhibit hypoxia-inducible factor (HIF) prolyl hydroxylase (PHD), a key enzyme that regulates the posttranslational stability and hence activity of HIF. METHODS: NIH3T3 cell line stably transfected with firefly luciferase under a HIF-1-inducible promoter was used to screen a Chembridge library of 34,000 small molecules of molecular weight 250 to 550 Da. Positive hits were considered at 4.5-fold higher luminescence than control. Selected compounds were validated in vitro. The most effective dose was then used to treat mice expressing firefly luciferase fused to the oxygen-dependent degradation domain (lucODD) in order to determine the location of the receptor for systemic treatment with small-molecule HIF PHD inhibitors. RESULTS: Twenty-three novel small molecules were discovered, the majority of which were hydrazones and hydrazines. Of the 23 compounds, each had different selectivity for expression of erythropoietin or vascular endothelial growth factor, two angiogenic, HIF-regulated gene products. In addition, each showed different selectivity for hepatocytes or kidney, or both or neither, when injected intraperitoneally in an in vivo reporter gene assay. CONCLUSION: The discovery of multiple small molecules that inhibit HIF PHD identifies new reagents to develop strategies to prevent the degradation of HIF by its selective PHD. These molecules are novel hypoxia mimetics that may provide new strategies to protect retinovasculature from hyperoxia.


Subject(s)
Enzyme Inhibitors/pharmacology , Hydrazones/pharmacology , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Reperfusion Injury/prevention & control , Retinal Diseases/prevention & control , Retinal Vessels/drug effects , Animals , Blotting, Western , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Hypoxia/prevention & control , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Luciferases/genetics , Mice , Mice, Transgenic , NIH 3T3 Cells , Promoter Regions, Genetic , Reperfusion Injury/enzymology , Retinal Diseases/enzymology , Transfection
13.
J Cell Biol ; 190(5): 881-92, 2010 Sep 06.
Article in English | MEDLINE | ID: mdl-20819940

ABSTRACT

Autophagy clears long-lived proteins and dysfunctional organelles and generates substrates for adenosine triphosphate production during periods of starvation and other types of cellular stress. Here we show that high mobility group box 1 (HMGB1), a chromatin-associated nuclear protein and extracellular damage-associated molecular pattern molecule, is a critical regulator of autophagy. Stimuli that enhance reactive oxygen species promote cytosolic translocation of HMGB1 and thereby enhance autophagic flux. HMGB1 directly interacts with the autophagy protein Beclin1 displacing Bcl-2. Mutation of cysteine 106 (C106), but not the vicinal C23 and C45, of HMGB1 promotes cytosolic localization and sustained autophagy. Pharmacological inhibition of HMGB1 cytoplasmic translocation by agents such as ethyl pyruvate limits starvation-induced autophagy. Moreover, the intramolecular disulfide bridge (C23/45) of HMGB1 is required for binding to Beclin1 and sustaining autophagy. Thus, endogenous HMGB1 is a critical pro-autophagic protein that enhances cell survival and limits programmed apoptotic cell death.


Subject(s)
Autophagy/physiology , HMGB1 Protein/metabolism , Antineoplastic Agents/metabolism , Apoptosis/genetics , Autophagy/genetics , Cell Line, Transformed , Cell Line, Tumor , Cytoplasm/genetics , Cytoplasm/metabolism , HCT116 Cells , HMGB1 Protein/chemistry , HMGB1 Protein/genetics , Humans , Mutation , Pancreatic Neoplasms/pathology , Protein Transport/genetics , RNA Interference , Starvation/genetics , Starvation/metabolism , Transfection
14.
Ophthalmology ; 116(3): 513-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19157560

ABSTRACT

PURPOSE: To determine the incidence of retinopathy of prematurity (ROP) over a 2-year period before and after a change in the practice of oxygen supplementation. DESIGN: Nonrandomized, retrospective study. PARTICIPANTS: All infants in a single Level III neonatal intensive care unit between the years of 2005 and 2007. METHODS: A prospective database recorded the gestational age, birth weight, stage and zone of ROP, threshold disease, treatment, final outcome and date of examination, maternal and infant demographics, and neonatal intensive care unit course. Year 1 (August 1, 2005 to July 31, 2006) includes a patient cohort who received the standard oxygen supplementation protocol, which has oxygen targets of 95% to 100% saturation. Year 2 (August 1, 2006 to July 31, 2007) includes a patient cohort who has strictly monitored oxygen targets of <34 weeks corrected gestational age oxygen limits of 80% to 95% and target 85% to 92% oxygen saturation and >34 weeks corrected gestational age limits of 85% to 100% and target 92% to 97% saturation. MAIN OUTCOME MEASURE: Incidence of ROP in year 1 before a change in oxygen protocol compared with the incidence of ROP in year 2 after a change in the oxygen protocol. RESULTS: A total of 114 children in year 1 and 108 children in year 2 were identified as having been born or transferred to the Fairview Nursery. Ninety-eight infants were examined before and 92 infants were examined after the change in oxygen standards, comprising 190 consecutive patients examined between September 2005 and October 2007. ROP was present in 35% of infants in group 1 before the change in oxygen protocol compared with 13% after the change in oxygen standards (P=0.001); stage 3 decreased from 11% to 2% (P=0.021); threshold disease decreased from 7% to 1% (P=0.066). Stage 0 (immature vessels, no ROP) incidence increased (pre/post-oxygen change 30%/51% stage 0, P=0.001). There were statistically significant differences in mode of delivery (P=0.007), sepsis <3 days of life (P=0.01), and oxygen at discharge (P=0.003). CONCLUSIONS: Lower oxygen targets at early gestational age and higher oxygen targets at older gestational age decrease the severity and incidence of ROP while inducing normal retinal development.


Subject(s)
Oxygen Consumption/physiology , Oxygen Inhalation Therapy/methods , Oxygen/administration & dosage , Retinopathy of Prematurity/therapy , Birth Weight , Child, Preschool , Female , Gestational Age , Humans , Incidence , Infant , Infant, Newborn , Male , Retinopathy of Prematurity/physiopathology , Retrospective Studies
15.
Proc Natl Acad Sci U S A ; 105(50): 19898-903, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19057008

ABSTRACT

Oxygen-induced retinopathy (OIR) in the mouse, like the analogous human disease retinopathy of prematurity, is an ischemic retinopathy dependent on oxygen-induced vascular obliteration. We tested the hypothesis that chemically overriding the oxygen-induced downregulation of hypoxia-inducible factor (HIF) activity would prevent vascular obliteration and subsequent pathologic neovascularization in the OIR model. Because the degradation of HIF-1alpha is regulated by prolyl hydroxylases, we examined the effect of systemic administration of a prolyl hydroxylase inhibitor, dimethyloxalylglycine, in the OIR model. Our results determine that stabilizing HIF activity in the early phase of OIR prevents the oxygen-induced central vessel loss and subsequent vascular tortuosity and tufting that is characteristic of OIR. Overall, these findings imply that simulating hypoxia chemically by stabilizing HIF activity during the causative ischemia phase (hyperoxia) of retinopathy of prematurity may be of therapeutic value in preventing progression to the proliferative stage of the disease.


Subject(s)
Amino Acids, Dicarboxylic/administration & dosage , Enzyme Inhibitors/administration & dosage , Oxygen/toxicity , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Retinopathy of Prematurity/prevention & control , Aerobiosis , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Disease Models, Animal , Erythropoietin/biosynthesis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/agonists , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Infant, Newborn , Kidney/metabolism , Liver/metabolism , Mice , Procollagen-Proline Dioxygenase/metabolism , Retina/metabolism , Retinopathy of Prematurity/chemically induced , Retinopathy of Prematurity/enzymology , Vascular Endothelial Growth Factor A/biosynthesis
16.
Immunity ; 29(1): 21-32, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18631454

ABSTRACT

The mammalian immune system discriminates between modes of cell death; necrosis often results in inflammation and adaptive immunity, whereas apoptosis tends to be anti-inflammatory and promote immune tolerance. We have examined apoptosis for the features responsible for tolerance; specifically, we looked at the roles of caspases and mitochondria. Our results show that caspase activation targeted the mitochondria to produce reactive oxygen species (ROS), which were critical to tolerance induction by apoptotic cells. ROS oxidized the potential danger signal high-mobility group box-1 protein (HMGB1) released from dying cells and thereby neutralized its stimulatory activity. Apoptotic cells failed to induce tolerance and instead stimulated immune responses by scavenging or by mutating a mitochondrial caspase target protein when ROS activity was prohibited. Similarly, blocking sites of oxidation in HMGB1 prevented tolerance induction by apoptotic cells. These results suggest that caspase-orchestrated mitochondrial events determine the impact of apoptotic cells on the immune response.


Subject(s)
Apoptosis/immunology , Caspases/immunology , HMGB1 Protein/immunology , Immune Tolerance/immunology , Mitochondria/metabolism , Animals , Dendritic Cells/immunology , HMGB1 Protein/metabolism , HeLa Cells , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Mitochondria/immunology , Oxidation-Reduction , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism
17.
J Biol Chem ; 283(9): 5699-707, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18096704

ABSTRACT

Hyperglycemia is an independent risk factor for development of vascular diabetic complications. Vascular dysfunction in diabetics manifests in a tissue-specific manner; macrovasculature is affected by atherosclerotic lesions, and microvascular complications are described as "aberrant angiogenesis": in the same patient angiogenesis is increased in some tissues (e.g. retinal neovascularization) and decreased in others (e.g. in skin). Molecular cell- and tissue-specific mechanisms regulating the response of vasculature to hyperglycemia remain unclear. Thrombospondin-1 (TSP-1), a potent antiangiogenic and proatherogenic protein, has been implicated in the development of several vascular diabetic complications (atherosclerosis, nephropathy, and cardiomyopathy). This study examines cell type-specific regulation of production of thrombospondin-1 by high glucose. We previously reported the increased expression of TSP-1 in the large arteries of diabetic animals. mRNA and protein levels were up-regulated in response to high glucose. Unlike in macrovascular cells, TSP-1 protein levels are dramatically decreased in response to high glucose in microvascular endothelial cells and retinal pigment epithelial cells (RPE). This down-regulation is post-transcriptional; mRNA levels are increased. In situ mRNA hybridization and immunohistochemistry revealed that the level of mRNA is up-regulated in RPE of diabetic rats, whereas the protein level is decreased. This cell type-specific posttranscriptional suppression of TSP-1 production in response to high glucose in microvascular endothelial cells and RPE is controlled by untranslated regions of TSP-1 mRNA that regulate coupling of TSP-1 mRNA to polysomes and its translation. The cell-specific regulation of TSP-1 suggests a potential mechanism for the aberrant angiogenesis in diabetics and TSP-1 involvement in development of various vascular diabetic complications.


Subject(s)
Angiogenesis Inhibitors/biosynthesis , Atherosclerosis/metabolism , Diabetes Complications/metabolism , Glucose/pharmacology , Hyperglycemia/metabolism , Neovascularization, Pathologic/metabolism , Sweetening Agents/pharmacology , Thrombospondin 1/biosynthesis , Animals , Atherosclerosis/etiology , Atherosclerosis/pathology , Cattle , Cells, Cultured , Diabetes Complications/pathology , Down-Regulation/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Hyperglycemia/complications , Hyperglycemia/pathology , In Situ Hybridization , Neovascularization, Pathologic/etiology , Organ Specificity/drug effects , Pigment Epithelium of Eye/metabolism , Pigment Epithelium of Eye/pathology , Protein Biosynthesis/drug effects , Rats , Rats, Zucker
18.
J Comp Neurol ; 501(2): 219-30, 2007 Mar 10.
Article in English | MEDLINE | ID: mdl-17226794

ABSTRACT

Hmgb1 belongs to a family of structure-specific DNA binding proteins with DNA chaperone-like properties that mediate chromatin remodeling in a wide range of nuclear processes including regulation of transcription, DNA repair, genome stability, and stress response. A diurnal oscillation of Hmgb1 at the protein level occurs in rat retinal photoreceptor cells and to a lesser extent in bipolar neurons. Expression of Hmgb1 was least at night at Zeitgeber time (ZT) 18 and maximal in the middle of the lights-on period (ZT6). Since rhythmic expression of Hmgb1 protein in photoreceptors continued in complete darkness, it is likely under control of a circadian clock. Within photoreceptor nuclei, Hmgb1 colocalized with acetylated histone H3, a marker of euchromatin. Outside the nucleus a distinct smaller-sized isoform of Hmgb1 was present in photoreceptor inner segments and bound to a membrane fraction with characteristics of endoplasmic reticulum membranes. The rhythmic expression of Hmgb1 protein may underlie the circadian change in chromatin remodeling in addition to histone acetylation.


Subject(s)
Circadian Rhythm , HMGB1 Protein/metabolism , Photoreceptor Cells/metabolism , Aged , Animals , Euchromatin/metabolism , Female , Gene Expression Regulation , Humans , Immunohistochemistry , Microscopy, Electron, Transmission/methods , Middle Aged , Photoreceptor Cells/ultrastructure , Rats , Rats, Long-Evans , Retina/cytology
19.
Invest Ophthalmol Vis Sci ; 47(11): 4935-41, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17065510

ABSTRACT

PURPOSE: To test whether triamcinolone acetonide (TA) inhibits angiogenesis induced by IL-6 or VEGF and whether this inhibition is through antagonism of the IL-6 or the VEGF receptor 2. METHODS: A rat cornea micropocket assay was used to initiate IL-6- and VEGF-mediated angiogenesis. The ability of TA or neutralizing VEGF antibody to inhibit IL-6- or VEGF-mediated neovascularization was analyzed by measuring vessel length, vessel extension, and vessel area. The phosphorylation of signal transduction activator 3 (STAT3), VEGF receptor, and extracellular signal-regulated kinase 1/2 (ERK1/2) was determined by Western blot in human umbilical vein endothelial cell (HUVEC) lysates after stimulus with IL-6 or VEGF, with and without TA pretreatment. The effect of IL-6 or TA on STAT3 expression in cornea was determined by Western blot. RESULTS: IL-6 induced corneal angiogenesis in a dose-dependent manner, with 350 ng producing a peak at day 6. VEGF antibodies and TA blocked IL-6-mediated limbal neovascularization. TA also directly inhibited angiogenesis stimulated by a VEGF pellet; the glucocorticoid receptor antagonist mifepristone neutralized TA inhibition of angiogenesis. TA did not inhibit IL-6-induced STAT3 phosphorylation and did not inhibit VEGF-induced phosphorylation of the VEGF receptor 2 or of ERK1/2 in endothelial cells, but TA decreased IL-6-induced STAT3 expression in cornea. CONCLUSIONS: IL-6- and VEGF-mediated corneal neovascularization are blocked by TA through the mifepristone-sensitive steroid receptor. TA inhibits IL-6-induced STAT3 expression in cornea, but it does not inhibit activation of the IL-6 or the VEGF receptor in cultured human endothelial cells. This finding has two implications. The fact that TA directly inhibits VEGF action implies that other factors may be critical to angiogenesis and sensitive to glucocorticoids.


Subject(s)
Corneal Neovascularization/drug therapy , Glucocorticoids/pharmacology , Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Triamcinolone Acetonide/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Blotting, Western , Chorioallantoic Membrane/blood supply , Corneal Neovascularization/chemically induced , Corneal Neovascularization/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Interleukin-6/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Rats , Rats, Inbred F344 , STAT3 Transcription Factor/metabolism , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/pharmacology
20.
Exp Cell Res ; 312(18): 3526-38, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16962095

ABSTRACT

Oxidative stress can induce a covalent disulfide bond between protein and peptide thiols that is reversible through enzymatic catalysis. This process provides a post-translational mechanism for control of protein function and may also protect thiol groups from irreversible oxidation. High mobility group protein B1 (Hmgb1), a DNA-binding structural chromosomal protein and transcriptional co-activator was identified as a substrate of glutaredoxin. Hmgb1 contains 3 cysteines, Cys23, 45, and 106. In mild oxidative conditions, Cys23 and Cys45 readily form an intramolecular disulfide bridge, whereas Cys106 remains in the reduced form. The disulfide bond between Cys23 and Cys45 is a target of glutathione-dependent reduction by glutaredoxin. Endogenous Hmgb1 as well as GFP-tagged wild-type Hmgb1 co-localize in the nucleus of CHO cells. While replacement of Hmgb1 Cys23 and/or 45 with serines did not affect the nuclear distribution of the mutant proteins, Cys106-to-Ser and triple cysteine mutations impaired nuclear localization of Hmgb1. Our cysteine targeted mutational analysis suggests that Cys23 and 45 induce conformational changes in response to oxidative stress, whereas Cys106 appears to be critical for the nucleocytoplasmic shuttling of Hmgb1.


Subject(s)
Active Transport, Cell Nucleus/physiology , HMGB1 Protein/metabolism , Oxidative Stress , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Cysteine/metabolism , Disulfides/metabolism , Glutaredoxins , HMGB1 Protein/chemistry , HMGB1 Protein/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Oxidation-Reduction , Oxidoreductases/metabolism , Protein Disulfide Reductase (Glutathione)/metabolism , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...