Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Front Cell Dev Biol ; 9: 630645, 2021.
Article de Anglais | MEDLINE | ID: mdl-33553187

RÉSUMÉ

Ischemic retinopathies (IRs), such as retinopathy of prematurity and diabetic retinopathy, are characterized by an initial phase of microvascular degeneration that results in retinal ischemia, followed by exaggerated pathologic neovascularization (NV). Mesenchymal stromal cells (MSCs) have potent pro-angiogenic and anti-inflammatory properties associated with tissue repair and regeneration, and in this regard exert protection to neurons in ischemic and degenerative conditions; however, the exact mechanisms underlying these functions remain largely unknown. Class III Semaphorins (A-G) are particularly implicated in regulating neural blood supply (as well as neurogenesis) by suppressing angiogenesis and affecting myeloid cell function; this is the case for distinct neuropillin-activating Sema3A as well as PlexinD1-activating Sema3E; but during IR the former Sema3A increases while Sema3E decreases. We investigated whether retinal vascular repair actions of MSCs are exerted by normalizing Semaphorin and downstream cytokines in IR. Intravitreal administration of MSCs or their secretome (MSCs-conditioned media [MSCs-CM]) significantly curtailed vasoobliteration as well as aberrant preretinal NV in a model of oxygen-induced retinopathy (OIR). The vascular repair effects of MSCs-CM in the ischemic retina were associated with restored levels of Sema3E. Vascular benefits of MSCs-CM were reversed by anti-Sema3E; while intravitreal injection of anti-angiogenic recombinant Sema3E (rSema3E) in OIR-subjected mice reproduced effects of MSCs-CM by inhibiting as expected preretinal NV but also by decreasing vasoobliteration. To explain these opposing vascular effects of Sema3E we found in OIR high retinal levels, respectively, of the pro- and anti-angiogenic IL-17A and Sema3A-regulating IL-1ß; IL-17A positively affected expression of IL-1ß. rSema3E decreased concentrations of these myeloid cell-derived pro-inflammatory cytokines in vitro and in vivo. Importantly, IL-17A suppression by MSCs-CM was abrogated by anti-Sema3E neutralizing antibody. Collectively, our findings provide novel evidence by which MSCs inhibit aberrant NV and diminish vasoobliteration (promoting revascularization) in retinopathy by restoring (at least in part) neuronal Sema3E levels that reduce pathological levels of IL-17A (and in turn other proinflammatory factors) in myeloid cells. The ability of MSCs to generate a microenvironment permissive for vascular regeneration by controlling the production of neuronal factors involved in immunomodulatory activities is a promising opportunity for stem cell therapy in ocular degenerative diseases.

2.
Development ; 145(1)2018 01 08.
Article de Anglais | MEDLINE | ID: mdl-29180574

RÉSUMÉ

MicroRNAs are key regulators of angiogenesis, as illustrated by the vascular defects observed in miR-126-deficient animals. The miR-126 duplex gives rise to two mature microRNAs (miR-126-3p and -5p). The vascular defects in these mutant animals were attributed to the loss of miR-126-3p but the role of miR-126-5p during normal angiogenesis in vivo remains unknown. Here, we show that miR-126-5p is expressed in endothelial cells but also by retinal ganglion cells (RGCs) of the mouse postnatal retina and participates in protecting endothelial cells from apoptosis during the establishment of the retinal vasculature. miR-126-5p negatively controls class 3 semaphorin protein (Sema3A) in RGCs through the repression of SetD5, an uncharacterized member of the methyltransferase family of proteins. In vitro, SetD5 controls Sema3A expression independently of its SET domain and co-immunoprecipitates with BRD2, a bromodomain protein that recruits transcription regulators onto the chromatin. Both SetD5 and BRD2 bind to the transcription start site and to upstream promoter regions of the Sema3a locus and BRD2 is necessary for the regulation of Sema3A expression by SetD5. Thus, neuronally expressed miR-126-5p regulates angiogenesis by protecting endothelial cells of the developing retinal vasculature from apoptosis.


Sujet(s)
Apoptose/physiologie , Cellules endothéliales/métabolisme , Methyltransferases/biosynthèse , microARN/biosynthèse , Neurones/métabolisme , Rétine/métabolisme , Animaux , Survie cellulaire/physiologie , Cellules endothéliales/cytologie , Souris , Souris knockout , microARN/génétique , Néovascularisation physiologique/physiologie , Neurones/cytologie , Éléments de réponse/physiologie , Rétine/cytologie , Sémaphorine-3A/génétique , Sémaphorine-3A/métabolisme
3.
J Neuroinflammation ; 14(1): 181, 2017 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-28874201

RÉSUMÉ

BACKGROUND: Tetrahydrobiopterin (BH4) is an essential cofactor in multiple metabolic processes and plays an essential role in maintaining the inflammatory and neurovascular homeostasis. In this study, we have investigated the deleterious effects of BH4 deficiency on retinal vasculature during development. METHODS: hph-1 mice, which display deficiency in BH4 synthesis, were used to characterize the inflammatory effects and the integrity of retinal microvasculature. BH4 levels in retinas from hph-1 and wild type (WT) mice were measured by LC-MS/MS. Retinal microvascular area and microglial cells number were quantified in hph-1 and WT mice at different ages. Retinal expression of pro-inflammatory, anti-angiogenic, and neuronal-derived factors was analyzed by qPCR. BH4 supplementation was evaluated in vitro, ex-vivo, and in vivo models. RESULTS: Our findings demonstrated that BH4 levels in the retina from hph-1 mice were significantly lower by ~ 90% at all ages analyzed compared to WT mice. Juvenile hph-1 mice showed iris atrophy, persistent fetal vasculature, significant increase in the number of microglial cells (p < 0.01), as well as a marked degeneration of the retinal microvasculature. Retinal microvascular alterations in juvenile hph-1 mice were associated with a decreased expression in Norrin (0.2-fold) and its receptor Frizzled-4 (FZD4; 0.51-fold), as well as with an augmented expression of pro-inflammatory factors such as IL-6 (3.2-fold), NRLP-3 (4.4-fold), IL-1ß (8.6-fold), and the anti-angiogenic factor thrombospondin-1 (TSP-1; 17.5-fold). We found that TSP-1 derived from activated microglial cells is a factor responsible of inducing microvascular degeneration, but BH4 supplementation markedly prevented hyperoxia-induced microglial activation in vitro and microvascular injury in an ex-vivo model of microvascular angiogenesis and an in vivo model of oxygen-induced retinopathy (OIR). CONCLUSION: Our findings reveal that BH4 is a key cofactor in regulating the expression of inflammatory and anti-angiogenic factors that play an important function in the maintenance of retinal microvasculature.


Sujet(s)
Microvaisseaux/métabolisme , Phénylcétonuries/métabolisme , Rétine/métabolisme , Dégénérescence de la rétine/métabolisme , Vaisseaux rétiniens/métabolisme , Animaux , Inflammation/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Souris , Souris de lignée C57BL , Souris transgéniques , Microvaisseaux/anatomopathologie , Phénylcétonuries/génétique , Phénylcétonuries/anatomopathologie , Complexe répresseur Polycomb-1/génétique , Rétine/anatomopathologie , Dégénérescence de la rétine/génétique , Dégénérescence de la rétine/anatomopathologie , Vaisseaux rétiniens/anatomopathologie
4.
Invest Ophthalmol Vis Sci ; 58(1): 461-469, 2017 01 01.
Article de Anglais | MEDLINE | ID: mdl-28122088

RÉSUMÉ

Purpose: Spectral-domain optical coherence tomography (SD-OCT) is widely used in clinical ophthalmology and recently gained popularity in laboratory research involving small rodents. Its noninvasive nature allows repeated measurements, thereby decreasing the number of animals required. However, when used at a conventional dosage, xylazine (an α2-adrenoceptor) can cause irreversible corneal calcification, especially among young rodents. In the present study, we test whether corneal calcification associated with xylazine is mediated by the α2-adrenoceptor. Methods: Our study tested Sprague-Dawley rats, Long-Evans rats, and CD-1 mice (postnatal day [P]14). Retinal images were captured by SD-OCT. Quantitative PCR (qPCR) was used to study gene expression, whereas receptor localization was examined by immunofluorescent staining followed by confocal microscopy. Calcium deposits were detected via von Kossa staining. Results: When used at dosages appropriate for adult animals, ketamine-xylazine anesthetics led to a high rate of respiratory failure, increased apoptotic activity in the corneal epithelium, and irreversible corneal calcification in P14 rat pups. Meanwhile, OCT image quality decreased drastically as a result of corneal calcification among animals recovering from anesthesia. α2-Adrenoceptor subtypes were highly expressed on P14, in line with rodents' age-specific sensitivity to xylazine. Clonidine, a potent α2-adrenoceptor agonist, dose-dependently induced corneal calcification, which could be prevented by an α2-adrenoceptor antagonist. Conclusions: These data suggest that α2-adrenoceptors contribute to corneal calcification in young rodents. Therefore, we developed a suitable OCT imaging protocol for this cohort, including a carefully tailored ketamine-xylazine dosage (60 mg/kg and 2.5 kg/mg, respectively).


Sujet(s)
Calcinose/prévention et contrôle , Cornée/effets des médicaments et des substances chimiques , Maladies de la cornée/prévention et contrôle , Tomographie par cohérence optique/méthodes , Xylazine/toxicité , Agonistes des récepteurs alpha-2 adrénergiques/administration et posologie , Agonistes des récepteurs alpha-2 adrénergiques/toxicité , Animaux , Calcinose/anatomopathologie , Calcium/métabolisme , Cornée/métabolisme , Cornée/anatomopathologie , Maladies de la cornée/induit chimiquement , Maladies de la cornée/anatomopathologie , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Immunohistochimie , Souris , Microscopie confocale , Rats , Rat Long-Evans , Rat Sprague-Dawley , Xylazine/administration et posologie
5.
Oxid Med Cell Longev ; 2017: 3940241, 2017.
Article de Anglais | MEDLINE | ID: mdl-29410732

RÉSUMÉ

Ischemic retinopathies (IRs), such as retinopathy of prematurity (ROP), diabetic retinopathy (DR), and (in many cases) age-related macular degeneration (AMD), are ocular disorders characterized by an initial phase of microvascular changes that results in ischemia, followed by a second phase of abnormal neovascularization that may culminate into retinal detachment and blindness. IRs are complex retinal conditions in which several factors play a key role during the development of the different pathological stages of the disease. Increasing evidence reveals that oxidative stress and inflammatory processes are important contributors to the pathogenesis of IRs. Despite the beneficial effects of the photocoagulation and anti-VEGF therapy during neovascularization phase, the need to identify novel targets to prevent initial phases of these ocular pathologies is still needed. In this review, we provide an update on the involvement of oxidative stress and inflammation in the progression of IRs and address some therapeutic interventions by using antioxidants and anti-inflammatory agents.


Sujet(s)
Rétinopathie diabétique/métabolisme , Rétinopathie hypertensive/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Stress oxydatif/physiologie , Rétinopathie du prématuré/métabolisme , Rétinopathie diabétique/anatomopathologie , Humains , Rétinopathie hypertensive/anatomopathologie , Stress oxydatif/génétique , Rétinopathie du prématuré/anatomopathologie
6.
Invest Ophthalmol Vis Sci ; 56(13): 7897-907, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26670826

RÉSUMÉ

PURPOSE: This study evaluated the effects of BIBF1120, a novel triple angiokinase inhibitor against pathological retinal neovascularization. METHODS: BIBF1120 effect on development of the normal retinal vasculature was evaluated in Sprague-Dawley rat pups. Two models of ischemic oxygen-induced retinopathy (OIR) and the aortic ring assay were used to assess the antiangiogenic effects of BIBF1120. In the vaso-obliteration model (VO), rat pups were exposed to 80% O2 from postnatal day (P) 5 to P10. In the preretinal neovascularization (NV) model, rat pups were exposed to cycling O2 (50% and 10%) from P1 to P14, followed by room air until P18. Animals were intravitreally or orally treated with BIBF1120. Retinal vasculature, VO, and NV were evaluated in retinal flat mounts. Retinal expression of VEGF, Delta-like ligand 4 (Dll4), Netrin-1, Ephrin-B2, and EphB4 was analyzed by quantitative PCR and Western blot analysis. RESULTS: BIBF1120 interfered with normal retinal vascular development and microvessel branching in the aortic assay. However, in VO model BIBF1120 did not accrue VO. On the contrary, in the NV model BIBF1120 accelerated normal retinal vascularization and robustly diminished preretinal neovascularization compared to vehicle (by ~80%). The expression levels of VEGF negative regulator Dll4 and repulsive cues EphrinB2 and EphB4 mRNA in the retina of vehicle-treated OIR animals were markedly increased compared to normoxia, but were normalized by BIBF1120. CONCLUSIONS: Data reveal efficacy of BIBF1120 on preretinal neovascularization and, of greater interest, on acceleration of normal vascularization, consistent with interference of major repulsive cues expressed in the retina during OIR. Accordingly, BIBF1120 appears to exhibit preferable properties compared to anti-VEGF therapies for the treatment of ischemic retinopathies.


Sujet(s)
Indoles/usage thérapeutique , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Néovascularisation rétinienne/traitement médicamenteux , Rétinopathie du prématuré/traitement médicamenteux , Animaux , Animaux nouveau-nés , Modèles animaux de maladie humaine , Femelle , Rats , Rat Sprague-Dawley , Néovascularisation rétinienne/anatomopathologie , Rétinopathie du prématuré/anatomopathologie
7.
J Immunol ; 195(7): 3402-15, 2015 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-26304990

RÉSUMÉ

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.


Sujet(s)
Inflammation/immunologie , JNK Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs , Peptides/pharmacologie , Naissance prématurée/prévention et contrôle , p38 Mitogen-Activated Protein Kinases/antagonistes et inhibiteurs , Animaux , Lignée cellulaire , Femelle , Interleukine-1 bêta/immunologie , Souris , Myomètre/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Grossesse , Récepteurs à l'interleukine-1/antagonistes et inhibiteurs , Récepteur de type Toll-2/immunologie , Récepteur de type Toll-4/immunologie , Utérus/immunologie , Protéines G rho/antagonistes et inhibiteurs , rho-Associated Kinases/antagonistes et inhibiteurs
8.
Am J Pathol ; 185(2): 581-95, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25478809

RÉSUMÉ

Ischemic retinopathies are characterized by sequential vaso-obliteration followed by abnormal intravitreal neovascularization predisposing patients to retinal detachment and blindness. Ischemic retinopathies are associated with robust inflammation that leads to generation of IL-1ß, which causes vascular degeneration and impairs retinal revascularization in part through the liberation of repulsive guidance cue semaphorin 3A (Sema3A). However, retinal revascularization begins as inflammation culminates in ischemic retinopathies. Because inflammation leads to activation of proteases involved in the formation of vasculature, we hypothesized that proteinase-activated receptor (Par)-2 (official name F2rl1) may modulate deleterious effects of IL-1ß. Par2, detected mostly in retinal ganglion cells, was up-regulated in oxygen-induced retinopathy. Surprisingly, oxygen-induced retinopathy-induced vaso-obliteration and neovascularization were unaltered in Par2 knockout mice, suggesting compensatory mechanisms. We therefore conditionally knocked down retinal Par2 with shRNA-Par2-encoded lentivirus. Par2 knockdown interfered with normal revascularization, resulting in pronounced intravitreal neovascularization; conversely, the Par2 agonist peptide (SLIGRL) accelerated normal revascularization. In vitro and in vivo exploration of mechanisms revealed that IL-1ß induced Par2 expression, which in turn down-regulated sequentially IL-1 receptor type I and Sema3A expression through Erk/Jnk-dependent processes. Collectively, our findings unveil an important mechanism by which IL-1ß regulates its own endothelial cytotoxic actions by augmenting neuronal Par2 expression to repress sequentially IL-1 receptor type I and Sema3A expression. Timely activation of Par2 may be a promising therapeutic avenue in ischemic retinopathies.


Sujet(s)
Protéines de l'oeil/métabolisme , Ischémie/métabolisme , Récepteurs à la thrombine/métabolisme , Rétinopathies/métabolisme , Neurones rétiniens/métabolisme , Animaux , Protéines de l'oeil/agonistes , Protéines de l'oeil/génétique , Inflammation/traitement médicamenteux , Inflammation/génétique , Inflammation/métabolisme , Inflammation/anatomopathologie , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Ischémie/traitement médicamenteux , Ischémie/génétique , Ischémie/anatomopathologie , Souris , Souris knockout , Oligopeptides/pharmacologie , Récepteurs à la thrombine/agonistes , Récepteurs à la thrombine/génétique , Rétinopathies/traitement médicamenteux , Rétinopathies/génétique , Rétinopathies/anatomopathologie , Neurones rétiniens/anatomopathologie , Sémaphorine-3A/génétique , Sémaphorine-3A/métabolisme
9.
Arterioscler Thromb Vasc Biol ; 34(2): 285-93, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-24285580

RÉSUMÉ

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.


Sujet(s)
Cortex cérébral/vascularisation , Cortex cérébral/effets des médicaments et des substances chimiques , Infarctus cérébral/traitement médicamenteux , Hypoxie-ischémie du cerveau/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Récepteurs couplés aux protéines G/agonistes , Acide succinique/pharmacologie , Protéines angiogéniques/métabolisme , Animaux , Animaux nouveau-nés , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Lignée cellulaire , Cortex cérébral/métabolisme , Cortex cérébral/anatomopathologie , Infarctus cérébral/étiologie , Infarctus cérébral/génétique , Infarctus cérébral/métabolisme , Infarctus cérébral/anatomopathologie , Infarctus cérébral/physiopathologie , Inhibiteurs des cyclooxygénases/pharmacologie , Dinoprostone/métabolisme , Modèles animaux de maladie humaine , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Hypoxie-ischémie du cerveau/étiologie , Hypoxie-ischémie du cerveau/génétique , Hypoxie-ischémie du cerveau/métabolisme , Hypoxie-ischémie du cerveau/anatomopathologie , Hypoxie-ischémie du cerveau/physiopathologie , Injections ventriculaires , Souris , Souris de lignée C57BL , Souris knockout , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/anatomopathologie , Neuroprotecteurs/administration et posologie , Neuroprotecteurs/métabolisme , Antagonistes des prostaglandines/pharmacologie , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Sous-type EP4 des récepteurs des prostaglandines E/effets des médicaments et des substances chimiques , Sous-type EP4 des récepteurs des prostaglandines E/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Acide succinique/administration et posologie , Acide succinique/métabolisme , Facteurs temps , Techniques de culture de tissus
10.
Arterioscler Thromb Vasc Biol ; 33(8): 1881-91, 2013 Aug.
Article de Anglais | MEDLINE | ID: mdl-23766263

RÉSUMÉ

OBJECTIVE: Proinflammatory cytokines contribute to the development of retinal vasculopathies. However, the role of these factors and the mechanisms by which they elicit their effects in retina are not known. We investigated whether activated microglia during early stages of ischemic retinopathy produces excessive interleukin-1ß (IL-1ß), which elicits retinal microvascular degeneration not directly but rather by triggering the release of the proapoptotic/repulsive factor semaphorin-3A (Sema3A) from neurons. APPROACH AND RESULTS: Sprague Dawley rats subjected to retinopathy induced by hyperoxia (80% O2; O2-induced retinopathy) exhibited retinal vaso-obliteration associated with microglial activation, NLRP3 upregulation, and IL-1ß and Sema3A release; IL-1ß was mostly generated by microglia. Intraperitoneal administration of IL-1 receptor antagonists (Kineret, or rytvela [101.10]) decreased these effects and enhanced retinal revascularization; knockdown of Sema3A resulted in microvessel preservation and, conversely, administration of IL-1ß caused vaso-obliteration. In vitro, IL-1ß derived from activated primary microglial cells, cultured under hyperoxia, stimulated the release of Sema3A in retinal ganglion cells-5, which in turn induced apoptosis of microvascular endothelium; antagonism of IL-1 receptor decreased microglial activation and on retinal ganglion cells-5 abolished the release of Sema3A inhibiting ensuing endothelial cell apoptosis. IL-1ß was not directly cytotoxic to endothelial cells. CONCLUSIONS: Our findings suggest that in the early stages of O2-induced retinopathy, retinal microglia are activated to produce IL-1ß, which sustains the activation of microglia and induces microvascular injury through the release of Sema3A from adjacent neurons. Interference with IL-1 receptor or Sema3A actions preserves the microvascular bed in ischemic retinopathies and, consequently, decreases ensued pathological preretinal neovascularization.


Sujet(s)
Interleukine-1 bêta/métabolisme , Ischémie/anatomopathologie , Microglie/anatomopathologie , Rétinopathies/anatomopathologie , Rétinite/anatomopathologie , Sémaphorine-3A/métabolisme , Animaux , Antirhumatismaux/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/immunologie , Protéines de transport , Cellules endothéliales/immunologie , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Techniques de knock-down de gènes , Hyperoxie/immunologie , Hyperoxie/métabolisme , Hyperoxie/anatomopathologie , Antagoniste du récepteur à l'interleukine-1/pharmacologie , Interleukine-1 bêta/antagonistes et inhibiteurs , Interleukine-1 bêta/immunologie , Ischémie/traitement médicamenteux , Ischémie/immunologie , Microcirculation/physiologie , Microglie/immunologie , Microglie/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine , Stress oxydatif/effets des médicaments et des substances chimiques , Stress oxydatif/immunologie , Peptides/pharmacologie , Culture de cellules primaires , Rats , Rat Sprague-Dawley , Récepteurs cytoplasmiques et nucléaires/immunologie , Récepteurs cytoplasmiques et nucléaires/métabolisme , Rétinopathies/traitement médicamenteux , Rétinopathies/immunologie , Cellules ganglionnaires rétiniennes/immunologie , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie , Rétinite/traitement médicamenteux , Rétinite/immunologie , Sémaphorine-3A/génétique , Sémaphorine-3A/immunologie
11.
Cell Logist ; 2(1): 28-42, 2012 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-22645708

RÉSUMÉ

Saccharomyces cerevisiae transport protein particle (TRAPP) is a family of related multisubunit complexes required for endoplasmic reticulum-to-Golgi transport (TRAPP I), endosome-to-Golgi transport (TRAPP II) or cytosol to vacuole targeting (TRAPP III). To gain insight into the relationship between these complexes, we generated random and targeted mutations in the Trs23p core subunit. Remarkably, at physiological salt concentrations only two peaks (TRAPP I and a high molecular weight peak) are detected in wild-type cells. As the salt was raised, the high molecular weight peak resolved into TRAPP II and III peaks. Deletion of a Saccharomycotina-specific domain of Trs23p resulted in destabilization of TRAPP I but had no effect on TRAPP II or III. This mutation had no observable growth phenotype, normal levels of Ypt1p-directed guanine nucleotide exchange factor activity in vivo and did not display any in vivo nor in vitro blocks in membrane traffic. Biochemical analysis indicated that TRAPP I could be produced from the TRAPP II/III peak in vitro by increasing the salt concentration. Our data suggest that the SMS domain of Trs23p is responsible for the in vitro appearance of TRAPP I in S. cerevisiae. The implications of these findings are discussed.

12.
Mol Biol Cell ; 22(12): 2083-93, 2011 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-21525244

RÉSUMÉ

TRAPP is a multisubunit tethering complex implicated in multiple vesicle trafficking steps in Saccharomyces cerevisiae and conserved throughout eukarya, including humans. Here we confirm the role of TRAPPC2L as a stable component of mammalian TRAPP and report the identification of four novel components of the complex: C4orf41, TTC-15, KIAA1012, and Bet3L. Two of the components, KIAA1012 and Bet3L, are mammalian homologues of Trs85p and Bet3p, respectively. The remaining two novel TRAPP components, C4orf41 and TTC-15, have no homologues in S. cerevisiae. With this work, human homologues of all the S. cerevisiae TRAPP proteins, with the exception of the Saccharomycotina-specific subunit Trs65p, have now been reported. Through a multidisciplinary approach, we demonstrate that the novel proteins are bona fide components of human TRAPP and implicate C4orf41 and TTC-15 (which we call TRAPPC11 and TRAPPC12, respectively) in ER-to-Golgi trafficking at a very early stage. We further present a binary interaction map for all known mammalian TRAPP components and evidence that TRAPP oligomerizes. Our data are consistent with the absence of a TRAPP I-equivalent complex in mammalian cells, suggesting that the fundamental unit of mammalian TRAPP is distinct from that characterized in S. cerevisiae.


Sujet(s)
Réticulum endoplasmique/métabolisme , Appareil de Golgi/métabolisme , Protéines du transport vésiculaire/génétique , Protéines du transport vésiculaire/métabolisme , Humains , Protéines membranaires/métabolisme , Transport des protéines , Interférence par ARN , Petit ARN interférent , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Protéines de Saccharomyces cerevisiae/génétique , Protéines de Saccharomyces cerevisiae/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE