Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cells ; 11(14)2022 07 13.
Article in English | MEDLINE | ID: mdl-35883628

ABSTRACT

The GPCR SUCNR1/GPR91 exerts proangiogenesis upon stimulation with the Krebs cycle metabolite succinate. GPCR signaling depends on the surrounding environment and intracellular localization through location bias. Here, we show by microscopy and by cell fractionation that in neurons, SUCNR1 resides at the endoplasmic reticulum (ER), while being fully functional, as shown by calcium release and the induction of the expression of the proangiogenic gene for VEGFA. ER localization was found to depend upon N-glycosylation, particularly at position N8; the nonglycosylated mutant receptor localizes at the plasma membrane shuttled by RAB11. This SUCNR1 glycosylation is physiologically regulated, so that during hypoxic conditions, SUCNR1 is deglycosylated and relocates to the plasma membrane. Downstream signal transduction of SUCNR1 was found to activate the prostaglandin synthesis pathway through direct interaction with COX-2 at the ER; pharmacologic antagonism of the PGE2 EP4 receptor (localized at the nucleus) was found to prevent VEGFA expression. Concordantly, restoring the expression of SUCNR1 in the retina of SUCNR1-null mice renormalized vascularization; this effect is markedly diminished after transfection of the plasma membrane-localized SUCNR1 N8A mutant, emphasizing that ER localization of the succinate receptor is necessary for proper vascularization. These findings uncover an unprecedented physiologic process where GPCR resides at the ER for signaling function.


Subject(s)
Receptors, G-Protein-Coupled , Succinic Acid , Animals , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Hypoxia , Mice , Receptors, G-Protein-Coupled/metabolism , Succinates , Succinic Acid/metabolism
2.
PLoS Biol ; 16(5): e2003619, 2018 05.
Article in English | MEDLINE | ID: mdl-29771909

ABSTRACT

During the development of the visual system, high levels of energy are expended propelling axons from the retina to the brain. However, the role of intermediates of carbohydrate metabolism in the development of the visual system has been overlooked. Here, we report that the carbohydrate metabolites succinate and α-ketoglutarate (α-KG) and their respective receptor-GPR91 and GPR99-are involved in modulating retinal ganglion cell (RGC) projections toward the thalamus during visual system development. Using ex vivo and in vivo approaches, combined with pharmacological and genetic analyses, we revealed that GPR91 and GPR99 are expressed on axons of developing RGCs and have complementary roles during RGC axon growth in an extracellular signal-regulated kinases 1 and 2 (ERK1/2)-dependent manner. However, they have no effects on axon guidance. These findings suggest an important role for these receptors during the establishment of the visual system and provide a foundational link between carbohydrate metabolism and axon growth.


Subject(s)
Carbohydrate Metabolism , Neuronal Outgrowth , Receptors, G-Protein-Coupled/metabolism , Receptors, Purinergic P2/metabolism , Retina/embryology , Animals , Ketoglutaric Acids/metabolism , MAP Kinase Signaling System , Mice , Mice, Knockout , Retina/metabolism , Retinal Ganglion Cells/metabolism , Succinic Acid/metabolism
3.
Sci Rep ; 6: 37391, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27874077

ABSTRACT

Pathological choroidal neovascularization (CNV) is the common cause of vision loss in patients with age-related macular degeneration (AMD). Macrophages possess potential angiogenic function in CNV. We have demonstrated that human T lymphocyte-derived microparticles (LMPs) exert a potent antiangiogenic effect in several pathological neovascularization models. In this study, we investigated the alteration of proangiogenic properties of macrophages by LMPs treatment in vitro and in vivo models. LMPs regulated the expression of several angiogenesis-related factors in macrophages and consequently stimulated their antiangiogenic effects evidenced by the suppression of the proliferation of human retinal endothelial cells in co-culture experiments. The involvement of CD36 receptor in LMPs uptake by macrophages was demonstrated by in vitro assays and by immunostaining of choroidal flat mounts. In addition, ex vivo experiments showed that CD36 mediates the antiangiogenic effect of LMPs in murine and human choroidal explants. Furthermore, intravitreal injection of LMPs in the mouse model of laser-induced CNV significantly suppressed CNV in CD36 dependent manner. The results of this study suggested an ability of LMPs to alter the gene expression pattern of angiogenesis-related factors in macrophages, which provide important information for a new therapeutic approach for efficiently interfering with both vascular and extravascular components of CNV.


Subject(s)
Cell-Derived Microparticles/metabolism , Choroidal Neovascularization/pathology , Lymphocytes/metabolism , Macrophages/metabolism , Neovascularization, Physiologic , Animals , Biomarkers/metabolism , CD36 Antigens/metabolism , Cell Polarity , Cell Proliferation , Gene Expression Regulation , Humans , Lasers , Mice , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells
4.
J Immunol ; 195(7): 3402-15, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26304990

ABSTRACT

Preterm birth (PTB) is firmly linked to inflammation regardless of the presence of infection. Proinflammatory cytokines, including IL-1ß, are produced in gestational tissues and can locally upregulate uterine activation proteins. Premature activation of the uterus by inflammation may lead to PTB, and IL-1 has been identified as a key inducer of this condition. However, all currently available IL-1 inhibitors are large molecules that exhibit competitive antagonism properties by inhibiting all IL-1R signaling, including transcription factor NF-κB, which conveys important physiological roles. We hereby demonstrate the efficacy of a small noncompetitive (all-d peptide) IL-1R-biased ligand, termed rytvela (labeled 101.10) in delaying IL-1ß-, TLR2-, and TLR4-induced PTB in mice. The 101.10 acts without significant inhibition of NF-κB, and instead selectively inhibits IL-1R downstream stress-associated protein kinases/transcription factor c-jun and Rho GTPase/Rho-associated coiled-coil-containing protein kinase signaling pathways. The 101.10 is effective at decreasing proinflammatory and/or prolabor genes in myometrium tissue and circulating leukocytes in all PTB models independently of NF-κB, undermining NF-κB role in preterm labor. In this work, biased signaling modulation of IL-1R by 101.10 uncovers a novel strategy to prevent PTB without inhibiting NF-κB.


Subject(s)
Inflammation/immunology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Peptides/pharmacology , Premature Birth/prevention & control , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Female , Interleukin-1beta/immunology , Mice , Myometrium/metabolism , NF-kappa B/metabolism , Pregnancy , Receptors, Interleukin-1/antagonists & inhibitors , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Uterus/immunology , rho GTP-Binding Proteins/antagonists & inhibitors , rho-Associated Kinases/antagonists & inhibitors
5.
Arterioscler Thromb Vasc Biol ; 34(2): 285-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24285580

ABSTRACT

OBJECTIVE: Prompt post-hypoxia-ischemia (HI) revascularization has been suggested to improve outcome in adults and newborn subjects. Other than hypoxia-inducible factor, sensors of metabolic demand remain largely unknown. During HI, anaerobic respiration is arrested resulting in accumulation of carbohydrate metabolic intermediates. As such succinate readily increases, exerting its biological effects via a specific receptor, G-protein-coupled receptor (GPR) 91. We postulate that succinate/GPR91 enhances post-HI vascularization and reduces infarct size in a model of newborn HI brain injury. APPROACH AND RESULTS: The Rice-Vannucci model of neonatal HI was used. Succinate was measured by mass spectrometry, and microvascular density was evaluated by quantification of lectin-stained cryosection. Gene expression was evaluated by real-time polymerase chain reaction. Succinate levels rapidly increased in the penumbral region of brain infarcts. GPR91 was foremost localized not only in neurons but also in astrocytes. Microvascular density increased at 96 hours after injury in wild-type animals; it was diminished in GPR91-null mice leading to an increased infarct size. Stimulation with succinate led to an increase in growth factors implicated in angiogenesis only in wild-type mice. To explain the mode of action of succinate/GPR91, we investigated the role of prostaglandin E2-prostaglandin E receptor 4, previously proposed in neural angiogenesis. Succinate-induced vascular endothelial growth factor expression was abrogated by a cyclooxygenase inhibitor and a selective prostaglandin E receptor 4 antagonist. This antagonist also abolished succinate-induced neovascularization. CONCLUSIONS: We uncover a dominant metabolic sensor responsible for post-HI neurovascular adaptation, notably succinate/GPR91, acting via prostaglandin E2-prostaglandin E receptor 4 to govern expression of major angiogenic factors. We propose that pharmacological intervention targeting GPR91 could improve post-HI brain recovery.


Subject(s)
Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Cerebral Infarction/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , Neuroprotective Agents/pharmacology , Receptors, G-Protein-Coupled/agonists , Succinic Acid/pharmacology , Angiogenic Proteins/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cell Line , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Infarction/etiology , Cerebral Infarction/genetics , Cerebral Infarction/metabolism , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hypoxia-Ischemia, Brain/etiology , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Injections, Intraventricular , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Prostaglandin Antagonists/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Prostaglandin E, EP4 Subtype/drug effects , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction/drug effects , Succinic Acid/administration & dosage , Succinic Acid/metabolism , Time Factors , Tissue Culture Techniques
6.
PLoS One ; 8(7): e69552, 2013.
Article in English | MEDLINE | ID: mdl-23922736

ABSTRACT

Angiogenesis of the microvasculature is central to the etiology of many diseases including proliferative retinopathy, age-related macular degeneration and cancer. A mouse model of microvascular angiogenesis would be very valuable and enable access to a wide range of genetically manipulated tissues that closely approximate small blood vessel growth in vivo. Vascular endothelial cells cultured in vitro are widely used, however, isolating pure vascular murine endothelial cells is technically challenging. A microvascular mouse explant model that is robust, quantitative and can be reproduced without difficulty would overcome these limitations. Here we characterized and optimized for reproducibility an organotypic microvascular angiogenesis mouse and rat model from the choroid, a microvascular bed in the posterior of eye. The choroidal tissues from C57BL/6J and 129S6/SvEvTac mice and Sprague Dawley rats were isolated and incubated in Matrigel. Vascular sprouting was comparable between choroid samples obtained from different animals of the same genetic background. The sprouting area, normalized to controls, was highly reproducible between independent experiments. We developed a semi-automated macro in ImageJ software to allow for more efficient quantification of sprouting area. Isolated choroid explants responded to manipulation of the external environment while maintaining the local interactions of endothelial cells with neighboring cells, including pericytes and macrophages as evidenced by immunohistochemistry and fluorescence-activated cell sorting (FACS) analysis. This reproducible ex vivo angiogenesis assay can be used to evaluate angiogenic potential of pharmacologic compounds on microvessels and can take advantage of genetically manipulated mouse tissue for microvascular disease research.


Subject(s)
Biological Assay/methods , Choroid/blood supply , Microvessels/physiology , Models, Biological , Neovascularization, Physiologic , Aging/physiology , Angiogenesis Inducing Agents/pharmacology , Animals , Biological Assay/standards , Choroid/drug effects , Culture Media/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microvessels/drug effects , Monocytes/cytology , Monocytes/metabolism , Neovascularization, Physiologic/drug effects , Pericytes/cytology , Pericytes/metabolism , Rats , Rats, Sprague-Dawley , Reference Standards , Reproducibility of Results , Retinal Pigment Epithelium/physiology
7.
Invest Ophthalmol Vis Sci ; 54(9): 6084-92, 2013 Sep 09.
Article in English | MEDLINE | ID: mdl-23920372

ABSTRACT

PURPOSE: Choroidal neovascularization (CNV) is a major cause of vision loss in which choroidal vessels penetrate the RPE-an important source of growth factors, including nerve growth factor (NGF), whose activation via the p75NTR receptor promotes apoptosis and inhibits angiogenesis. We demonstrated previously that human T-lymphocyte-derived microparticles (LMPs) significantly inhibit angiogenesis in several models of ocular neovascularization. We investigated how LMPs modulate pro- and antiangiogenic microenvironments during choroidal angiogenesis. METHODS: Antiangiogenic effects of LMPs were investigated using a rat model of choroidal angiogenesis. The impact of LMPs on expression of major angiogenic factors was assessed by real-time quantitative PCR (qPCR). To determine whether p75NTR signalling was implicated in LMPs-induced activities, we used a specific antibody and short hairpin RNA (shRNA) targeting p75NTR. Cellular apoptosis was determined via evaluation of activated caspase-3 and annexin V binding. RESULTS: The LMPs time-dependently inhibited choroidal angiogenesis by more than 64% after 48 hours of treatment. Removal of the RPE from choroidal explants abolished the antiangiogenic effects of LMPs. The mRNA levels of pigment epithelium-derived factor (PEDF) and NGF were increased significantly following LMPs treatment of intact, but not RPE-removed choroids. Downregulation of PEDF and p75NTR significantly blocked the antiangiogenic effects of LMPs. Finally, induction of choroidal endothelial cell apoptosis by LMPs was dependent on p75NTR. CONCLUSIONS: We demonstrate for the first time to our knowledge that LMPs markedly inhibit choroidal angiogenesis via mechanisms that are dependent on the integrity of the RPE, and that are mediated largely by the PEDF and proapoptotic activities of p75NTR.


Subject(s)
Apoptosis , Choroidal Neovascularization/genetics , Gene Expression Regulation, Developmental , RNA/genetics , Receptors, Nerve Growth Factor/genetics , T-Lymphocytes/metabolism , Animals , Animals, Newborn , Cells, Cultured , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Nerve Tissue Proteins , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Growth Factor , Receptors, Nerve Growth Factor/biosynthesis
8.
Am J Physiol Regul Integr Comp Physiol ; 304(1): R10-22, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23152113

ABSTRACT

Acute renal failure (ARF) is a serious medical complication characterized by an abrupt and sustained decline in renal function. Despite significant advances in supportive care, there is currently no effective treatment to restore renal function. PGE(2) is a lipid hormone mediator abundantly produced in the kidney, where it acts locally to regulate renal function; several studies suggest that modulating EP(4) receptor activity could improve renal function following kidney injury. An optimized peptidomimetic ligand of EP(4) receptor, THG213.29, was tested for its efficacy to improve renal function (glomerular filtration rate, renal plasma flow, and urine output) and histological changes in a model of ARF induced by either cisplatin or renal artery occlusion in Sprague-Dawley rats. THG213.29 modulated PGE(2)-binding dissociation kinetics, indicative of an allosteric binding mode. Consistently, THG213.29 antagonized EP(4)-mediated relaxation of piglet saphenous vein rings, partially inhibited EP(4)-mediated cAMP production, but did not affect Gα(i) activation or ß-arrestin recruitment. In vivo, THG213.29 significantly improved renal function and histological changes in cisplatin- and renal artery occlusion-induced ARF models. THG213.29 increased mRNA expression of heme-oxygenase 1, Bcl2, and FGF-2 in renal cortex; correspondingly, in EP(4)-transfected HEK293 cells, THG213.29 augmented FGF-2 and abrogated EP(4)-dependent overexpression of inflammatory IL-6 and of apoptotic death domain-associated protein and BCL2-associated agonist of cell death. Our results demonstrate that THG213.29 represents a novel class of diuretic agent with noncompetitive allosteric modulator effects on EP(4) receptor, resulting in improved renal function and integrity following acute renal failure.


Subject(s)
Acute Kidney Injury/drug therapy , Kidney/drug effects , Kidney/physiology , Oligopeptides/therapeutic use , Receptors, Prostaglandin E, EP4 Subtype/agonists , Recovery of Function/drug effects , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , Cisplatin/adverse effects , Cyclic AMP/biosynthesis , Disease Models, Animal , Dogs , Female , Fibroblast Growth Factor 2/biosynthesis , Glomerular Filtration Rate/drug effects , HEK293 Cells , Heme Oxygenase-1/biosynthesis , Humans , Interleukin-6/biosynthesis , Male , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats , Rats, Sprague-Dawley , Renal Plasma Flow/physiology , Saphenous Vein/drug effects , Saphenous Vein/pathology , Swine/physiology
9.
Neonatology ; 100(4): 343-53, 2011.
Article in English | MEDLINE | ID: mdl-21968165

ABSTRACT

Retinopathy of prematurity (ROP), an ocular disease characterized by the onset of vascular abnormalities in the developing retina, is the major cause of visual impairment and blindness in premature neonates. ROP is a complex condition in which various factors participate at different stages of the disease leading to microvascular degeneration followed by neovascularization, which in turn predisposes to retinal detachment. Current ablative therapies (cryotherapy and laser photocoagulation) used in the clinic for the treatment of ROP have limitations and patients can still have long-term effects even after successful treatment. New treatment modalities are still emerging. The most promising are the therapies directed against VEGF; more recently the use of preventive dietary supplementation with ω-3 polyunsaturated fatty acid may also be promising. Other than pharmacologic and nutritional approaches, cell-based strategies for vascular repair are likely to arise from advances in regenerative medicine using stem cells. In addition to all of these, a greater understanding of other factors involved in regulating pathologic retinal angiogenesis continues to emerge, suggesting potential targets for therapeutic approaches. This review summarizes an update on the current state of knowledge on ROP from our and other laboratories, with particular focus on the role of nitro-oxidative stress and notably trans-arachidonic acids in microvascular degeneration, semaphorin 3 operating as vasorepulsive molecules in the avascular hypoxic retina and in turn impairing revascularization, succinate and its receptor GPR91 in neuron-mediated retinal neovascularization, and ω-3 lipids as modulators of preretinal neovascularization.


Subject(s)
Infant, Premature , Retinopathy of Prematurity/etiology , Gestational Age , Humans , Infant, Newborn , Lipid Peroxidation , Neovascularization, Pathologic , Oxidative Stress , Oxygen/physiology , Oxygen/therapeutic use , Receptors, G-Protein-Coupled , Retina/embryology , Retinal Vessels/embryology , Retinopathy of Prematurity/prevention & control , Retinopathy of Prematurity/therapy , Risk Factors , Semaphorins , Succinic Acid , Vascular Endothelial Growth Factor A
10.
Mol Cell Biol ; 23(4): 1316-33, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12556491

ABSTRACT

As do cytokine receptors and receptor tyrosine kinases, G protein-coupled receptors (GPCRs) signal to Janus kinases (Jaks) and signal transducers and activators of transcription (STATs). However, the early biochemical events linking GPCRs to this signaling pathway have been unclear. Here we show that GPCR-stimulated Rac activity and the subsequent generation of reactive oxygen species are necessary for activating tyrosine phosphorylation of Jaks and STAT-dependent transcription. The requirement for Rac activity can be overcome by addition of hydrogen peroxide. Expression of activated mutants of Rac1 is sufficient to activate Jak2 and STAT-dependent transcription, and the activation of Jak2 correlates with the ability of Rac1 to bind to NADPH oxidase subunit p67(phox). We further show that GPCR agonists stimulate tyrosine phosphorylation of STAT1 and STAT3 proteins in a Rac-dependent manner. The tyrosine phosphorylation of STAT3 is biphasic; the first peak of phosphorylation is weak and correlates with rapid activation of Jaks by GPCRs, whereas the second peak is stronger and requires the synthesis of an autocrine factor. Rho also plays an essential role in the induction of STAT transcriptional activity. Our results highlight a novel role for Rho GTPases in mediating the regulatory effects of GPCRs on STAT-dependent gene expression.


Subject(s)
DNA-Binding Proteins/metabolism , GTP-Binding Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/metabolism , Angiotensin II/pharmacology , Animals , Antioxidants/pharmacology , Bacterial Toxins/pharmacology , Cells, Cultured , DNA-Binding Proteins/genetics , Humans , Janus Kinase 2 , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Mutation , Oxidants/pharmacology , Phosphorylation , Protein-Tyrosine Kinases/genetics , Rats , Reactive Oxygen Species/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , STAT1 Transcription Factor , STAT2 Transcription Factor , STAT3 Transcription Factor , Signal Transduction , Thrombin/pharmacology , Trans-Activators/genetics , Transcription, Genetic , Tyrosine/metabolism , rac1 GTP-Binding Protein/drug effects , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/drug effects , rho GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...