CCL

Összesen 9 találat.
#/oldal:
Részletezés:
Rendezés:

1.

001-es BibID:BIBFORM118666
035-os BibID:(Scopus)85184251918 (WOS)001155051600001
Első szerző:Ababneh, Haneen Muntasir
Cím:High glucose promotes osteogenic differentiation of human lens epithelial cells through hypoxia-inducible factor (HIF) activation / Haneen Ababneh, Enikő Balogh, Dávid Máté Csiki, Gréta Lente, Ferenc Fenyvesi, Andrea Tóth, Viktória Jeney
Dátum:2024
ISSN:0021-9541 1097-4652
Megjegyzések:Cataract, a leading cause of blindness, is characterised by lens opacification. Type 2 diabetes is associated with a two- to fivefold higher prevalence of cataracts. The risk of cataract formation increases with the duration of diabetes and the severity of hyperglycaemia. Hydroxyapatite deposition is present in cataractous lenses that could be the consequence of osteogenic differentiation and calcification of lens epithelial cells (LECs). We hypothesised that hyperglycaemia might promote the osteogenic differentiation of human LECs (HuLECs). Osteogenic medium (OM) containing excess phosphate and calcium with normal (1 g/L) or high (4.5 g/L) glucose was used to induce HuLEC calcification. High glucose accelerated and intensified OM-induced calcification of HuLECs, which was accompanied by hyperglycaemia-induced upregulation of the osteogenic markers Runx2, Sox9, alkaline phosphatase and osteocalcin, as well as nuclear translocation of Runx2. High glucose-induced calcification was abolished in Runx2-deficient HuLECs. Additionally, high glucose stabilised the regulatory alpha subunits of hypoxia-inducible factor 1 (HIF-1), triggered nuclear translocation of HIF-1? and increased the expression of HIF-1 target genes. Gene silencing of HIF-1? or HIF-2? attenuated hyperglycaemia-induced calcification of HuLECs, while hypoxia mimetics (desferrioxamine, CoCl2) enhanced calcification of HuLECs under normal glucose conditions. Overall, this study suggests that high glucose promotes HuLEC calcification via Runx2 and the activation of the HIF-1 signalling pathway. These findings may provide new insights into the pathogenesis of diabetic cataracts, shedding light on potential factors for intervention to treat this sight-threatening condition.
Tárgyszavak:Természettudományok Biológiai tudományok idegen nyelvű folyóiratközlemény külföldi lapban
folyóiratcikk
cataract
hyperglycaemia
hypoxia-inducible factor (HIF)
lens calcification
lens epithelial cell
osteogenic differentiation
Megjelenés:Journal Of Cellular Physiology. - [Epub ahead of print] (2024). -
További szerzők:Balogh Enikő (1987-) (molekuláris biológus) Csiki Dávid Máté (1993-) (biomérnök) Lente Gréta (1994-) (klinikai laboratóriumi kuatató) Fenyvesi Ferenc (1977-) (gyógyszerész, gyógyszertechnológus) Tóth Andrea (1992-) (molekuláris biológus) Jeney Viktória (1971-) (vegyész, kémia tanár)
Pályázati támogatás:NKFIH K146669
Egyéb
Internet cím:Szerző által megadott URL
DOI
Intézményi repozitóriumban (DEA) tárolt változat
Borító:

2.

001-es BibID:BIBFORM047427
Első szerző:Adyshev, Djanybek M.
Cím:Ezrin/radixin/moesin proteins differentially regulate endothelial hyperpermeability after thrombin / Djanybek M. Adyshev, Steven M. Dudek, Nurgul Moldobaeva, Kyung-mi Kim, Shwu-Fan Ma, Anita Kasa, Joe G. N. Garcia, Alexander D. Verin
Dátum:2013
ISSN:1040-0605
Megjegyzések:Endothelial cell (EC) barrier disruption induced by inflammatory agonists such as thrombin leads to potentially lethal physiological dysfunction such as alveolar flooding, hypoxemia and pulmonary edema. Thrombin stimulates paracellular gap and F-actin stress fiber formation, triggers actomyosin contraction and alters EC permeability through multiple mechanisms that include protein kinase C (PKC) activation. We previously have shown that the ezrin, radixin, and moesin (ERM) actin-binding proteins differentially participate in S1P-induced EC barrier enhancement. Phosphorylation of a conserved threonine residue in the C terminus of ERM proteins causes conformational changes in ERM to unmask binding sites and is considered a hallmark of ERM activation. In the present study we test the hypothesis that ERM proteins are phosphorylated on this critical threonine residue by thrombin-induced signaling events and explore the role of the ERM family in modulating thrombin-induced cytoskeletal rearrangement and EC barrier function. Thrombin promotes ERM phosphorylation at this threonine residue (Ezrin-567, Radixin-564, Moesin-558) in a PKC-dependent fashion and induces translocation of phosphorylated ERM to the EC periphery. Thrombin-induced ERM threonine phosphorylation is likely synergistically mediated by protease-activated receptors PAR1 and PAR2. Using the siRNA approach, depletion of either moesin alone, or of all three ERM proteins, significantly attenuates thrombin-induced increase in EC barrier permeability (TER), cytoskeletal rearrangements, paracellular gap formation and accumulation of di-phospho-MLC. In contrast, radixin depletion exerts opposing effects on these indices. These data suggest that ERM proteins play important differential roles in the thrombin-induced modulation of EC permeability, with moesin promoting barrier dysfunction and radixin opposing it
Tárgyszavak:Orvostudományok Elméleti orvostudományok idegen nyelvű folyóiratközlemény külföldi lapban
thrombin
ERM
PKC
phosphorylation
Megjelenés:American Journal of Physiology-Lung Cellular and Molecular Physiology 305 : 3 (2013), p. L240-L255. -
További szerzők:Dudek, Steven Moldobaeva, Nurgul Kim, Kyung-mi Ma, Shwu-Fan Kovács-Kása Anita (1983-) Garcia, Joe G. N. Verin, Alexander
Internet cím:Szerző által megadott URL
DOI
Intézményi repozitóriumban (DEA) tárolt változat
Borító:

3.

001-es BibID:BIBFORM074060
Első szerző:Bátori Róbert Károly (biológus, biotechnológus)
Cím:Differential mechanisms of adenosine- and ATpγS-induced microvascular endothelial barrier strengthening / Róbert Bátori, Sanjiv Kumar, Zsuzsanna Bordán, Mary Cherian-Shaw, Anita Kovács-Kása, Justin A. MacDonald, David J. R. Fulton, Ferenc Erdődi, Alexander D. Verin
Dátum:2019
ISSN:0021-9541
Tárgyszavak:Természettudományok Biológiai tudományok idegen nyelvű folyóiratközlemény külföldi lapban
Endothelial barrier
Megjelenés:Journal of Cellular Physiology. - 234 : 5 (2019), p. 5863-5879. -
További szerzők:Kumar, Sanjiv Bordán Zsuzsanna Cherian-Shaw, Mary Kovács-Kása Anita (1983-) MacDonald, Justin A. Fulton, David Erdődi Ferenc (1953-) (biokémikus) Verin, Alexander
Internet cím:Szerző által megadott URL
DOI
Intézményi repozitóriumban (DEA) tárolt változat
Borító:

4.

001-es BibID:BIBFORM074172
Első szerző:Csortos Csilla (biokémikus)
Cím:TIMAP is a positive regulator of pulmonary endothelial barrier function / Csilla Csortos, Istvan Czikora, Natalia V. Bogatcheva, Djanybek M. Adyshev, Christophe Poirier, Gabor Olah, Alexander D. Verin
Dátum:2008
ISSN:1040-0605 1522-1504
Megjegyzések:TGF-?-inhibited membrane-associated protein, TIMAP, is expressed at high levels in endothelial cells (EC). It is regarded as a member of the MYPT (myosin phosphatase target subunit) family of protein phosphatase 1 (PP1) regulatory subunits; however, its function in EC is not clear. In our pull-down experiments, recombinant TIMAP binds preferentially the ?-isoform of the catalytic subunit of PP1 (PP1c?) from pulmonary artery EC. As PP1c?, but not PP1c?, binds with MYPT1 into functional complex, these results suggest that TIMAP is a novel regulatory subunit of myosin phosphatase in EC. TIMAP depletion by small interfering RNA (siRNA) technique attenuates increases in transendothelial electrical resistance induced by EC barrier-protective agents (sphingosine-1-phosphate, ATP) and enhances the effect of barrier-compromising agents (thrombin, nocodazole) demonstrating a barrier-protective role of TIMAP in EC. Immunofluorescent staining revealed colocalization of TIMAP with membrane/cytoskeletal protein, moesin. Moreover, TIMAP coimmunoprecipitates with moesin suggesting the involvement of TIMAP/moesin interaction in TIMAP-mediated EC barrier enhancement. Activation of cAMP/PKA cascade by forskolin, which has a barrier-protective effect against thrombin-induced EC permeability, attenuates thrombin-induced phosphorylation of moesin at the cell periphery of control siRNA-treated EC. On the contrary, in TIMAP-depleted EC, forskolin failed to affect the level of moesin phosphorylation at the cell edges. These results suggest the involvement of TIMAP in PKA-mediated moesin dephosphorylation and the importance of this dephosphorylation in TIMAP-mediated EC barrier protection.protein phosphorylation and dephosphorylation are known to be the key signaling events affecting the status of vascular endothelial barrier (11). Cytoskeletal and intercellular junctional proteins are regulated via reversible phosphorylation of serine (Ser), threonine (Thr), or tyrosine (Tyr) side chains. Based on many recent data, it is apparent that several types of protein phosphatases are intimately involved in the regulation of endothelial barrier function (10, 17, 27?29). However, their regulation is not yet completely understood.Protein phosphatase 1 (PP1) is a multimeric phosphoserine/phosphothreonine-specific phosphatase. One of the four different isoforms, ?, ?, ?1, or ?2, of the catalytic subunit (PP1c) binds to one (or two) protein from a pool of regulatory subunits (R). The holoenzyme forms possess diverse cellular functions. A common structural element of R proteins is a short, conserved PP1c binding motif, (R/K)VXF (3, 9, 10). Different R subunits may direct PP1 holoenzymes to distinct subcellular locations and increase or suppress the activity toward specific substrates (3, 9). Myosin light chain phosphatase (or myosin phosphatase, MP), for example, is composed of PP1c? and two regulatory subunits, namely, a larger targeting/regulatory subunit (myosin phosphatase target subunit, MYPT) and a small regulatory subunit (M20) (2, 10, 14). The activity of MP holoenzyme is increased toward phosphorylated myosin compared with the activity of the PP1c monomer (15).It was recently shown that MP function is not limited to myosin dephosphorylation. The MP regulatory subunit MYPT1 can directly bind to F-actin binding proteins including ERM proteins (ezrin-radixin-moesin family). These proteins could be phosphorylated by either protein kinase C? or Rho kinase (12, 20); phosphorylation renders unfolded ERM protein, enabling its interaction with actin and membrane proteins (20, 21). ERM dephosphorylation by MP seems to affect ERM conformation and cytoskeletal/membrane binding capacities (12, 20). These data indicate that MP not only dephosphorylates myosin, but it is also involved in the regulation of F-actin cytoskeleton.Recently, other proteins of the MYPT family, namely MYPT3, TIMAP (TGF-?-inhibited membrane-associated protein), and myosin binding subunit 85 (MBS85), were identified and characterized from different sources (8, 25, 26). They share some structural features with MYPT1, e.g., all of these proteins contain the PP1c binding motif followed by ankyrin repeats. On the other hand, MYPT3, TIMAP, and MBS85 have their own special features as well. For example, both TIMAP and MYPT3 have COOH-terminal prenylation motif suggesting possible membrane association. The high level of homology with MYPT1 implies that TIMAP, MYPT3, and MBS85 may be regulatory subunits of PP1; however, their physiological significance is not known.TIMAP is a 64-kDa protein expressed at high levels in endothelial cells (EC). As TIMAP mRNA synthesis is strongly downregulated by TGF-?1 (8), it is likely to assume that TIMAP may be an important component of endothelial response to TGF-?1, including apoptosis, capillary morphogenesis, and barrier dysfunction. It is highly homologous to MYPT3 (?45% amino acid homology) and shares its structural features, i.e., PP1c binding motif, ankyrin repeats, prenylation motif, and possible nuclear localization signals (8). Yeast and bacterial two-hybrid screening revealed several potential protein partners for TIMAP (1, 16). For instance, TIMAP interacts with the 37/67-kDa laminin receptor (LAMR1). It was suggested that TIMAP targets PP1c to LAMR1, and LAMR1 is a TIMAP-dependent PP1c substrate (16). Although protein-protein interaction between TIMAP and PP1c was shown by immunoprecipitation, its role in regulating PP1c activity is not clarified yet. In the present work, we present evidence for specific interaction between TIMAP and PP1c?. Furthermore, we show that TIMAP has a barrier-protective role in human pulmonary artery endothelial cells (HPAEC), and we propose that ERM proteins are among its targets.
Tárgyszavak:Orvostudományok Elméleti orvostudományok idegen nyelvű folyóiratközlemény külföldi lapban
transendothelial electrical resistance
small interfering RNA
moesin interaction with protein phosphatase 1
Megjelenés:American Journal Of Physiology-Lung Cellular And Molecular Physiology. - 295 : 3 (2008), p. L440-L450. -
További szerzők:Czikora István (1979-) (vegyész, biokémikus) Bogatcheva, Natalia V. Adyshev, Djanybek M. Poirier, Christophe Oláh Gábor Verin, Alexander
Internet cím:Szerző által megadott URL
DOI
Intézményi repozitóriumban (DEA) tárolt változat
Borító:

5.

001-es BibID:BIBFORM001291
Első szerző:Csortos Csilla (biokémikus)
Cím:Regulation of vascular endothelial cell barrier function and cytoskeleton structure by protein phosphatases of the PPP family / Csortos Cs., Kolosova I., Verin A. D.
Dátum:2007
Tárgyszavak:Orvostudományok Elméleti orvostudományok idegen nyelvű folyóiratközlemény külföldi lapban
endothelial barrier function
Ser/Thr protein phosphatases
Megjelenés:American Journal of Physiology-Lung Cellular and Molecular Phsiology. - 293 (2007), p. L843-L854. -
További szerzők:Kolosova, Irina Verin, Alexander
Internet cím:elektronikus változat
DOI
Borító:

6.

001-es BibID:BIBFORM002464
Első szerző:Helyes Zsuzsanna
Cím:Role of transient receptor potential vanilloid 1 receptors in endotoxin-induced / Zsuzsanna Helyes, Krisztián Elekes, József Németh, Gábor Pozsgai, Katalin Sándor, László Kereskai, Rita Börzsei, Erika Pintér, Árpád Szabó, János Szolcsányi
Dátum:2007
Megjegyzések:Airways are densely innervated by capsaicin-sensitive sensory neurons expressing transient receptor potential vanilloid 1 (TRPV1) receptors/ion channels, which play an important regulatory role in inflammatory processes via the release of sensory neuropeptides. The aim of the present study was to investigate the role of TRPV1 receptors in endotoxin-induced airway inflammation and consequent bronchial hyperreactivity with functional, morphological, and biochemical techniques using receptor gene-deficient mice. Inflammation was evoked by intranasal administration of Escherichia coli lipopolysaccharide (60 gammal, 167 gammag/ml) in TRPV1 knockout (TRPV1-/-) mice and their wild-type counterparts (TRPV1+/+) 24 h before measurement. Airway reactivity was assessed by unrestrained whole body plethysmography, and its quantitative indicator, enhanced pause (Penh), was calculated after inhalation of the bronchoconstrictor carbachol. Histological examination and spectrophotometric myeloperoxidase measurement was performed from the lung. Somatostatin concentration was measured in the lung and plasma with radioimmunoassay. Bronchial hyperreactivity, histological lesions (perivascular/peribronchial edema, neutrophil/ macrophage infiltration, goblet cell hyperplasia), and myeloperoxidase activity were significantly greater in TRPV-/- mice. Inflammation markedly elevated lung and plasma somatostatin concentrations in TRPV1+/+ but not TRPV1-/- animals. In TRPV1-/- mice, exogenous administration of somatostatin-14 (4 x 100 gamma g/kg ip) diminished inflammation and hyperreactivity. Furthermore, in wildtype mice, antagonizing somatostatin receptors by cyclo-somatostatin (4 x 250 gamma g/kg ip) increased these parameters. This study provides the first evidence for a novel counterregulatory mechanism during endotoxin-induced airway inflammation, which is mediated by somatostatin released from sensory nerve terminals in response to activation of TRPV1 receptors of the lung. It reaches the systemic circulation and inhibits inflammation and consequent bronchial hyperreactivity.
Tárgyszavak:Orvostudományok Elméleti orvostudományok idegen nyelvű folyóiratközlemény külföldi lapban
capsaicin-sensitive afferents
inflammatory airway hyperreactivity
Megjelenés:American journal of physiology. Lung cellular and molecular physiology. - 292 (2007), p. L1173-L1181. -
További szerzők:Sándor Katalin Szolcsányi János (Pécs) Szabó Árpád Pintér Erika Börzsei Rita Kereskai László Pozsgai Gábor Elekes Krisztián Németh József (1954-) (vegyész, analitikus)
Internet cím:elektronikus változat
DOI
Borító:

7.

001-es BibID:BIBFORM029024
Első szerző:Kim, Kyung-mi
Cím:Molecular characterization of myosin phosphatase in endothelium / Kyung-mi Kim, Csilla Csortos, Istvan Czikora, David Fulton, Nagavedi S. Umapathy, Gabor Olah, Alexander D. Verin
Dátum:2012
ISSN:0021-9541
Megjegyzések:The phosphorylation status of myosin light chain (MLC) is regulated by both MLC kinases and type 1 Ser/Thr phosphatase (PPase 1), MLC phosphatase (MLCP) activities. The activity of the catalytic subunit of MLCP (CS1β) towards myosin depends on its associated regulatory subunit, namely myosin PPase targeting subunit 1 (MYPT1). Our previously published data strongly suggested the involvement of MLCP in endothelial cell (EC) barrier regulation. In this study, our new data demonstrate that inhibition of MLCP by either CS1β or MYPT1 siRNA-based depletion results in significant attenuation of purine nucleotide (ATP and adenosine)-induced EC barrier enhancement. Consistent with the data, thrombin-induced EC F-actin stress fiber formation and permeability increase were attenuated by the ectopic expression of constitutively active (C/A) MYPT1. The data demonstrated for the first time direct involvement of MLCP in EC barrier enhancement/protection. Cloning of MYPT1 in human pulmonary artery EC (HPAEC) revealed the presence of two MYPT1 isoforms, long and variant 2 (V2) lacking 56 amino acids from 553 to 609 of human MYPT1 long, which were previously identified in HeLa and HEK 293 cells. Our data demonstrated that in Cos-7 cells ectopically expressed EC MYPT1 isoforms co-immunoprecipitated with intact CS1β suggesting the importance of PPase 1 activity for the formation of functional complex of MYPT1/CS1β. Interestingly, MYPT1 V2 shows decreased binding affinity compared to MYPT1 long for radixin (novel MLCP substrate and a member of ERM family proteins). These results suggest functional difference between EC MYPT1 isoforms in the regulation of MLCP activity and cytoskeleton. J. Cell. Physiol. 227: 1701-1708, 2012. © 2011 Wiley Periodicals, Inc.
Tárgyszavak:Orvostudományok Elméleti orvostudományok idegen nyelvű folyóiratközlemény külföldi lapban
egyetemen (Magyarországon) készült közlemény
Megjelenés:Journal of Cellular Physiology. - 227 : 4 (2012), p. 1701-1708. -
További szerzők:Fulton, David Umapathy, Nagavedi S. Oláh Gábor Verin, Alexander Csortos Csilla (1956-) (biokémikus) Czikora István (1979-) (vegyész, biokémikus)
Internet cím:Szerző által megadott URL
DOI
Intézményi repozitóriumban (DEA) tárolt változat
Borító:

8.

001-es BibID:BIBFORM110434
035-os BibID:(scopus)85151405198
Első szerző:Kohler Zoltán M.
Cím:Tilorone increases glucose uptake in vivo and in skeletal muscle cells by enhancing Akt2/AS160 signaling and glucose transporter levels / Kohler Zoltan M., Trencsenyi Gyorgy, Juhasz Laszlo, Zvara Agnes, Szabo Judit P., Dux Laszlo, Puskas Laszlo G., Rovo Laszlo, Keller-Pinter Aniko
Dátum:2023
ISSN:0021-9541 1097-4652
Tárgyszavak:Orvostudományok Elméleti orvostudományok idegen nyelvű folyóiratközlemény külföldi lapban
folyóiratcikk
Megjelenés:Journal Of Cellular Physiology. - 238 : 5 (2023), p. 1080-1094. -
További szerzők:Trencsényi György (1978-) (biológus, biokémikus, molekuláris biológus) Juhász László Zvara Ágnes Péli-Szabó Judit (1977-) (vegyész) Dux László Puskás László G. Rovó László Keller-Pintér Anikó
Pályázati támogatás:GINOP?2.3.2?15?2016?00040
GINOP
EFOP-3.6.2?16?2017?00006
EFOP
FK 134684
Egyéb
K 132446
Egyéb
TKP2021?EGA?28
Egyéb
TKP2021?EGA
Egyéb
UNKP?21?5?SZTE?571
UNKP
Internet cím:Szerző által megadott URL
DOI
Intézményi repozitóriumban (DEA) tárolt változat
Borító:

9.

001-es BibID:BIBFORM094525
035-os BibID:(WoS)000651650200001 (Scopus)85106224539 (PubMed)34008188
Első szerző:Matta Csaba (molekuláris biológus, genetikus, angol szakfordító)
Cím:Transcriptome-based screening of ion channels and transporters in a migratory chondroprogenitor cell line isolated from late-stage osteoarthritic cartilage / Csaba Matta, Rebecca Lewis, Christopher Fellows, Gyula Diszhazi, Janos Almassy, Nicolai Miosge, James Dixon, Marcos C. Uribe, Sean May, Szilard Poliska, Richard Barrett-Jolley, Janos Fodor, Peter Szentesi, Tibor Hajdú, Aniko Keller-Pinter, Erin Henslee, Fatima H. Labeed, Michael P. Hughes, Ali Mobasheri1
Dátum:2021
ISSN:0021-9541 1097-4652
Tárgyszavak:Orvostudományok Elméleti orvostudományok idegen nyelvű folyóiratközlemény külföldi lapban
folyóiratcikk
Megjelenés:Journal of Cellular Physiology. - 236 : 11 (2021), p. 7421-7439. -
További szerzők:Lewis, Rebecca Fellows, Christopher R. Diszházi Gyula (1992-) (gyógyszerész) Almássy János (1981-) (élettanász, biológus, angol-magyar szakfordító) Miosge, Nicolai Dixon, James E. Uribe, Marcos C. May, Sean Póliska Szilárd (1978-) (biológus) Barrett-Jolley, Richard Fodor János (1973-) (élettanász, biotechnológus) Szentesi Péter (1967-) (élettanász) Hajdú Tibor (1988-) (általános orvos) Keller-Pintér Anikó Henslee, Erin Labeed, Fatima H. Hughes, Michael P. Mobasheri, Ali
Internet cím:Szerző által megadott URL
DOI
Intézményi repozitóriumban (DEA) tárolt változat
Borító:
Rekordok letöltése1