Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Boglarka Racz is active.

Publication


Featured researches published by Boglarka Racz.


Neuroscience Letters | 2004

Effects of pituitary adenylate cyclase activating polypeptide in retinal degeneration induced by monosodium-glutamate.

Andrea Tamas; Robert Gábriel; Boglarka Racz; Viktoria Denes; Peter Kiss; Andrea Lubics; István Lengvári; Dóra Reglődi

Pituitary adenylate cyclase activating polypeptide (PACAP) is a pleiotropic neuropeptide with a wide range of effects in the central and peripheral nervous systems. PACAP has well-documented neurotrophic and neuroprotective actions in both in vitro and in vivo models of different neuronal injuries. The aim of the present study was to investigate the possible neuroprotective effect of PACAP in retinal degeneration induced by monosodium-glutamate (MSG) in neonatal rats. Preceding the MSG treatment, PACAP (1 or 100pmol/5mul) was injected unilaterally into the vitreous body on postnatal days 1, 5 and 9. Immediately after the PACAP treatment, pups were treated with 2mg/g body weight MSG subcutaneously. At 3 weeks of age, rats were sacrificed and retinas were removed and processed for histological examination. Our results show that MSG treatment caused severe degeneration, primarily of the inner retinal layers. The thickness of the entire retina was only approximately half of that of the normal retinas, and the inner nuclear layer seemed to be fused with the ganglionic cell layer, with no discernible inner plexiform layer. Retinas of animals treated with 1pmol PACAP showed a similar degree of degeneration. However, retinas of rats treated with 100pmol PACAP showed significantly less damage, with clearly distinguishable inner retinal layers. In summary, our present study shows that local PACAP treatment could attenuate the retinal degeneration induced by the excitotoxic effects of glutamate.


Regulatory Peptides | 2006

The neuroprotective effects of PACAP in monosodium glutamate-induced retinal lesion involve inhibition of proapoptotic signaling pathways

Boglarka Racz; Ferenc Gallyas; Peter Kiss; Gábor K. Tóth; Orsolya Hegyi; Balázs Gasz; Balázs Borsiczky; Andrea Ferencz; Erzsébet Roth; Andrea Tamas; István Lengvári; Andrea Lubics; Dora Reglodi

Pituitary adenylate cyclase activating polypeptide (PACAP) and its receptors are present in the retina and exert several distinct functions. PACAP has well-known neuroprotective effects in neuronal cultures in vitro and against different insults in vivo. Recently we have shown that PACAP is neuroprotective against monosodium glutamate (MSG)-induced retinal degeneration. In the present study we investigated the possible signal transduction pathways involved in the protective effect of intravitreal PACAP administration against apoptotic retinal degeneration induced by neonatal MSG treatment. MSG induced activation of proapoptotic signaling proteins and reduced the levels of antiapoptotic molecules in neonatal retinas. Co-treatment with PACAP attenuated the MSG-induced activation of caspase-3 and JNK, inhibited the MSG-induced cytosolic translocation of apoptosis inducing factor (AIF) and cytochrome c, and increased the level of phospho-Bad. Furthermore, PACAP treatment alone decreased cytosolic AIF and cytochrome c levels, while PACAP6-38 increased cytochrome c release, caspase-3 and JNK activity and decreased phospho-Bad activity. In summary, our results show that PACAP treatment attenuated the MSG-induced changes in apoptotic signaling molecules in vivo and suggest that also endogenously present PACAP has neuroprotective effects. These results may have further clinical implications in reducing glutamate-induced excitotoxicity in several ophthalmic diseases.


Annals of the New York Academy of Sciences | 2010

Pituitary adenylate cyclase activating polypeptide in the retina: focus on the retinoprotective effects.

Tamas Atlasz; Krisztina Szabadfi; Peter Kiss; Boglarka Racz; Ferenc Gallyas; Andrea Tamas; V. Gaal; Zs. Márton; Robert Gábriel; Dora Reglodi

Pituitary adenylate cyclase activating polypeptide (PACAP) is a neurotrophic and neuroprotective peptide that has been shown to exert protective effects against different neuronal injuries, such as traumatic brain and spinal cord injury, models of neurodegenerative diseases, and cerebral ischemia. PACAP and its receptors are present in the retina. In this study, we summarize the current knowledge on retinal PACAP with focus on the retinoprotective effects. Results of histological, immunohistochemical, and molecular biological analysis are reviewed. In vitro, PACAP shows protection against glutamate, thapsigargin, anisomycin, and anoxia. In vivo, the protective effects of intravitreal PACAP treatment have been shown in the following models of retinal degeneration in rats: excitotoxic injury induced by glutamate and kainate, ischemic injury, degeneration caused by UV‐A light, optic nerve transection, and streptozotocin‐induced diabetic retinopathy. Studying the molecular mechanism has revealed that PACAP acts by activating antiapoptotic and inhibiting proapoptotic signaling pathways in the retina in vivo. These studies strongly suggest that PACAP is an excellent candidate retinoprotective agent that could be a potential therapeutic substance in various retinal diseases.


Regulatory Peptides | 2008

PKA-Bad-14-3-3 and Akt-Bad-14-3-3 signaling pathways are involved in the protective effects of PACAP against ischemia/reperfusion-induced cardiomyocyte apoptosis

Boglarka Racz; Balázs Gasz; Ferenc Gallyas; Peter Kiss; Andrea Tamas; Zalán Szántó; Andrea Lubics; I. Lengvári; Gábor K. Tóth; Orsolya Hegyi; Erzsébet Roth; Dora Reglodi

The neuropeptide PACAP (pituitary adenylate cyclase activating polypeptide) and its receptors are widely expressed in the nervous system and various other tissues. PACAP has well-known anti-apoptotic effects in neuronal cell lines. Recent data suggest that PACAP exerts anti-apoptotic effects also in non-neuronal cells. The peptide is present in the cardiovascular system, and has various distinct effects. The aim of the present study was to investigate whether PACAP is protective against in vitro ischemia/reperfusion-induced apoptosis in cardiomyocytes. Cultured cardiomyocytes were exposed to 60 min ischemia followed by 120 min reperfusion. The addition of PACAP1-38 significantly increased cell viability and decreased the ratio of apoptotic cells as measured by MTT test and flow cytometry. PACAP induced the phosphorylation of Akt and protein kinase A. In the present study we also examined the possible involvement of Akt- and protein kinase A-induced phosphorylation and thus inactivation of Bad, a pro-apoptotic member of the Bcl-2 family. It was found that ischemia significantly decreased the levels of phosphorylated Bad, which was counteracted by PACAP. Furthermore, PACAP increased the levels of Bcl-xL and 14-3-3 protein, both of which promote cell survival, and decreased the apoptosis executor caspase-3 cleavage. All effects of PACAP1-38 were inhibited by the PACAP antagonist PACAP6-38. In summary, our results show that PACAP has protective effects against ischemia/reperfusion-induced cardiomyocyte apoptosis and provides new insights into the signaling mechanisms involved in the PACAP-mediated anti-apoptotic effects.


Neurotoxicity Research | 2007

Effects of pituitary adenylate cyclase activating polypeptide (PACAP) on the PKA-Bad-14-3-3 signaling pathway in glutamate-induced retinal injury in neonatal rats.

Boglarka Racz; Ferenc Gallyas; Peter Kiss; Andrea Tamas; Andrea Lubics; István Lengvári; Erzsébet Roth; Gábor K. Tóth; Orsolya Hegyi; Zsófia Verzár; Csaba Fabricsek; Dora Reglodi

The neuropeptide PACAP (pituitary adenylate cyclase activating polypeptide) and its receptors are widely expressed in the nervous system including the retina. PACAP has well-known neuroprotective effects in neuronal culturesin vitro and against different insultsin vivo. Recently, we have shown that PACAP1-38 is neuroprotective against monosodium glutamate (MSG)-induced retinal degeneration. Studying the molecular mechanisms of this protection has revealed that PACAP1-38 stimulates antiapoptotic mechanisms such as phosphorylation of ERK1/2 and inhibits pro-apoptotic signaling molecules such as JNK1/2, p38MAPK, caspase-3 and the translocation of mitochondrial cytochromec and apoptosis inducing factor in glutamate-treated retinasin vivo. In the present study we investigated the effects of PACAP1-38 on a further signal transduction pathway possibly involved in the protective effect of intravitreal PACAP1-38 administration against apoptotic retinal degeneration induced by neonatal MSG treatment. The focus of the present study was the protein kinase A (PKA)-Bad-14-3-3 transduction pathway.In vivo MSG treatment led to a reduction in the levels of anti-apoptotic molecules (phospho-PKA phospho-Bad, Bcl-xL and 14-3-3 proteins) in the retina. Co-treatment with PACAP1-38 counteracted these effects: the level of phospho-PKA, phospho-Bad, Bcl-xL and 14-3-3 were increased. All effects of PACAP1-38 were inhibited by the PACAP antagonist PACAP6-38. In summary, our results show that PACAP1-38 activates the PKA-Bad-14-3-3 pathway which is inhibited by MSG treatment. Our results also provide new insights into the signaling mechanisms possibly involved in the PACAP-mediated anti-apoptotic effects.


Annals of the New York Academy of Sciences | 2007

Expression and Protective Role of Heme Oxygenase-1 in Delayed Myocardial Preconditioning

Gábor Jancsó; Barbara Cserepes; Balázs Gasz; László Benko; Balázs Borsiczky; Andrea Ferenc; Mária Kürthy; Boglarka Racz; János Lantos; János Gál; Endre Arató; Lószló Sínayc; György Wéber; Erzsébet Roth

Abstract:  In the study the authors aimed to demonstrate the expression and protective effect of heme oxygenase‐1 (HO‐1) in the delayed preconditioning (PC) on cultured myocardiac cells. Neonatal rat cardiac myocytes were exposed to ischemic (ischemic medium [IM] for 20 min) and pharmacological (adenosine, epinephrine, opioid) PC. Twenty‐four hours later cells were subjected to a simulated ischemia (SI )—culturing for 3 h in IM, followed by 2‐h reperfusion in normal medium‐–and then lactate dehydrogenase (LDH), live/death ratio, and apoptosis were measured. For demonstrating the protective role of HO‐1, its enzymatic activity was competitively inhibited by administration of zinc protoporphyrin IX (ZnPPIX), and HO‐1 synthesis was blocked with HO‐1 siRNA. Cells in control group were cultured under normoxic conditions. In SI group, cells underwent only an SI without PC. HO‐1 expression in all of the groups was demonstrated with immunostaining. Our results showed a significant decrease of LDH release, apoptosis, and cell death in PC groups versus SI group, which has been risen in ZnPPIX‐ and HO‐1 siRNA‐treated groups. HO‐1 immunostaining showed an appreciable HO‐1 expression in PC groups, which was abolished with HO‐1 siRNA administration, but not in ZnPPIX group. The results therefore suggest that HO‐1 expression increases in both ischemic and pharmacological PC, and HO‐1 has cellular protective effect against cell death and apoptosis in ischemia‐reperfusion‐induced oxidative injury.


Free Radical Biology and Medicine | 2010

Regulation of MKP-1 expression and MAPK activation by PARP-1 in oxidative stress: A new mechanism for the cytoplasmic effect of PARP-1 activation

Boglarka Racz; Katalin Hanto; Antal Tapodi; Izabella Solti; Nikoletta Kálmán; Péter B. Jakus; Krisztina Kovacs; Balazs Debreceni; Ferenc Gallyas; Balazs Sumegi

Previously, it was suggested that the release of nuclearly formed ADP-ribose polymers or ADP-ribosylated proteins could be responsible for the cytosolic and mitochondrial effects of poly(ADP-ribose) polymerase (PARP)-1 activation in oxidative stress. In this report, we provide a novel alternative mechanism. We found that reactive oxygen species-activated PARP-1 regulated the activation of JNK and p38 mitogen-activated protein kinases (MAPKs) because inhibition of PARP-1 by pharmacons, small interfering RNA silencing of PARP-1 expression, or the transdominant expression of enzymatically inactive PARP-1 resulted in the inactivation of these MAPKs. This regulation was achieved by increased expression and enlarged cytoplasmic localization of MAPK phosphatase-1 (MKP-1) upon PARP-1 inhibition in oxidative stress because changes in MKP-1 expression were reflected in the phosphorylation states of JNK and p38. Furthermore, we found that in MKP-1-silenced cells, PARP inhibition was unable to exert its protective effect, indicating the pivotal roles of JNK and p38 in mediating the oxidative-stress-induced cell death as well as that of increased MKP-1 expression in mediating the protective effect of PARP inhibition. We suggest that regulation of a protein that can directly influence cytoplasmic signaling cascades at the expression level represents a novel mechanism for the cytoplasmic action of PARP-1 inhibition.


Neuropeptides | 2011

Mice deficient in pituitary adenylate cyclase activating polypeptide (PACAP) show increased susceptibility to in vivo renal ischemia/reperfusion injury

Peter Szakaly; E. Laszlo; Krisztina Kovacs; Boglarka Racz; Gabriella Horvath; Andrea Ferencz; Andrea Lubics; Peter Kiss; Andrea Tamas; R. Brubel; Balazs Opper; Akemichi Baba; Hitoshi Hashimoto; Jozsef Farkas; Attila Matkovits; Tamás Magyarlaki; Zsuzsanna Helyes; Dora Reglodi

Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide with well-known cytoprotective effects. We have reported earlier that PACAP decreases mortality and the degree of tubular atrophy in a rat model of renal ischemia/reperfusion injury. Recently, we have shown that kidney cultures isolated from PACAP deficient mice show increased susceptibility to renal oxidative stress. Based on these previous studies, we raised the question whether PACAP deficient mice display increased sensitivity to in vivo kidney ischemia/reperfusion. PACAP⁻/⁻ mice underwent 45 or 60 min of renal ischemia followed by 2 weeks reperfusion. Kidneys were processed for histological analysis. Sections stained with PAS-haematoxylin were graded for the following parameters: degree of tubular dilation, Bowmanns capsule dilation, lymphocyte and macrophage infiltration, thyroidization and the disappearance of the PAS-positive glycocalyx from under the brush border. In other sets of experiments, tissue cytokine expression and the level of the endogenous antioxidant superoxide dismutase (SOD) were also determined after 60 min ischemia/reperfusion. Our results show that while intact kidneys were not different between wild-type and PACAP deficient mice, marked differences were observed in the histological structures in groups that underwent ischemia/reperfusion. PACAP deficient mice had a worse histological outcome, with significantly higher histological scores for all tested parameters. Cytokine expression was also markedly different between wild-type and PACAP deficient mice. In addition, the level of SOD was significantly lower in PACAP⁻/⁻ animals after ischemia/reperfusion. In conclusion, the lack of endogenous PACAP leads to higher susceptibility to in vivo renal ischemia/reperfusion, suggesting that PACAP has an endogenous renoprotective effect.


International Journal of Molecular Sciences | 2010

Novel Neuroprotective Strategies in Ischemic Retinal Lesions

Krisztina Szabadfi; Laszlo Mester; Dora Reglodi; Peter Kiss; Norbert Babai; Boglarka Racz; Krisztina Kovacs; Aliz Szabo; Andrea Tamas; Robert Gábriel; Tamas Atlasz

Retinal ischemia can be effectively modeled by permanent bilateral common carotid artery occlusion, which leads to chronic hypoperfusion-induced degeneration in the entire rat retina. The complex pathways leading to retinal cell death offer a complex approach of neuroprotective strategies. In the present review we summarize recent findings with different neuroprotective candidate molecules. We describe the protective effects of intravitreal treatment with: (i) urocortin 2; (ii) a mitochondrial ATP-sensitive K+ channel opener, diazoxide; (iii) a neurotrophic factor, pituitary adenylate cyclase activating polypeptide; and (iv) a novel poly(ADP-ribose) polymerase inhibitor (HO3089). The retinoprotective effects are demonstrated with morphological description and effects on apoptotic pathways using molecular biological techniques.


Journal of Leukocyte Biology | 2011

Suppressing LPS-induced early signal transduction in macrophages by a polyphenol degradation product: a critical role of MKP-1

Zsuzsanna Tucsek; Balázs Radnai; Boglarka Racz; Balazs Debreceni; Janos K. Priber; Tamas Dolowschiak; Tamas Palkovics; Ferenc Gallyas; Balazs Sumegi; Balazs Veres

Macrophages represent the first defense line against bacterial infection and therefore, play a crucial role in early inflammatory response. In this study, we investigated the role of MAPKs and MKP‐1 activation in regulation of an early inflammatory response in RAW 264.7 macrophage cells. We induced the inflammatory response by treating the macrophages with LPS and inhibited an early inflammatory response by using ferulaldehyde, a water‐soluble end‐product of dietary polyphenol degradation that we found previously to exert its beneficial anti‐inflammatory effects during the early phase of in vivo inflammation. We found that LPS‐induced ROS and nitrogen species formations were reduced by ferulaldehyde in a concentration‐dependent manner, and ferulaldehyde protected mitochondria against LPS‐induced rapid and massive membrane depolarization. LPS induced early suppression of MKP‐1, which was accompanied by activation of JNK, ERK, and p38 MAPK. By reversing LPS‐induced early suppression of MKP‐1, ferulaldehyde diminished MAPK activation, thereby inhibiting NF‐κB activation, mitochondrial depolarization, and ROS production. Taken together, our data suggest that ferulaldehyde exerts its early anti‐inflammatory effect by preserving the mitochondrial membrane integrity and shifting the expression of MKP‐1 forward in time in macrophages.

Collaboration


Dive into the Boglarka Racz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge