Network


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

Hotspot


Dive into the research topics where Sonja Sucic is active.

Publication


Featured researches published by Sonja Sucic.


Journal of Biological Chemistry | 2010

The N Terminus of Monoamine Transporters Is a Lever Required for the Action of Amphetamines

Sonja Sucic; Stefan Dallinger; Barbara Zdrazil; René Weissensteiner; Trine N. Jørgensen; Marion Holy; Oliver Kudlacek; Stefan Seidel; Joo Hee Cha; Ulrik Gether; Amy Hauck Newman; Gerhard F. Ecker; Michael Freissmuth; Harald H. Sitte

The serotonin transporter (SERT) terminates neurotransmission by removing serotonin from the synaptic cleft. In addition, it is the site of action of antidepressants (which block the transporter) and of amphetamines (which induce substrate efflux). We explored the functional importance of the N terminus in mediating the action of amphetamines by focusing initially on the highly conserved threonine residue at position 81, a candidate site for phosphorylation by protein kinase C. Molecular dynamics simulations of the wild type SERT, compared with its mutations SERTT81A and SERTT81D, suggested structural changes in the inner vestibule indicative of an opening of the inner vestibule. Predictions from this model (e.g. the preferential accumulation of SERTT81A in the inward conformation, its reduced turnover number, and a larger distance between its N and C termini) were verified. Most importantly, SERTT81A (and the homologous mutations in noradrenaline and dopamine) failed to support amphetamine-induced efflux, and this was not remedied by aspartate at this position. Amphetamine-induced currents through SERTT81A were comparable with those through the wild type transporter. Both abundant Na+ entry and accumulation of SERTT81A in the inward facing conformation ought to favor amphetamine-induced efflux. Thus, we surmised that the N terminus must play a direct role in driving the transporter into a state that supports amphetamine-induced efflux. This hypothesis was verified by truncating the first 64 amino acids and by tethering the N terminus to an additional transmembrane helix. Either modification abolished amphetamine-induced efflux. We therefore conclude that the N terminus of monoamine transporters acts as a lever that sustains reverse transport.


Journal of Biological Chemistry | 2011

The Serotonin Transporter Is an Exclusive Client of the Coat Protein Complex II (COPII) Component SEC24C

Sonja Sucic; Ali El-Kasaby; Oliver Kudlacek; Subhodeep Sarker; Harald H. Sitte; Philippe Marin; Michael Freissmuth

The transporters for serotonin (SERT), dopamine, and noradrenaline have a conserved hydrophobic core but divergent N and C termini. The C terminus harbors the binding site for the coat protein complex II (COPII) cargo-binding protein SEC24. Here we explored which SEC24 isoform was required for export of SERT from the endoplasmic reticulum (ER). Three lines of evidence argue that SERT can only exit the ER by recruiting SEC24C: (i) Mass spectrometry showed that a peptide corresponding to the C terminus of SERT recruited SEC24C-containing COPII complexes from mouse brain lysates. (ii) Depletion of individual SEC24 isoforms by siRNAs revealed that SERT was trapped in the ER only if SEC24C was down-regulated, in both, cells that expressed SERT endogenously or after transfection. The combination of all siRNAs was not more effective than that directed against SEC24C. A SERT mutant in which the SEC24C-binding motif (607RI608) was replaced by alanine was insensitive to down-regulation of SEC24C levels. (iii) Overexpression of a SEC24C variant with a mutation in the candidate cargo-binding motif (SEC24C-D796V/D797N) but not of the corresponding mutant SEC24D-D733V/D734N reduced SERT surface levels. In contrast, noradrenaline and dopamine transporters and the more distantly related GABA transporter 1 relied on SEC24D for ER export. These observations demonstrate that closely related transporters are exclusive client cargo proteins for different SEC24 isoforms. The short promoter polymorphism results in reduced SERT cell surface levels and renders affected individuals more susceptible to depression. By inference, variations in the Sec24C gene may also affect SERT cell surface levels and thus be linked to mood disorders.


Molecular Pharmacology | 2010

The High-Affinity Binding Site for Tricyclic Antidepressants Resides in the Outer Vestibule of the Serotonin Transporter

Subhodeep Sarker; René Weissensteiner; Ilka Steiner; Harald H. Sitte; Gerhard F. Ecker; Michael Freissmuth; Sonja Sucic

The structure of the bacterial leucine transporter from Aquifex aeolicus (LeuTAa) has been used as a model for mammalian Na+/Cl−-dependent transporters, in particular the serotonin transporter (SERT). The crystal structure of LeuTAa liganded to tricyclic antidepressants predicts simultaneous binding of inhibitor and substrate. This is incompatible with the mutually competitive inhibition of substrates and inhibitors of SERT. We explored the binding modes of tricyclic antidepressants by homology modeling and docking studies. Two approaches were used subsequently to differentiate between three clusters of potential docking poses: 1) a diagnostic SERTY95F mutation, which greatly reduced the affinity for [3H]imipramine but did not affect substrate binding; 2) competition binding experiments in the presence and absence of carbamazepine (i.e., a tricyclic imipramine analog with a short side chain that competes with [3H]imipramine binding to SERT). Binding of releasers (para-chloroamphetamine, methylene-dioxy-methamphetamine/ecstasy) and of carbamazepine were mutually exclusive, but Dixon plots generated in the presence of carbamazepine yielded intersecting lines for serotonin, MPP+, paroxetine, and ibogaine. These observations are consistent with a model, in which 1) the tricyclic ring is docked into the outer vestibule and the dimethyl-aminopropyl side chain points to the substrate binding site; 2) binding of amphetamines creates a structural change in the inner and outer vestibule that precludes docking of the tricyclic ring; 3) simultaneous binding of ibogaine (which binds to the inward-facing conformation) and of carbamazepine is indicative of a second binding site in the inner vestibule, consistent with the pseudosymmetric fold of monoamine transporters. This may be the second low-affinity binding site for antidepressants.


Journal of Biological Chemistry | 2012

Ca2+/Calmodulin-dependent Protein Kinase IIα (αCaMKII) Controls the Activity of the Dopamine Transporter IMPLICATIONS FOR ANGELMAN SYNDROME

Thomas Steinkellner; Jae-Won Yang; Therese Montgomery; Wei-Qiang Chen; Marie-Therese Winkler; Sonja Sucic; Gert Lubec; Michael Freissmuth; Ype Elgersma; Harald H. Sitte; Oliver Kudlacek

Background: αCaMKII modulates amphetamine-induced dopamine transporter-mediated substrate efflux. Results: Mice with ablated or blunted αCaMKII function show decreased amphetamine-triggered efflux. Conclusion: Dopamine transporter function is impaired in mice with targeted αCaMKII mutations and in a mouse model of the Angelman syndrome. Significance: Such new insights into dopamine transporter function may further illuminate the complex pathophysiology of the Angelman syndrome. The dopamine transporter (DAT) is a crucial regulator of dopaminergic neurotransmission, controlling the length and brevity of dopaminergic signaling. DAT is also the primary target of psychostimulant drugs such as cocaine and amphetamines. Conversely, methylphenidate and amphetamine are both used clinically in the treatment of attention-deficit hyperactivity disorder and narcolepsy. The action of amphetamines, which induce transport reversal, relies primarily on the ionic composition of the intra- and extracellular milieus. Recent findings suggest that DAT interacting proteins may also play a significant role in the modulation of reverse dopamine transport. The pharmacological inhibition of the serine/threonine kinase αCaMKII attenuates amphetamine-triggered DAT-mediated 1-methyl-4-phenylpyridinium (MPP+) efflux. More importantly, αCaMKII has also been shown to bind DAT in vitro and is therefore believed to be an important player within the DAT interactome. Herein, we show that αCaMKII co-immunoprecipitates with DAT in mouse striatal synaptosomes. Mice, which lack αCaMKII or which express a permanently self-inhibited αCaMKII (αCaMKIIT305D), exhibit significantly reduced amphetamine-triggered DAT-mediated MPP+ efflux. Additionally, we investigated mice that mimic a neurogenetic disease known as Angelman syndrome. These mice possess reduced αCaMKII activity. Angelman syndrome mice demonstrated an impaired DAT efflux function, which was comparable with that of the αCaMKII mutant mice, indicating that DAT-mediated dopaminergic signaling is affected in Angelman syndrome.


Journal of Biological Chemistry | 2013

Switching the Clientele A LYSINE RESIDING IN THE C TERMINUS OF THE SEROTONIN TRANSPORTER SPECIFIES ITS PREFERENCE FOR THE COAT PROTEIN COMPLEX II COMPONENT SEC24C

Sonja Sucic; Florian Koban; Ali El-Kasaby; Oliver Kudlacek; Thomas Stockner; Harald H. Sitte; Michael Freissmuth

Background: The serotonin transporter (SERT) relies exclusively on SEC24 paralog C for its ER export. Results: A lysine to tyrosine mutation in the C terminus of SERT switches its preference from SEC24C to SEC24D. Conclusion: The position +2 from the RI/RL/KL export motif determines SEC24 paralog requirement. Significance: The role of SEC24C in ER export of neurotransmitter transporters is relevant to psychiatric disorders, e.g., bipolar disease and depression. The serotonin transporter (SERT) maintains serotonergic neurotransmission via rapid reuptake of serotonin from the synaptic cleft. SERT relies exclusively on the coat protein complex II component SEC24C for endoplasmic reticulum (ER) export. The closely related transporters for noradrenaline and dopamine depend on SEC24D. Here, we show that discrimination between SEC24C and SEC24D is specified by the residue at position +2 downstream from the ER export motif (607RI608 in SERT). Substituting Lys610 with tyrosine, the corresponding residue found in the noradrenaline and dopamine transporters, switched the SEC24 isoform preference: SERT-K610Y relied solely on SEC24D to reach the cell surface. This analysis was extended to other SLC6 (solute carrier 6) transporter family members: siRNA-dependent depletion of SEC24C, but not of SEC24D, reduced surface levels of the glycine transporter-1a, the betaine/GABA transporter and the GABA transporter-4. Experiments with dominant negative versions of SEC24C and SEC24D recapitulated these findings. We also verified that the presence of two ER export motifs (in concatemers of SERT and GABA transporter-1) supported recruitment of both SEC24C and SEC24D. To the best of our knowledge, this is the first report to document a change in SEC24 specificity by mutation of a single residue in the client protein. Our observations allowed for deducing a rule for SLC6 family members: a hydrophobic residue (Tyr or Val) in the +2 position specifies interaction with SEC24D, and a hydrophilic residue (Lys, Asn, or Gln) recruits SEC24C. Variations in SEC24C are linked to neuropsychiatric disorders. The present findings provide a mechanistic explanation. Variations in SEC24C may translate into distinct surface levels of neurotransmitter transporters.


The FASEB Journal | 2012

The cysteines of the extracellular loop are crucial for trafficking of human organic cation transporter 2 to the plasma membrane and are involved in oligomerization

Sabine Brast; Alexander Grabner; Sonja Sucic; Harald H. Sitte; Edwin Hermann; Hermann Pavenstädt; Eberhard Schlatter; Giuliano Ciarimboli

Human organic cation transporter 2 (hOCT2) is involved in transport of many endogenous and exogenous organic cations, mainly in kidney and brain cells. Because the quaternary structure of transmembrane proteins plays an essential role for their cellular trafficking and function, we investigated whether hOCT2 forms oligomeric complexes, and if so, which part of the transporter is involved in the oligomerization. A yeast 2‐hybrid mating‐based split‐ubiquitin system (mbSUS), fluorescence resonance energy transfer, Western blot analysis, crosslinking experiments, immunofluorescence, and uptake measurements of the fluorescent organic cation 4‐(4‐(dimethylamino) styryl)‐N‐methylpyridinium were applied to human embryonic kidney 293 (HEK293) cells transfected with hOCT2 and partly also to freshly isolated human proximal tubules. The role of cysteines for oligomerization and trafficking of the transporter to the plasma membranes was investigated in cysteine mutants of hOCT2. hOCT2 formed oligomers both in the HEK293 expression system and in native human kidneys. The cysteines of the large extracellular loop are important to enable correct folding, oligomeric assembly, and plasma membrane insertion of hOCT2. Mutation of the first and the last cysteines of the loop at positions 51 and 143 abolished oligomer formation. Thus, the cysteines of the extracellular loop are important for correct trafficking of the transporter to the plasma membrane and for its oligomerization.—Brast, S., Grabner, A., Sucic, S., Sitte, H. H., Hermann, E., Pavenstädt, H., Schlatter, E., and Ciarimboli, G. The cysteines of the extracellular loop are crucial for trafficking of human organic cation transporter 2 to the plasma membrane and are involved in oligomerization. FASEB J. 26, 976‐986 (2012). www.fasebj.org


Scientific Reports | 2015

STAT3 controls IL6-dependent regulation of serotonin transporter function and depression-like behavior

Eryan Kong; Sonja Sucic; Francisco J. Monje; Giorgia Savalli; Weifei Diao; Deeba Khan; Marianne Ronovsky; Maureen Cabatic; Florian Koban; Michael Freissmuth; Daniela D. Pollak

Experimental evidence suggests a role for the immune system in the pathophysiology of depression. A specific involvement of the proinflammatory cytokine interleukin 6 (IL6) in both, patients suffering from the disease and pertinent animal models, has been proposed. However, it is not clear how IL6 impinges on neurotransmission and thus contributes to depression. Here we tested the hypothesis that IL6-induced modulation of serotonergic neurotransmission through the STAT3 signaling pathway contributes to the role of IL6 in depression. Addition of IL6 to JAR cells, endogenously expressing SERT, reduced SERT activity and downregulated SERT mRNA and protein levels. Similarly, SERT expression was reduced upon IL6 treatment in the mouse hippocampus. Conversely, hippocampal tissue of IL6-KO mice contained elevated levels of SERT and IL6-KO mice displayed a reduction in depression-like behavior and blunted response to acute antidepressant treatment. STAT3 IL6-dependently associated with the SERT promoter and inhibition of STAT3 blocked the effect of IL6 in-vitro and modulated depression-like behavior in-vivo. These observations demonstrate that IL6 directly controls SERT levels and consequently serotonin reuptake and identify STAT3-dependent regulation of SERT as conceivable neurobiological substrate for the involvement of IL6 in depression.


Journal of Biological Chemistry | 2015

A Salt Bridge Linking the First Intracellular Loop with the C Terminus Facilitates the Folding of the Serotonin Transporter

Florian Koban; Ali El-Kasaby; Cornelia Häusler; Thomas Stockner; Benedikt M. Simbrunner; Harald H. Sitte; Michael Freissmuth; Sonja Sucic

Background: The C terminus of the serotonin transporter (SERT) is required for folding. Results: Replacing C-terminal residues (Phe604, Ile608, and Ile612 by Gln and Glu615 by Lys) caused misfolding of SERT. A charge reversal (R152E) rescued SERT-E615K. Conclusion: An amphipathic C-terminal helix interacts with the first intracellular loop to facilitate folding of SERT. Significance: These data provide insights into the folding trajectory of SERT and related transporters. The folding trajectory of solute carrier 6 (SLC6) family members is of interest because point mutations result in misfolding and thus cause clinically relevant phenotypes in people. Here we examined the contribution of the C terminus in supporting folding of the serotonin transporter (SERT; SLC6A4). Our working hypothesis posited that the amphipathic nature of the C-terminal α-helix (Thr603–Thr613) was important for folding of SERT. Accordingly, we disrupted the hydrophobic moment of the α-helix by replacing Phe604, Ile608, or Ile612 by Gln. The bulk of the resulting mutants SERT-F604Q, SERT-I608Q, and SERT-I612Q were retained in the endoplasmic reticulum, but their residual delivery to the cell surface still depended on SEC24C. This indicates that the amphipathic nature of the C-terminal α-helix was dispensable to endoplasmic reticulum export. The folding trajectory of SERT is thought to proceed through the inward facing conformation. Consistent with this conjecture, cell surface expression of the misfolded mutants was restored by (i) introducing second site suppressor mutations, which trap SERT in the inward facing state, or (ii) by the pharmacochaperone noribogaine, which binds to the inward facing conformation. Finally, mutation of Glu615 at the end of the C-terminal α-helix to Lys reduced surface expression of SERT-E615K. A charge reversal mutation in intracellular loop 1 restored surface expression of SERT-R152E/E615K to wild type levels. These observations support a mechanistic model where the C-terminal amphipathic helix is stabilized by an intramolecular salt bridge between residues Glu615 and Arg152. This interaction acts as a pivot in the conformational search associated with folding of SERT.


Journal of Biological Chemistry | 2014

A cytosolic relay of heat shock proteins HSP70-1A and HSP90β monitors the folding trajectory of the serotonin transporter.

Ali El-Kasaby; Florian Koban; Harald H. Sitte; Michael Freissmuth; Sonja Sucic

Background: Naturally occurring mutations in solute carrier 6 (SLC6) family members impair folding of these transporters. Results: Folding-deficient SERT mutants bound heat shock protein (HSP)70-1A, HSP90β, and co-chaperones. Noribogaine synergized with HSP inhibitors in rescuing these SERT mutants. Conclusion: Folding of SERT is assisted by a cytosolic HSP relay. Significance: The folding trajectory of SERT is relevant to folding diseases arising from mutated SLC6 transporters. Mutations in the C terminus of the serotonin transporter (SERT) disrupt folding and export from the endoplasmic reticulum. Here we examined the hypothesis that a cytosolic heat shock protein relay was recruited to the C terminus to assist folding of SERT. This conjecture was verified by the following observations. (i) The proximal portion of the SERT C terminus conforms to a canonical binding site for DnaK/heat shock protein of 70 kDa (HSP70). A peptide covering this segment stimulated ATPase activity of purified HSP70-1A. (ii) A GST fusion protein comprising the C terminus of SERT pulled down HSP70-1A. The interaction between HSP70-1A and SERT was visualized in live cells by Förster resonance energy transfer: it was restricted to endoplasmic reticulum-resident transporters and enhanced by an inhibitor that traps HSP70-1A in its closed state. (iv) Co-immunoprecipitation confirmed complex formation of SERT with HSP70-1A and HSP90β. Consistent with an HSP relay, co-chaperones (e.g. HSC70-HSP90-organizing protein) were co-immunoprecipitated with the stalled mutants SERT-R607A/I608A and SERT-P601A/G602A. (v) Depletion of HSP90β by siRNA or its inhibition increased the cell surface expression of wild type SERT and SERT-F604Q. In contrast, SERT-R607A/I608A and SERT-P601A/G602A were only rendered susceptible to inhibition of HSP70 and HSP90 by concomitant pharmacochaperoning with noribogaine. (vi) In JAR cells, inhibition of HSP90 also increased the levels of SERT, indicating that endogenously expressed transporter was also susceptible to control by HSP90β. These findings support the concept that the folding trajectory of SERT is sampled by a cytoplasmic chaperone relay.


Molecular Pharmacology | 2015

Ligand Selectivity among the Dopamine and Serotonin Transporters Specified by the Forward Binding Reaction.

Peter S. Hasenhuetl; Klaus Schicker; Xaver Koenig; Yang Li; Subhodeep Sarker; Thomas Stockner; Sonja Sucic; Harald H. Sitte; Michael Freissmuth; Walter Sandtner

The membrane transporters for the monoamines serotonin (SERT) and dopamine (DAT) are prominent targets of various psychoactive substances, including competitive inhibitors, such as tricyclic antidepressants, methylphenidate, and cocaine. Upon rapid application of a substrate, SERT and DAT display an inwardly directed current comprised of a peak and a steady-state component. Binding of a competitive inhibitor to the transporter leads to reduction of the peak current amplitude because occupancy of the transporter by an inhibitor prevents the induction of the peak current by the substrate. We show that the inhibitory effect on the peak current can be used to study the association rate constant (kon), dissociation rate constant (koff), and equilibrium dissociation constant (KD) of chemically distinct SERT and DAT inhibitors, with high temporal precision and without the need of high-affinity radioligands as surrogates. We exemplify our approach by measuring the kinetics of cocaine, methylphenidate, and desipramine binding to SERT and DAT. Our analysis revealed that the selectivity of methylphenidate and desipramine for DAT and SERT, respectively, can be accounted for by their rate of association and not by the residence time in their respective binding sites.

Collaboration


Dive into the Sonja Sucic's collaboration.

Top Co-Authors

Avatar

Michael Freissmuth

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Harald H. Sitte

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Ali El-Kasaby

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Oliver Kudlacek

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Florian Koban

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Ameya Kasture

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Subhodeep Sarker

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Thomas Stockner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Walter Sandtner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge