Network


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

Hotspot


Dive into the research topics where Csaba Tömböly is active.

Publication


Featured researches published by Csaba Tömböly.


Peptides | 2002

In vitro quantitative study of the degradation of endomorphins.

Csaba Tömböly; Antal Péter; Géza Tóth

The catabolism of the endomorphins was investigated in detail. The endomorphins were degraded relatively slowly in the rat brain homogenate (t1/2(endomorphin-1)=4.94 min; t1/2(endomorphin-2)=3.81 min). The inhibition of metalloproteases and aminopeptidases stabilised the endomorphins to the greatest extent. The digestion of endomorphins tritiated specifically on Tyr(1), Pro(2) or Phe(3) established also that only the aminopeptidase pathways were essential for inactivation of the endomorphins, and that the tetrapeptides were degraded by cleavage of the Pro(2)-Trp(3) or Pro(2)-Phe(3) bond. The end-products of the catabolism were amino acids; the fragments Tyr-Pro-OH and Pro-Trp-Phe-NH2 were present as intermediates. Metabolites produced by brain carboxypeptidases were not detected.


Life Sciences | 1999

Antinociceptive effects of intrathecal endomorphin-1 and -2 in rats

Gyöngyi Horváth; Margit Szikszay; Csaba Tömböly; György Benedek

Endomorphin-1 and endomorphin-2 were recently postulated to be endogenous mu-opioid receptor agonists. We have investigated the antinociceptive and antihyperalgesic effects of intrathecally administered endomorphins in cumulative doses (0.1-100 microg) on acute and inflammatory pain sensations in awake rats. In the tail-flick test, both peptides caused a dose-dependent short-lasting antinociception, except at the highest dose, which caused motor impairment also. The dose-response curves revealed the development of acute tolerance (tachyphylaxis) to endomorphin. Similarly in the carrageenan-injected paw, the endomorphins (10 microg) exerted transient antinociceptive effects. These are the first data to demonstrate decreased responsivity in models of both acute and inflammatory pain after intrathecal administration of endomorphin-1 and -2 in awake rats.


Journal of Chromatography A | 1999

Liquid chromatographic study of the enzymatic degradation of endomorphins, with identification by electrospray ionization mass spectrometry.

Antal Péter; Géza Tóth; Csaba Tömböly; Georges Laus; Dirk Tourwé

The recently discovered native endomorphins play an important role in opioid analgesia, but their metabolic fate in the organism remains relatively little known. This paper describes the application of high-performance liquid chromatography combined with electrospray ionization mass spectrometry to identify the degradation products resulting from the incubation of endomorphins with proteolytic enzymes. The native endomorphin-1, H-Tyr-Pro-Trp-Phe-NH2 (1), and endomorphin-2, H-Tyr-Pro-Phe-Phe-NH2 (2), and an analog of endomorphin-2, H-Tyr-Pro-Phe-Phe-OH (3), were synthetized, and the levels of their resistance against carboxypeptidase A, carboxypeptidase Y, aminopeptidase M and proteinase A were determined. The patterns of peptide metabolites identified by this method indicated that carboxypeptidase Y first hydrolyzes the C-terminal amide group to a carboxy group, and then splits the peptides at the Trp3-Phe4 or Phe3-Phe4 bond. The remaining fragment peptides are stable against the enzymes investigated. Carboxypeptidase A degrades only analog 3 at the Phe3-Phe4 bond. Aminopeptidase M cleaves the peptides at the Pro2-Trp3 or Pro2-Phe3 bond. The C-terminal fragments hydrolyze further, giving amino acids and Phe-NH2-s while the N-terminal part displays a resistance to further aminopeptidase M digestion. Proteinase A exhibits a similar effect to carboxypeptidase Y: the C-terminal amide group is first converted to a carboxy group, and one amino acid is then split off from the C-terminal side.


European Journal of Neuroscience | 2000

Specific activation of the μ opioid receptor (MOR) by endomorphin 1 and endomorphin 2

Krisztina Monory; Marie Claude Bourin; Mariana Spetea; Csaba Tömböly; Géza Tóth; Hans W.D Matthes; Brigitte L. Kieffer; Jacques Hanoune; Anna Borsodi

The recently discovered endomorphin 1 (Tyr‐Pro‐Trp‐Phe‐NH2) and endomorphin 2 (Tyr‐Pro‐Phe‐Phe‐NH2) were investigated with respect to their direct receptor‐binding properties, and to their ability to activate G proteins and to inhibit adenylyl cyclase in both cellular and animal models. Both tetrapeptides activated G proteins and inhibited adenylyl cyclase activity in membrane preparations from cells stably expressing the μ opioid receptor, an effect reversed by the μ receptor antagonist CTAP (d‐Phe‐Cys‐Tyr‐d‐Trp‐Arg‐Thr‐Pen‐Thr‐NH2), but they had no influence on cells stably expressing the δ opioid receptor. To further establish the selectivity of these peptides for the μ opioid receptor, brain preparations of mice lacking the μ opioid receptor gene were used to study their binding and signalling properties. Endomorphin 2, tritiated by a dehalotritiation method resulting in a specific radioactivity of 1.98 TBq/mmol (53.4 Ci/mmol), labelled the brain membranes of wild‐type mice with a Kd value of 1.77 nm and a Bmax of 63.33 fmol/mg protein. In membranes of mice lacking the μreceptor gene, no binding was observed, and both endomorphins failed to stimulate [35S]guanosine‐5′‐O‐(3‐thio)triphosphate ([35S]GTPγS) binding and to inhibit adenylyl cyclase. These data show that endomorphins are capable of activating G proteins and inhibiting adenylyl cyclase activity, and all these effects are mediated by the μ opioid receptors.


Journal of Medicinal Chemistry | 2008

New endomorphin analogues containing alicyclic β-amino acids: Influence on bioactive conformation and pharmacological profile

Attila Keresztes; Mária Szücs; Attila Borics; Katalin E. Kövér; Eniko Forró; Ferenc Fülöp; Csaba Tömböly; Antal Péter; Annamária Páhi; Gabriella Fábián; Mariann Murányi; Géza Tóth

Endomorphins were subjected to a number of structural modifications in a search for their bioactive conformations. The alicyclic beta-amino acids cis-(1 S,2 R)ACPC/ACHC, cis-(1 R,2 S)ACPC/ACHC, trans-(1 S,2 S)ACPC/ACHC, and trans-(1 R,2 R)ACPC/ACHC were introduced into endomorphins to examine the conformational effects on the bioactivity. Use of a combination of receptor binding techniques, (1)H NMR, and molecular modeling allowed the conclusion that Pro (2) substitution by these residues causes changes in structure, proteolytic stability, and pharmacological activity. It seems that the size of the alicyclic beta-amino acids does not have marked influence on the receptor binding affinities and/or selectivities. Among the new analogues, the cis-(1 S,2 R)ACPC (2) and cis-(1 S,2 R)ACHC (2)-containing derivatives displayed the highest binding potencies and efficacies in receptor binding and ligand-stimulated [ (35)S]GTPgammaS functional experiments. Molecular dynamic simulations and (1)H NMR studies of the cis-ACPC/ACHC-containing analogues revealed that many conformations are accessible, though it is most likely that these peptides bind to the mu-opioid receptor in a compact, folded structure rather than extended.


Peptides | 2006

Endomorphin-1 and endomorphin-2 differentially interact with specific binding sites for substance P (SP) aminoterminal SP1–7 in the rat spinal cord

Milad Botros; Mathias Hallberg; Tobias Johansson; Qin Zhou; Gunnar Lindeberg; Per Anders Frändberg; Csaba Tömböly; Géza Tóth; Pierre Le Grevès; Fred Nyberg

Endomorphin-1 (EM-1) and endomorphin-2 (EM-2) represent two opioid active tetrapeptides with high affinity and selectivity for the mu-opioid (MOP) receptor. Both EM-1 and EM-2 exhibit strong inhibition of pain signals in the central nervous system (CNS). In contrast to these compounds, the undecapeptide substance P (SP) facilitates pain influx in the CNS. SP has been implicated in a number of functions in the central nervous system, including pain processing and reward. Its aminoterminal fragment SP1-7 has been shown to modulate several actions of SP in the CNS, the nociceptive effect included. Although the actions of SP1-7 have been known for long no specific receptor for the SP fragment has yet been cloned. In this study, we demonstrate the presence of specific binding sites for the heptapeptide in the rat spinal cord. The binding affinity for unlabeled SP1-7 to the specific sites for the labeled heptapeptide highly exceeded those of SP and other C- or N-terminal fragments thereof. The NK-1, NK-2 and NK-3 receptor ligands [Sar9, Met(O2)11]SP, R396 and senktide, respectively, showed no or negligible binding. Moreover, both EM-1 and EM-2 were found to interact with SP1-7 binding. However, a significant difference in binding affinity between the two opioid active tetrapeptides was observed. As recorded from replacement curves the affinity of EM-2 was 10 times weaker than that for SP1-7 but about 100 times higher than that of EM-1. Among other Tyr-Pro-containing peptides Tyr-MIF-1 but not Tyr-W-MIF-1 exhibited affinity of similar potency as EM-2. These results strengthen the previously observed differences between EM-1 and EM-2 in various functional studies. Moreover, using a cell line (C6) expressing the MOP receptor it was shown that the labeled SP1-7 did not interact with binding to this receptor and no functional response was seen for the SP heptapeptide on the MOP receptor by means of stimulation in the GTPgammaS assay. This suggests that the identified SP1-7 binding sites, with high affinity also for EM-2, are not identical to the MOP receptor and apparently not to any of the known tachykinin receptors.


Peptides | 2001

Behavioral and neuroendocrine actions of endomorphin-2

Erika Bujdosó; Miklós Jászberényi; Csaba Tömböly; Géza Tóth; Gyula Telegdy

The effects of intracerebroventricularly administered endomorphin-2 (EM2) on open-field activity and the hypothalamo-pituitary-adrenal (HPA) system were investigated. EM2 (0.25-1 microg) significantly increased both the locomotor and the rearing activity, resulting in a bell-shaped dose-response curve. EM2 also enhanced corticosterone release, with an even more profound downturn phase at higher concentrations. The corticotropin-releasing hormone (CRH) antagonist alpha-helical CRH9-41 completely abolished the EM2-evoked endocrine and behavioral responses. These findings reinforce the hypothesis that the endomorphins may play a significant role in the regulation of locomotion, rearing activity and the HPA system through the release of CRH.


Peptides | 2008

Endomorphins interact with the substance P (SP) aminoterminal SP1–7 binding in the ventral tegmental area of the rat brain

Milad Botros; Tobias Johansson; Qin Zhou; Gunnar Lindeberg; Csaba Tömböly; Géza Tóth; Pierre Le Grevès; Fred Nyberg; Mathias Hallberg

We have recently identified a specific binding site for the tachykinin peptide substance P (SP) fragment SP(1-7) in the rat spinal cord. This site appeared very specific for SP(1-7) as the binding affinity of this compound highly exceeded those of other SP fragments. We also observed that endomorphin-2 (EM-2) exhibited high potency in displacing SP(1-7) from this site. In the present work using a [(3)H]-labeled derivative of the heptapeptide we have identified and characterized [(3)H]-SP(1-7) binding in the rat ventral tegmental area (VTA). Similarly to the [(3)H]-SP(1-7) binding in the spinal cord the affinity of unlabeled SP(1-7) to the specific site in VTA was significantly higher than those of other SP fragments. Further, the tachykinin receptor NK-1, NK-2 and NK-3 ligands showed no or negligible binding to the identified site. However, the mu-opioid peptide (MOP) receptor agonists DAMGO, EM-1 and EM-2 did, and significant difference was observed in the binding affinity between the two endomorphins. As recorded from displacement curves the affinity of EM-2 for the SP(1-7) site was 4-5 times weaker than that for SP(1-7) but about 5 times higher than that of EM-1. The opioid receptor antagonists naloxone and naloxonazine showed weak or negligible binding. It was concluded that the specific site identified for SP(1-7) binding in the rat VTA is distinct from the MOP receptor although it exhibits high affinity for EM-2.


Endocrine | 2001

Effects of endomorphin-1 on open-field behavior and on the hypothalamic-pituitary-adrenal system.

Erika Bujdosó; Miklós Jászberényi; Csaba Tömböly; Géza Tóth; Gyula Telegdy

The effects of endomorphin-1 (EM1) on behavioral responses and on the hypothalamic-pituitary-adrenal system were investigated in mice. Locomotor activity was measured in an “open-field” apparatus, with parallel recording of the numbers of rearings and groomings. Different doses of the peptide (250 ng to 5 µg) were administered to the animals intracerebroventricularly 30 min before the tests. EM1 caused significant increases in the locomotor activity and the number of rearings. The effect of EM1 on the basal corticosterone secretion was also investigated. At a dose of 5 µg, the peptide significantly increased plasma corticosterone level. The corticotropin-releasing hormone (CRH) antagonist α-helical CRH9–41, applied 30 min prior to EM1 administration, completely abolished the increases in both locomotion and the number of rearings and attenuated the corticosterone release evoked by EM1. These results suggest that the EM1-induced increases in locomotion and rearing activity as well as the pituitary-adrenal activation are mediated by CRH.


Neuropharmacology | 2005

Autoradiography in opioid triple knockout mice reveals opioid and opioid receptor like binding of naloxone benzoylhydrazone

Veronica Cox; Siân Clarke; Tracy Czyzyk; Micheal Ansonoff; Joshua Nitsche; Ming Sing Hsu; Anna Borsodi; Csaba Tömböly; Géza Tóth; R.G. Hill; John E. Pintar; Ian Kitchen

Naloxone benzoylhydrazone (NalBzoH) is a ligand used to study opioid receptors. It has been suggested to act at a novel kappa3 receptor but also appears to bind to classical opioid receptors, and possibly the ORL1 receptor. We have used opioid receptor triple knockout mice, deficient in genes coding for the mu, delta and kappa-receptor, to characterise the relative contributions of opioid and ORL1 activity to the binding of this ligand, by carrying out receptor autoradiography with [3H]NalBzoH. As competing ligands we have used diprenorphine and nociceptin at 1 microM, alone or in combination, to determine the contribution of opioid and ORL1 receptor binding. At 4 nM [3H]NalBzoH showed labelling in wild-type brains indicative of broad spectrum classical opioid receptor binding. In the triple knockout brains all labelling was completely absent, suggesting that at this concentration there is no binding to ORL1 sites. However at 50 nM [3H]NalBzoH showed labelling in triple knockout brains with a distribution pattern indicative of ORL1 labelling. Quantitative analysis showed that nociceptin displaced typically 30% of the residual labelling in knockout brains whilst diprenorphine had relatively little effect. The data show that at 50 nM NalBzoH no binding was detected other than to classical opioid receptors or to ORL1 in an approximate ratio of 2:1.

Collaboration


Dive into the Csaba Tömböly's collaboration.

Top Co-Authors

Avatar

Géza Tóth

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Sándor Benyhe

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Anna Borsodi

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Szabolcs Dvorácskó

Hungarian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Dirk Tourwé

Vrije Universiteit Brussel

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Janecka

Medical University of Łódź

View shared research outputs
Top Co-Authors

Avatar

Adriano Mollica

University of Chieti-Pescara

View shared research outputs
Top Co-Authors

Avatar

Azzurra Stefanucci

University of Chieti-Pescara

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge