Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 49
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Biochem Biophys Rep ; 38: 101672, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38434142

RÉSUMÉ

Autophagy has emerged as a critical innate immune mechanism for host elimination of intracellular pathogens, however, the role of the autophagy receptor Optineurin during mycobacterial infection is not fully understood. To address this lacuna, we infected bone marrow-derived macrophages (BMDMs) derived from Optn+/+ and Optn-/- mice with Mycobacterium smegmatis, and observed the infection outcome at sequential time points. While low multiplicity of infection (MOI) did not show any significant difference between BMDMs from the two groups, at high MOI Optn-/- mice-derived BMDMs showed significantly lower colony forming unit counts, as well as lower cell counts at 12 h and 24 h post-infection. Quantification of cell numbers and nuclear morphologies at various time points post-infection indicated a markedly higher cell death in the Optineurin-deficient macrophages. Optineurin-deficient BMDMs showed significantly lower levels of the autophagosomal protein LC3-II upon infection, indicating a potential role for Optineurin in regulating autophagy during mycobacterial infection. Moreover, when stimulated by bacterial LPS, Optineurin deficient macrophages, showed altered levels of the inflammatory cytokine pro-IL-1ß. These observations taken together suggest a novel regulatory role for Optineurin during mycobacterial infection. Its deficiency leads to an impairment in macrophage responses, directly impacting the pathophysiology of infection.

2.
FEBS J ; 290(12): 3110-3127, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36705529

RÉSUMÉ

Optineurin/OPTN polymorphism, M98K is associated with normal tension glaucoma in certain populations, and genetic evidence shows its interaction with tumour necrosis factor-alpha (TNFα) polymorphism in causing glaucoma. Endoplasmic reticulum (ER) stress is also associated with glaucoma. We hypothesized that M98K-OPTN may sensitize retinal ganglion cells to various types of stress. To test this hypothesis, stable clones of a retinal cell line, 661W, expressing either wild-type (WT)-OPTN or M98K-OPTN were generated and examined for their survival under various stress conditions. Compared with WT-OPTN expressing cells, M98K-OPTN expressing cells showed significantly lower cell survival and higher activation of caspase-3 and caspase-8 upon treatment with tunicamycin (an inducer of ER stress) or TNFα. Levels of ER stress sensors IRE1α, PERK and ATF6 were significantly higher in M98K-OPTN expressing cells. Tunicamycin treatment resulted in significantly higher induction of ER stress marker CHOP and several other ER stress response genes regulated by IRE1α-XBP1, PERK-ATF4 and ATF6 pathways, in M98K-OPTN expressing cells. Splicing of XBP1 and ATF6 activation was higher in tunicamycin-treated M98K-OPTN expressing cells. Increased levels of PERK and IRE1α proteins in M98K-OPTN expressing cells were dependent on autophagy. Overall, our results show that M98K-OPTN sensitizes retinal cells to TNFα and ER stress-induced cell death. We also show that M98K-OPTN alters ER stress response signalling, which possibly enhances the sensitivity of retinal cells to ER stress. Our results provide support to the hypothesis that M98K-OPTN may cooperate with other genetic or environmental factors to cause retinal ganglion cell death associated with glaucoma.


Sujet(s)
Glaucome , Protein-Serine-Threonine Kinases , Humains , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Endoribonucleases/génétique , Endoribonucleases/métabolisme , Tunicamycine/pharmacologie , Glaucome/génétique , Stress du réticulum endoplasmique/génétique , Cellules ganglionnaires rétiniennes/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie
3.
Prog Mol Biol Transl Sci ; 194: 67-78, 2023.
Article de Anglais | MEDLINE | ID: mdl-36631201

RÉSUMÉ

Transferrin receptor (TFRC) is a transmembrane protein that plays a crucial role in mediating homeostasis of iron in the cell. The binding of transferrin (that is bound to iron) to TFRC at the cell membrane generally starts endocytosis of TFRC-transferrin complex, which leads to formation of vesicles that are positive for TFRC. These vesicles travel to the early endosomes and later to the endocytic recycling compartment. Release of iron occurs in the early endosomes because of acidic pH. Major fraction of the transferrin and TFRC is transported back to the cell membrane; however, a minor fraction of it is transported to lysosomes through the process of autophagy. Optineurin (OPTN) is a multi-functional adaptor protein that plays a pivotal role in the control of TFRC trafficking, recycling and autophagy dependent degradation. Optineurin also plays a role in cargo-selective and non-selective autophagy. Here, we review our understanding of the function of OPTN in regulating TFRC trafficking, recycling and autophagy dependent degradation. We also discuss the mechanisms by which certain disease-associated mutations of OPTN alter these processes.


Sujet(s)
Protéines du cycle cellulaire , Endocytose , Protéines de transport membranaire , Récepteurs à la transferrine , Humains , Transport biologique , Protéines du cycle cellulaire/génétique , Endocytose/génétique , Endosomes/métabolisme , Fer/métabolisme , Protéines de transport membranaire/génétique , Transport des protéines/génétique , Récepteurs à la transferrine/métabolisme , Transferrines/métabolisme
4.
Biochem Biophys Res Commun ; 641: 42-49, 2023 01 22.
Article de Anglais | MEDLINE | ID: mdl-36521284

RÉSUMÉ

NLRP3 is a cytoplasmic receptor protein, which initiates caspase-1 mediated inflammatory immune response upon detection of invading pathogen or a wide array of internal distress signals. Several gain-of function mutations of NLRP3 cause hereditary disorder of cold-induced hyper-inflammation known as familial cold autoinflammatory syndrome-1 (FCAS1). Although, caspase-1 activation and downstream interleukin-1ß/interleukin-18 maturation are common effectors in pathophysiology of this disorder, molecular mechanisms of how exposure to subnormal temperature triggers mutant NLRP3-inflammsome activity is not understood. Here, we show that endogenous NLRP3 is in complex with HSC70 (HSPA8), and this interaction is reduced upon exposure to cold. FCAS-causing NLRP3-L353P and NLRP3-R260W mutants show enhanced interaction with HSC70. Upon exposure to subnormal temperature, NLRP3-L353P and NLRP3-R260W show enhanced inflammasome formation, increased caspase-1 activation and reduced interaction with HSC70. Knockdown of HSC70 results in increased inflammasome formation by L353P and R260W mutants of NLRP3. Our results suggest that interaction with HSC70 suppresses inflammasome formation by FCAS-causing NLRP3 mutants at physiological temperature, and loss of this inhibitory association at subnormal temperature causes aggravated inflammasome formation and caspase-1 activation leading to interleukin-1ß maturation. These results provide evidence for HSC70 being a cold-sensor and a temperature-dependent regulator of inflammatory signaling by FCAS-causing NLRP3 mutants.


Sujet(s)
Syndromes périodiques associés à la cryopyrine , Inflammasomes , Humains , Caspase-1/métabolisme , Syndromes périodiques associés à la cryopyrine/génétique , Inflammasomes/métabolisme , Interleukine-1 bêta/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéines du choc thermique HSC70
6.
J Biochem ; 171(5): 555-565, 2022 May 11.
Article de Anglais | MEDLINE | ID: mdl-35134975

RÉSUMÉ

An important feature of several neurodegenerative diseases is the formation of pathological structures containing aggregated proteins. The autophagy receptor optineurin/OPTN is frequently observed in these structures. The role played by optineurin in these aggregates is not clear. In this study, we explored whether optineurin has a cytoprotective role in the cells having mutant protein aggregates. We overexpressed mutant huntingtin having 97 glutamine repeats (mHtt) and mutant ataxin-3 having 130 glutamine repeats (mAtax-3) in wild-type and optineurin-deficient neuronal (N2A) and non-neuronal cells (Optn-/- mouse embryonic fibroblasts) and determined the percentage of dead cells with mutant protein aggregates. Optineurin-deficient cells having mHtt or mAtax-3 aggregates showed higher cell death as compared to wild-type cells having mutant protein aggregates. Confocal microscopy revealed that optineurin formed a shell around mHtt and mAtax-3 aggregates through its C-terminal domain. The C-terminal domain of optineurin, which lacks LC3-interacting region required for autophagy, was necessary and sufficient to reduce cytotoxicity of mHtt and mAtax-3 aggregates. Our results show that in the absence of optineurin, mutant protein aggregates are highly toxic, revealing an autophagy-independent cytoprotective function of optineurin, which is mediated by its C-terminal domain.


Sujet(s)
Cytoprotection , Agrégats de protéines , Animaux , Ataxine-3/génétique , Autophagie/physiologie , Fibroblastes , Glutamine , Souris , Protéines mutantes
7.
FEBS J ; 289(24): 8037-8049, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-34535969

RÉSUMÉ

Familial cold autoinflammatory syndrome (FCAS) is a subset of heritable autoinflammatory disorders wherein inflammatory symptoms aggravate upon exposure of the individual to subnormal temperature. In the past two decades, several mutations in various genes such as NLRP3, NLRP12, PLCG2 and NLRC4 have been identified that cause cold-triggered inflammation. However, our understanding of the mechanisms by which cells perceive subnormal temperature, and what keeps the inflammation under check until exposure to low temperature, is very limited. We hypothesise that recognition of FCAS-associated mutants as misfolded polypeptides by temperature-sensitive HSC70 (HSPA8) chaperone determines the FCAS phenotype. At 37 °C, HSC70 would interact with the mutant proteins, keeping them almost inactive, and loss of interaction at low temperature due to a conformational change in HSC70 would lead to their activation. The proposed mechanism of low temperature sensing in the context of FCAS may have wider implications for HSC70 as a cold temperature sensor in various pathological conditions where symptoms get aggravated upon exposure to low temperature.


Sujet(s)
Basse température , Syndromes périodiques associés à la cryopyrine , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine , Température , Syndromes périodiques associés à la cryopyrine/génétique , Inflammation
8.
Neurochem Int ; 148: 105075, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34023378

RÉSUMÉ

Studies carried out on the pathogenesis of glaucoma using murine cell lines and animal models require to be validated in human cells. Therefore, we explored the possibility of using human primary retinal cells (hPRCs) in culture as a model for molecular studies and testing of potential therapeutic drugs. For this purpose, central retinal tissue, obtained from the enucleated eyes of patients with anterior staphyloma, was digested with trypsin and grown in a medium containing supplements (basic fibroblast growth factor and fetal bovine serum). hPRCs at passage 1 and 2, show expression of either GFAP, a glial cell marker, or ß-III tubulin, a retinal ganglion cell (RGC)-specific marker. But at passages 3-5 nearly all of hPRCs express several RGC-specific markers (Brn3 proteins, Thy-1, ß-III tubulin, RBPMS and NeuN) but not GFAP. Expression of these markers indicated that these cells may have functional properties of RGCs. As RGCs are sensitive to glaucoma-associated mutants of OPTN, we analysed the survival of hPRCs upon overexpression of OPTN mutants. Glaucoma-associated mutants, E50K-OPTN and M98K-OPTN, induced significantly higher cell death in hPRCs compared to WT-OPTN, whereas an amyotrophic lateral sclerosis-associated mutant, E478G-OPTN, did not. TBK1 inhibitor Amlexanox protected hPRCs from E50K-OPTN and M98K-OPTN induced cell death. M98K-OPTN induced cell death was suppressed by inhibitors of CaMKKß and AMPK in hPRCs as well as in 661W, a mouse cell line that expresses several markers of RGCs and RGC precursor cells. Our results suggest that hPRCs under appropriate culture condition show RGC-like properties. These cells can be used to explore the molecular mechanisms of cell death relevant for glaucoma pathogenesis and for testing of cytoprotective compounds.


Sujet(s)
Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Glaucome/métabolisme , Protéines de transport membranaire/génétique , Protéines de transport membranaire/métabolisme , Mutation/génétique , Cellules ganglionnaires rétiniennes/métabolisme , Apoptose/génétique , Apoptose/physiologie , Glaucome/anatomopathologie , Humains , Rétine/métabolisme , Rétine/anatomopathologie , Cellules ganglionnaires rétiniennes/anatomopathologie , Transduction du signal/génétique
9.
FEBS J ; 288(15): 4576-4595, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33548116

RÉSUMÉ

Mutations in OPTN are associated with glaucoma, an eye disease, and also with amyotrophic lateral sclerosis (ALS), a motor neuron disease. A 2-bp insertion in OPTN (691_692insAG or 2bpIns-OPTN) is associated with both glaucoma and ALS. This mutation results in frame shift after 127 amino acids, giving rise to a protein with C-terminal aberrant sequence. We have explored the mechanism of induction of cell death by this mutant in a motor neuron cell line, NSC-34, and also in a retinal cell line, 661W. Compared to wild-type OPTN, this mutant induced more cell death in NSC-34 and 661W cells. This mutant localizes predominantly in the nucleus whereas normal OPTN localizes in the cytoplasm. Deletion analysis of 2bpIns-OPTN showed that the aberrant sequence was not essential for cell death induction. This mutant interacts with TANK-binding kinase 1 (Tbk1) but not with OPTN and activates Tbk1. This mutant induced ER stress in NSC-34 cells as seen by induction of C/EBP homologous protein (CHOP) and some other genes. Induction of CHOP, autophagosomal protein LC3-II and cell death by this mutant were abrogated by Tbk1 knockdown and also by 4-phenylbutyric acid, that inhibits ER stress. Induction of CHOP and cell death by 2bpIns-OPTN was autophagy dependent as shown by the effect of Atg5 knockdown. This mutant caused increased formation of LC3-positive aggregates. Treatment of cells with autophagy inducer rapamycin reduced LC3-positive aggregates, CHOP and cell death induced by 2bpIns-OPTN. These results suggest that constitutive activation of Tbk1 by 2bpIns-OPTN leads to impaired autophagy that results in ER stress and cell death.


Sujet(s)
Sclérose latérale amyotrophique/génétique , Apoptose , Protéines du cycle cellulaire/génétique , Glaucome/génétique , Protéines de transport membranaire/génétique , Mutation , Neurones/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Autophagie , Protéine-5 associée à l'autophagie/métabolisme , Protéines du cycle cellulaire/métabolisme , Stress du réticulum endoplasmique , Cellules HEK293 , Humains , Protéines de transport membranaire/métabolisme , Protéines associées aux microtubules/métabolisme , Liaison aux protéines , Facteur de transcription CHOP/métabolisme
10.
Biochem Biophys Res Commun ; 534: 297-302, 2021 01 01.
Article de Anglais | MEDLINE | ID: mdl-33272572

RÉSUMÉ

We have investigated the physiological role of the autophagy receptor Optineurin/Optn in endoplasmic reticulum (ER) stress response using cellular and animal models. In comparison to their normal counterparts, Optn-deficient mouse embryonic fibroblasts showed significantly higher cell death and caspase-3 activation upon treatment with tunicamycin and thapsigargin, inducers of ER stress. The transcript levels of some of the genes regulated by the IRE1-XBP1 and PERK-ATF4 pathways were upregulated in Optn-deficient cells, in comparison with normal cells, upon treatment with tunicamycin, and also in the brain cortex and liver of tunicamycin treated Optn-deficient mice. Also, the basal levels of IRE1α and PERK were higher in Optn-deficient cells. These results suggest that Optn modulates ER stress-induced signaling pathways and provides protection from ER stress-induced cell death.


Sujet(s)
Protéines du cycle cellulaire/physiologie , Stress du réticulum endoplasmique/physiologie , Protéines de transport membranaire/physiologie , Animaux , Autophagie/physiologie , Protéines du cycle cellulaire/déficit , Protéines du cycle cellulaire/génétique , Mort cellulaire/physiologie , Cellules cultivées , Stress du réticulum endoplasmique/génétique , Endoribonucleases/génétique , Endoribonucleases/métabolisme , Fibroblastes/cytologie , Fibroblastes/physiologie , Expression des gènes , Protéines de transport membranaire/déficit , Protéines de transport membranaire/génétique , Souris , Souris knockout , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Transduction du signal/physiologie , Réponse aux protéines mal repliées/génétique , eIF-2 Kinase/génétique , eIF-2 Kinase/métabolisme
11.
Proc Natl Acad Sci U S A ; 116(43): 21694-21703, 2019 10 22.
Article de Anglais | MEDLINE | ID: mdl-31597739

RÉSUMÉ

NLRC4 [nucleotide-binding domain and leucine-rich repeat (NLR) family, caspase recruitment domain (CARD) containing 4] is an innate immune receptor, which, upon detection of certain pathogens or internal distress signals, initiates caspase-1-mediated interleukin-1ß maturation and an inflammatory response. A gain-of-function mutation, H443P in NLRC4, causes familial cold autoinflammatory syndrome (FCAS) characterized by cold-induced hyperactivation of caspase-1, enhanced interleukin-1ß maturation, and inflammation. Although the H443P mutant shows constitutive activity, the mechanism involved in hyperactivation of caspase-1 by NLRC4-H443P upon exposure of cells to lower temperature is not known. Here, we show that heat shock cognate protein 70 (HSC70) complexes with NLRC4 and negatively regulates caspase-1 activation by NLRC4-H443P in human cells. Compared with NLRC4, the structurally altered NLRC4-H443P shows enhanced interaction with HSC70. Nucleotide binding- and leucine-rich repeat domains of NLRC4, but not its CARD, can engage in complex formation with HSC70. Knockdown of HSC70 enhances apoptosis-associated speck-like protein containing a CARD (ASC)-speck formation and caspase-1 activation by NLRC4-H443P. Exposure to subnormal temperature results in reduced interaction of NLRC4-H443P with HSC70, and an increase in its ability to form ASC specks and activate caspase-1. Unlike the NLRC4-H443P mutant, another constitutively active mutant (NLRC4-V341A) associated with autoinflammatory diseases, but not FCAS, showed neither enhanced interaction with HSC70 nor an increase in inflammasome formation upon exposure to subnormal temperature. Our results identify HSC70 as a negative regulator of caspase-1 activation by the temperature-sensitive NLRC4-H443P mutant. We also show that low-temperature-induced hyperactivation of caspase-1 by NLRC4-H443P is due to loss of inhibition by HSC70.


Sujet(s)
Protéines adaptatrices de signalisation CARD/génétique , Protéines de liaison au calcium/génétique , Caspase-1/immunologie , Syndromes périodiques associés à la cryopyrine/génétique , Protéines du choc thermique HSC70/métabolisme , Apoptose/génétique , Apoptose/immunologie , Protéines adaptatrices de signalisation CARD/métabolisme , Lignée cellulaire , Basse température , Activation enzymatique/génétique , Mutation gain de fonction/génétique , Cellules HEK293 , Protéines du choc thermique HSC70/génétique , Humains , Inflammation/immunologie , Interleukine-1 bêta/immunologie
12.
Biochim Biophys Acta Mol Cell Res ; 1865(11 Pt A): 1526-1538, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-30327196

RÉSUMÉ

Optineurin (Optn) is an autophagy receptor that performs various functions in cargo-selective and non-selective autophagy. Here, we have identified and characterized a splice variant of mouse optineurin mRNA, which produces a truncated protein lacking N-terminal 157 amino acids (d157mOptn). This mRNA and protein are expressed in several tissues and cells. d157mOptn has an intact LC3-interacting region and a serine (S187) in it. However, unlike normal optineurin, the d157mOptn was not phosphorylated at this site when expressed in mammalian cells, and showed reduced interaction with TBK1 (tank binding kinase) that mediates phosphorylation at S187 (S177 in human OPTN). This phosphorylation of Optn required intact N-terminal sequence as well as functional C-terminal ubiquitin-binding domain. Unlike normal optineurin, d157mOptn was unable to promote autophagosome and autolysosome formation upon expression in Optn-deficient cells. d157mOptn was recruited to mutant huntingtin aggregates, but unlike wild type optineurin, it was unable to clear these aggregates by autophagy in neuronal NSC-34 cells. Phospho-TBK1 was seen around mutant Huntingtin aggregates in Optn overexpressing cells but it was reduced in cells overexpressing d157mOptn. Thus, we have identified an isoform of mouse optineurin which is defective in cargo-selective and non-selective autophagy possibly due to loss of phosphorylation and impaired interaction with TBK1. This isoform, which inhibits autophagosome formation in neuronal cells, might be involved in selectively modulating some of the functions of Optn, such as autophagy. Our results provide an insight into the role of N-terminal domain of Optn in various autophagic functions.


Sujet(s)
Autophagie/génétique , Épissage des ARN , Facteur de transcription TFIIIA/génétique , Facteur de transcription TFIIIA/métabolisme , Animaux , Autophagosomes/métabolisme , Marqueurs biologiques , Protéines du cycle cellulaire , Lignée cellulaire , Fibroblastes , Humains , Protéine huntingtine/métabolisme , Immunohistochimie , Protéines de transport membranaire , Souris , Souris knockout , Mutation , Neurones/métabolisme , Phosphorylation , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme
13.
Commun Integr Biol ; 11(2): 1-4, 2018.
Article de Anglais | MEDLINE | ID: mdl-30083281

RÉSUMÉ

Autophagy is an essential physiological process that maintains cellular homeostasis by eliminating harmful protein aggregates, damaged organelles and certain pathogens through lysosomal degradation. During autophagy specialized structures, known as autophagosomes are formed that recruit the cargo through autophagy receptors, and deliver it to lysosomes. Optineurin (Optn) is an autophagy receptor that mediates cargo selective autophagy. Recently, we have identified a novel function of Optn that promotes autophagosome formation during non-selective autophagy. Optn-deficient cells show reduced formation of autophagosomal protein LC3-II and lower number of autophagosomes as well as autolysosomes. Interestingly, formation of phagophores is increased in Optn-deficient cells. This suggests that Optn promotes autophagosome formation by potentiating LC3-II production and phagophore maturation. Phosphorylation of Optn at Ser-177 is required for promoting autophagosome formation. Here, we discuss various aspects of the role of Optn in the formation of autophagosomes and Atg16L1-positive vesicles. We also discuss the potential role of Rab1a-Optn interaction.

14.
Front Immunol ; 9: 1287, 2018.
Article de Anglais | MEDLINE | ID: mdl-29951055

RÉSUMÉ

Optineurin (OPTN) is an adaptor protein that is involved in mediating a variety of cellular processes such as signaling, vesicle trafficking, and autophagy. Certain mutations in OPTN (gene OPTN) are associated with primary open angle glaucoma, a leading cause of irreversible blindness, and amyotrophic lateral sclerosis, a fatal motor neuron disease. Glaucoma-associated mutations of OPTN are mostly missense mutations. OPTN mediates its functions by interacting with various proteins and altered interactions of OPTN mutants with various proteins primarily contribute to functional defects. It interacts with Rab8, myosin VI, Huntigtin, TBC1D17, and transferrin receptor to mediate various membrane vesicle trafficking pathways. It is an autophagy receptor that mediates cargo-selective as well as non-selective autophagy. Glaucoma-associated mutants of OPTN, E50K, and M98K, cause defective vesicle trafficking, autophagy, and signaling that contribute to death of retinal ganglion cells (RGCs). Transgenic mice expressing E50K-OPTN show loss of RGCs and persistent reactive gliosis. TBK1 protein kinase, which mediates E50K-OPTN and M98K-OPTN induced cell death, is emerging as a potential drug target. Autoimmunity has been implicated in glaucoma but involvement of OPTN or its mutants in autoimmnity has not been explored. In this review, we highlight the main functions of OPTN and how glaucoma-associated mutants alter these functions. We also discuss some of the controversies, such as the role of OPTN in signaling to transcription factor NF-κB, interferon signaling, and use of RGC-5 cell line as a cell culture model.


Sujet(s)
Études d'associations génétiques , Prédisposition génétique à une maladie , Glaucome/génétique , Glaucome/métabolisme , Mutation , Facteur de transcription TFIIIA/génétique , Facteur de transcription TFIIIA/métabolisme , Allèles , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/métabolisme , Animaux , Auto-immunité , Autophagie/génétique , Protéines du cycle cellulaire , Modèles animaux de maladie humaine , Glaucome/immunologie , Humains , Protéines de transport membranaire , Facteur de transcription NF-kappa B/métabolisme , Transport des protéines , Cellules ganglionnaires rétiniennes/métabolisme , Transduction du signal
15.
J Biol Chem ; 293(1): 132-147, 2018 01 05.
Article de Anglais | MEDLINE | ID: mdl-29133525

RÉSUMÉ

Autophagy is a quality-control mechanism that helps to maintain cellular homeostasis by removing damaged proteins and organelles through lysosomal degradation. During autophagy, signaling events lead to the formation of a cup-shaped structure called the phagophore that matures into the autophagosome. Recruitment of the autophagy-associated Atg12-5-16L1 complex to Wipi2-positive phagophores is crucial for producing microtubule-associated protein 1 light chain 3-II (LC3-II), which is required for autophagosome formation. Here, we explored the role of the autophagy receptor optineurin (Optn) in autophagosome formation. Fibroblasts from Optn knock-out mouse showed reduced LC3-II formation and a lower number of autophagosomes and autolysosomes during both basal and starvation-induced autophagy. However, the number of Wipi2-positive phagophores was not decreased in Optn-deficient cells. We also found that the number of Atg12/16L1-positive puncta and recruitment of the Atg12-5-16L1 complex to Wipi2-positive puncta are reduced in Optn-deficient cells. Of note, Optn was recruited to Atg12-5-16L1-positive puncta, and interacted with Atg5 and also with Atg12-5 conjugate. A disease-associated Optn mutant, E478G, defective in ubiquitin binding, was also defective in autophagosome formation and recruitment to the Atg12-5-16L1-positive puncta. Moreover, we noted that Optn phosphorylation at Ser-177 was required for autophagosome formation but not for Optn recruitment to the phagophore. These results suggest that Optn potentiates LC3-II production and maturation of the phagophore into the autophagosome, by facilitating the recruitment of the Atg12-5-16L1 complex to Wipi2-positive phagophores.


Sujet(s)
Protéines associées à l'autophagie/métabolisme , Protéines de transport/métabolisme , Protéines de l'oeil/métabolisme , Protéines membranaires/métabolisme , Facteur de transcription TFIIIA/métabolisme , Animaux , Autophagosomes/métabolisme , Autophagie/physiologie , Protéines du cycle cellulaire , Femelle , Cellules HEK293 , Humains , Mâle , Protéines de transport membranaire , Souris , Souris knockout , Protéines associées aux microtubules/métabolisme , Protéines de liaison aux phosphates , Liaison aux protéines
16.
Sci Rep ; 7(1): 16855, 2017 12 04.
Article de Anglais | MEDLINE | ID: mdl-29203899

RÉSUMÉ

A photoreceptor cell line, 661W, derived from a mouse retinal tumor that expresses several markers of cone photoreceptor cells has been described earlier. However, these cells can be differentiated into neuronal cells. Here, we report that this cell line expressed certain markers specific to retinal ganglion cells such as Rbpms, Brn3b (Pou4f2), Brn3c (Pou4f3), Thy1 and γ-synuclein (Sncg), and some other markers of neuronal cells (beta-III tubulin, NeuN and MAP2). These cells also expressed Opn1mw, a cone-specific marker and nestin, a marker for neural precursor cells. Two glaucoma-associated mutants of OPTN, E50K and M98K, but not an amyotrophic lateral sclerosis-associated mutant, E478G, induced cell death selectively in 661W cells. However, in a motor neuron cell line, NSC34, E478G mutant of OPTN but not E50K and M98K induced cell death. We conclude that 661W is a retinal ganglion precursor-like cell line, which shows properties of both retinal ganglion and photoreceptor cells. We suggest that these cells could be utilized for exploring the mechanisms of cell death induction and cytoprotection relevant for glaucoma pathogenesis. RGC-5 cell line which probably arose from 661W cells showed expression of essentially the same markers of retinal ganglion cells and neuronal cells as seen in 661W cells.


Sujet(s)
Apoptose , Protéines de l'oeil/métabolisme , Glaucome/anatomopathologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Marqueurs biologiques/métabolisme , Protéines du cycle cellulaire , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Protéines de liaison à l'ADN , Protéines de l'oeil/génétique , Glaucome/métabolisme , Protéines de transport membranaire , Souris , Protéines associées aux microtubules/métabolisme , Mutagenèse dirigée , Protéines de tissu nerveux/métabolisme , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Protéines nucléaires/métabolisme , Pyrimidines/pharmacologie , Cellules ganglionnaires rétiniennes/cytologie , Cellules ganglionnaires rétiniennes/métabolisme , Staurosporine/pharmacologie , Thiophènes/pharmacologie , Antigènes Thy-1/métabolisme , Facteur de transcription Brn-3C/métabolisme , Tubuline/métabolisme
17.
J Biol Chem ; 292(4): 1218-1230, 2017 01 27.
Article de Anglais | MEDLINE | ID: mdl-27974463

RÉSUMÉ

Nod-like receptor family card containing 4 (NLRC4)/Ipaf is involved in recognition of pathogen-associated molecular patterns leading to caspase-1 activation and cytokine release, which mediate protective innate immune response. Point mutations in NLRC4 cause autoinflammatory syndromes. Although all the mutations result in constitutive caspase-1 activation, their phenotypic presentations are different, implying that these mutations cause different alterations in properties of NLRC4. NLRC4 interacts with SUG1 and induces caspase-8-mediated cell death. Here, we show that one of the autoinflammatory syndrome-causing mutants of NLRC4, H443P, but not T337A and V341A, constitutively activates caspase-8 and induces apoptotic cell death in human lung epithelial cells. Compared with wild type NLRC4, the H443P mutant shows stronger interaction with SUG1 and with ubiquitinated cellular proteins. Phosphorylation of NLRC4 at Ser533 plays a crucial role in caspase-8 activation and cell death. However, H443P mutant does not require Ser533 phosphorylation for caspase-8 activation and cell death. Caspase-8 activation by NLRC4 and its H443P mutant are dependent on the adaptor protein FADD. A phosphomimicking mutant of NLRC4, S533D does not require SUG1 activity for inducing cell death. Ubiquitin-tagged NLRC4 could induce cell death and activate caspase-8 independent of Ser533 phosphorylation. Our work suggests that SUG1-mediated signaling results in enhanced ubiquitination and regulates FADD-dependent caspase-8 activation by NLRC4. We show that the autoinflammation-associated H443P mutant is altered in interaction with SUG1 and ubiquitinated proteins, triggering constitutive caspase-8-mediated cell death dependent on FADD but independent of Ser533 phosphorylation.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de signalisation CARD/métabolisme , Protéines de liaison au calcium/métabolisme , Caspase 8/métabolisme , Protéine à domaine de mort associée à Fas/métabolisme , Protéines à domaine LIM/métabolisme , Mutation faux-sens , Transduction du signal , Facteurs de transcription/métabolisme , Ubiquitination , Cellules A549 , ATPases associated with diverse cellular activities , Protéines adaptatrices de la transduction du signal/génétique , Substitution d'acide aminé , Protéines adaptatrices de signalisation CARD/génétique , Protéines de liaison au calcium/génétique , Caspase 8/génétique , Mort cellulaire , Activation enzymatique/génétique , Protéine à domaine de mort associée à Fas/génétique , Maladies auto-inflammatoires héréditaires/génétique , Maladies auto-inflammatoires héréditaires/métabolisme , Humains , Protéines à domaine LIM/génétique , Proteasome endopeptidase complex , Facteurs de transcription/génétique
19.
Exp Eye Res ; 144: 54-63, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26302410

RÉSUMÉ

Certain mutations in optineurin (gene OPTN) are associated with primary open angle glaucoma. Optineurin is ubiquitously expressed but it shows high level of expression in certain cells and tissues including retinal ganglion cells. It interacts with many proteins, often acting as an adaptor to link two or more proteins. These interactions play a crucial role in mediating various functions of optineurin such as membrane vesicle trafficking, autophagy, signal transduction etc. Autophagy is basically a quality control mechanism to remove damaged proteins and organelles through lysosomal degradation. Optineurin was identified as an autophagy receptor that directly interacts with autophagosomal protein, LC3, and ubiquitin. These interactions are important for autophagy receptor function. Autophagy receptors recruit their cargo and take it to autophagosomes which fuse with lysosomes to form autolysosomes where degradation of proteins takes place. Optineurin interacts with a motor protein, myosinVI, and this interaction is involved in mediating fusion of autophagosomes with lysosomes. A glaucoma-associated mutant of optineurin, E50K, impairs autophagy as well as vesicle trafficking, leading to death of retinal cells by apoptosis. E50K-OPTN-induced block in autophagy is dependent on a GTPase activating protein, TBC1D17. The E50K mutant also causes other changes in the cells such as altered interaction with TBK1 protein kinase, aggregate formation, generation of reactive oxygen species and inhibition of proteasome, which may contribute to pathogenesis. A polymorphism of optineurin, M98K, associated with glaucoma, causes enhanced autophagy leading to transferrin receptor degradation and apoptotic death of retinal cells. M98K-OPTN-induced autophagic cell death is dependent on Rab12 GTPase. Thus, an optimum level of optineurin-mediated autophagy is crucial for survival of retinal cells, and impaired autophagy is likely to contribute to glaucoma pathogenesis. How impaired autophagy caused by optineurin mutants leads to apoptosis and cell death, is yet to be explored.


Sujet(s)
Autophagie , Glaucome à angle ouvert/génétique , Polymorphisme de nucléotide simple , Facteur de transcription TFIIIA/génétique , Autophagie/génétique , Protéines du cycle cellulaire , Glaucome à angle ouvert/métabolisme , Humains , Protéines de transport membranaire , Récepteurs à la transferrine/métabolisme
20.
PLoS One ; 10(9): e0138289, 2015.
Article de Anglais | MEDLINE | ID: mdl-26376340

RÉSUMÉ

Certain missense mutations in optineurin/OPTN and amplification of TBK1 are associated with normal tension glaucoma. A glaucoma-associated variant of OPTN, M98K, induces autophagic degradation of transferrin receptor (TFRC) and death in retinal cells. Here, we have explored the role of Tbk1 in M98K-OPTN-induced autophagy and cell death, and the effect of Tbk1 overexpression in retinal cells. Cell death induced by M98K-OPTN was dependent on Tbk1 as seen by the effect of Tbk1 knockdown and blocking of Tbk1 activity by a chemical inhibitor. Inhibition of Tbk1 also restores M98K-OPTN-induced transferrin receptor degradation. M98K-OPTN-induced autophagosome formation, autophagy and cell death were dependent on its phosphorylation at S177 by Tbk1. Knockdown of OPTN reduced starvation-induced autophagosome formation. M98K-OPTN expressing cells showed higher levels of Tbk1 activation and enhanced phosphorylation at Ser177 compared to WT-OPTN expressing cells. M98K-OPTN-induced activation of Tbk1 and its ability to be phosphorylated better by Tbk1 was dependent on ubiquitin binding. Phosphorylated M98K-OPTN localized specifically to autophagosomes and endogenous Tbk1 showed increased localization to autophagosomes in M98K-OPTN expressing cells. Overexpression of Tbk1 induced cell death and caspase-3 activation that were dependent on its catalytic activity. Tbk1-induced cell death possibly involves autophagy, as shown by the effect of Atg5 knockdown, and requirement of autophagic function of OPTN. Our results show that phosphorylation of Ser177 plays a crucial role in M98K-OPTN-induced autophagosome formation, autophagy flux and retinal cell death. In addition, we provide evidence for cross talk between two glaucoma associated proteins and their inter-dependence to mediate autophagy-dependent cell death.


Sujet(s)
Glaucome/génétique , Phagosomes/physiologie , Protein-Serine-Threonine Kinases/métabolisme , Récepteurs à la transferrine/métabolisme , Cellules ganglionnaires rétiniennes/anatomopathologie , Facteur de transcription TFIIIA/métabolisme , Animaux , Autophagie , Technique de Western , Protéines du cycle cellulaire , Cellules cultivées , Technique d'immunofluorescence indirecte , Cellules HeLa , Humains , Immunoprécipitation , Protéines de transport membranaire , Souris , Microscopie confocale , Phosphorylation , Liaison aux protéines , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/génétique , Transport des protéines , Petit ARN interférent/génétique , Cellules ganglionnaires rétiniennes/métabolisme , Transduction du signal , Facteur de transcription TFIIIA/antagonistes et inhibiteurs , Facteur de transcription TFIIIA/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE