Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Biochim Biophys Acta ; 1863(9): 2322-32, 2016 09.
Article in English | MEDLINE | ID: mdl-27321910

ABSTRACT

E-prostanoid receptor subtype 2 (EP2) agonists are currently under clinical development as hypotensive agents for the treatment of ocular hypertension. However, the effects of EP2 receptor agonists on trabecular meshwork (TM) alterations leading to primary open-angle glaucoma (POAG) are still unknown. Here, we evaluated whether EP2 receptor activation exhibits protective functions on TM cell death induced by endoplasmic reticulum (ER) stress. We show that the EP2 receptor agonist butaprost protects TM cell death mediated by the ER stress inducer tunicamycin through a cyclic AMP (cAMP)-dependent mechanism, but independent of the classical cAMP sensors, protein kinase A and exchange proteins activated by cAMP. The ER stress-induced intrinsic apoptosis inhibited by the EP2 receptor agonist was correlated with a decreased accumulation of the cellular stress sensor p53. In addition, p53 down-regulation was associated with inhibition of its transcriptional activity, which led to decreased expression of the pro-apoptotic p53-upregulated modulator of apoptosis (PUMA). The stabilization of p53 by nutlin-3a abolished butaprost-mediated cell death protection. In conclusion, we showed that EP2 receptor activation protects against ER stress-dependent mitochondrial apoptosis through down-regulation of p53. The specific inhibition of this pathway could reduce TM alterations observed in POAG patients.


Subject(s)
Apoptosis , Cytoprotection , Down-Regulation , Endoplasmic Reticulum Stress , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Signal Transduction , Trabecular Meshwork/pathology , Tumor Suppressor Protein p53/metabolism , Adult , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Cell Death/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytochromes c/metabolism , Cytoprotection/drug effects , Down-Regulation/drug effects , Endoplasmic Reticulum Chaperone BiP , Guanine Nucleotide Exchange Factors/metabolism , Heat-Shock Proteins/metabolism , Humans , Male , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Transcription Factor CHOP/metabolism , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects
2.
Nat Commun ; 14(1): 3728, 2023 06 22.
Article in English | MEDLINE | ID: mdl-37349339

ABSTRACT

Loss of NBEAL2 function leads to grey platelet syndrome (GPS), a bleeding disorder characterized by macro-thrombocytopenia and α-granule-deficient platelets. A proportion of patients with GPS develop autoimmunity through an unknown mechanism, which might be related to the proteins NBEAL2 interacts with, specifically in immune cells. Here we show a comprehensive interactome of NBEAL2 in primary T cells, based on mass spectrometry identification of altogether 74 protein association partners. These include LRBA, a member of the same BEACH domain family as NBEAL2, recessive mutations of which cause autoimmunity and lymphocytic infiltration through defective CTLA-4 trafficking. Investigating the potential association between NBEAL2 and CTLA-4 signalling suggested by the mass spectrometry results, we confirm by co-immunoprecipitation that CTLA-4 and NBEAL2 interact with each other. Interestingly, NBEAL2 deficiency leads to low CTLA-4 expression in patient-derived effector T cells, while their regulatory T cells appear unaffected. Knocking-down NBEAL2 in healthy primary T cells recapitulates the low CTLA-4 expression observed in the T cells of GPS patients. Our results thus show that NBEAL2 is involved in the regulation of CTLA-4 expression in conventional T cells and provide a rationale for considering CTLA-4-immunoglobulin therapy in patients with GPS and autoimmune disease.


Subject(s)
Gray Platelet Syndrome , Humans , Adaptor Proteins, Signal Transducing/metabolism , Blood Platelets/metabolism , Blood Proteins/genetics , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Gray Platelet Syndrome/genetics , Gray Platelet Syndrome/metabolism
3.
Invest Ophthalmol Vis Sci ; 57(4): 1816-25, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27082296

ABSTRACT

PURPOSE: Prostaglandin F2α analogues are the first-line medication for the treatment of ocular hypertension (OHT), and prostanoid EP2 receptor agonists are under clinical development for this indication. The goal of this study was to investigate the effects of F prostanoid (FP) and EP2 receptor activation on the myofibroblast transition of primary trabecular meshwork (TM) cells, which could be a causal mechanism of TM dysfunction in glaucoma. METHODS: Human primary TM cells were treated with either latanoprost or butaprost and TGF-ß2. Trabecular meshwork contraction was measured in a three-dimensional (3D) TM cell-populated collagen gel (CPCG) model. Expression of α-smooth muscle actin (α-SMA) and phosphorylation of myosin light chain (MLC) were determined by Western blot. Assembly of actin stress fibers and collagen deposition were evaluated by immunocytochemistry. Involvement of p38, extracellular signal-regulated kinase (ERK), and Rho-associated kinase (ROCK) pathways as well as matrix metalloproteinase activation was tested with specific inhibitors. RESULTS: In one source of validated adult TM cells, latanoprost induced cell contraction as observed by CPCG surface reduction and increased actin polymerization, α-SMA expression, and MLC phosphorylation, whereas butaprost inhibited TGF-ß2-induced CPCG contraction, actin polymerization, and MLC phosphorylation. Both agonists inhibited TGF-ß2-dependent collagen deposition. The latanoprost effects were mediated by p38 pathway. CONCLUSIONS: Latanoprost decreased TM collagen accumulation but promoted a contractile phenotype in a source of adult TM cells that could modulate the conventional outflow pathway. In contrast, butaprost attenuated both TM contraction and collagen deposition induced by TGF-ß2, thereby inhibiting myofibroblast transition of TM cells. These results open new perspectives for the management of OHT.


Subject(s)
Glaucoma/drug therapy , Myofibroblasts/metabolism , Prostaglandins F, Synthetic/pharmacology , RNA/genetics , Receptors, Prostaglandin E, EP2 Subtype/drug effects , Receptors, Prostaglandin/drug effects , Trabecular Meshwork/metabolism , Actins/biosynthesis , Actins/genetics , Adult , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Antihypertensive Agents/pharmacology , Blotting, Western , Cell Survival , Cells, Cultured , Dinoprost , Glaucoma/genetics , Glaucoma/metabolism , Humans , Immunohistochemistry , Latanoprost , Male , Myofibroblasts/drug effects , Myofibroblasts/pathology , Myosin Light Chains/metabolism , Neuroprotective Agents , Prostaglandins E, Synthetic , Rats , Real-Time Polymerase Chain Reaction , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Signal Transduction/drug effects , Trabecular Meshwork/drug effects , Trabecular Meshwork/pathology
SELECTION OF CITATIONS
SEARCH DETAIL