Network


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

Hotspot


Dive into the research topics where Alina Smorodchenko is active.

Publication


Featured researches published by Alina Smorodchenko.


Journal of Experimental Medicine | 2003

Treatment of Relapsing Paralysis in Experimental Encephalomyelitis by Targeting Th1 Cells through Atorvastatin

Orhan Aktas; Sonia Waiczies; Alina Smorodchenko; Jan Dörr; Bibiane Seeger; Timour Prozorovski; Stephanie Sallach; Matthias Endres; Stefan Brocke; Robert Nitsch; Frauke Zipp

Statins, known as inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, exhibit numerous functions related to inflammation, such as MHC class II down-regulation, interference with T cell adhesion, and induction of apoptosis. Here we demonstrate that both subcutaneous and oral administration of atorvastatin inhibit the development of actively induced chronic experimental autoimmune encephalomyelitis in SJL/J mice and significantly reduce the inflammatory infiltration into the central nervous system (CNS). When treatment was started after disease onset, atorvastatin reduced the incidence of relapses and protected from the development of further disability. Both the reduced autoreactive T cell response measured by proliferation toward the encephalitogenic peptide PLP139–151 and the cytokine profile indicate a potent blockade of T helper cell type 1 immune response. In in vitro assays atorvastatin not only inhibited antigen-specific responses, but also decreased T cell proliferation mediated by direct TCR engagement independently of MHC class II and LFA-1. Inhibition of proliferation was not due to apoptosis induction, but linked to a negative regulation on cell cycle progression. However, early T cell activation was unaffected, as reflected by unaltered calcium fluxes. Thus, our results provide evidence for a beneficial role of statins in the treatment of autoimmune attack on the CNS.


Journal of Immunology | 2004

Green Tea Epigallocatechin-3-Gallate Mediates T Cellular NF-κB Inhibition and Exerts Neuroprotection in Autoimmune Encephalomyelitis

Orhan Aktas; Timour Prozorovski; Alina Smorodchenko; Nicolai E. Savaskan; Roland Lauster; Peter-Michael Kloetzel; Carmen Infante-Duarte; Stefan Brocke; Frauke Zipp

Recent studies in multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), point to the fact that even in the early phase of inflammation, neuronal pathology plays a pivotal role in the sustained disability of affected individuals. We show that the major green tea constituent, (−)-epigallocatechin-3-gallate (EGCG), dramatically suppresses EAE induced by proteolipid protein 139–151. EGCG reduced clinical severity when given at initiation or after the onset of EAE by both limiting brain inflammation and reducing neuronal damage. In orally treated mice, we found abrogated proliferation and TNF-α production of encephalitogenic T cells. In human myelin-specific CD4+ T cells, cell cycle arrest was induced, down-regulating the cyclin-dependent kinase 4. Interference with both T cell growth and effector function was mediated by blockade of the catalytic activities of the 20S/26S proteasome complex, resulting in intracellular accumulation of IκB-α and subsequent inhibition of NF-κB activation. Because its structure implicates additional antioxidative properties, EGCG was capable of protecting against neuronal injury in living brain tissue induced by N-methyl-d-aspartate or TRAIL and of directly blocking the formation of neurotoxic reactive oxygen species in neurons. Thus, a natural green tea constituent may open up a new therapeutic avenue for young disabled adults with inflammatory brain disease by combining, on one hand, anti-inflammatory and, on the other hand, neuroprotective capacities.


Neuron | 2005

Neuronal Damage in Autoimmune Neuroinflammation Mediated by the Death Ligand TRAIL

Orhan Aktas; Alina Smorodchenko; Stefan Brocke; Carmen Infante-Duarte; Ulf Schulze Topphoff; Johannes Vogt; Timour Prozorovski; Susanne Meier; Venera Osmanova; Elena E. Pohl; Ingo Bechmann; Robert Nitsch; Frauke Zipp

Here, we provide evidence for a detrimental role of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in neural death in T cell-induced experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Clinical severity and neuronal apoptosis in brainstem motor areas were substantially reduced upon brain-specific blockade of TRAIL after induction of EAE through adoptive transfer of encephalitogenic T cells. Furthermore, TRAIL-deficient myelin-specific lymphocytes showed reduced encephalitogenicity when transferred to wild-type mice. Conversely, intracerebral delivery of TRAIL to animals with EAE increased clinical deficits, while naive mice were not susceptible to TRAIL. Using organotypic slice cultures as a model for living brain tissue, we found that neurons were susceptible to TRAIL-mediated injury induced by encephalitogenic T cells. Thus, in addition to its known immunoregulatory effects, the death ligand TRAIL contributes to neural damage in the inflamed brain.


The Journal of Neuroscience | 2004

Direct Impact of T Cells on Neurons Revealed by Two-Photon Microscopy in Living Brain Tissue

Robert Nitsch; Elena E. Pohl; Alina Smorodchenko; Carmen Infante-Duarte; Orhan Aktas; Frauke Zipp

Encephalitogenic T cells invade the brain during neuroinflammation such as multiple sclerosis (MS), inducing damage to myelin sheaths and oligodendrocytes. Only recently, neuronal structures were reported to be a crucial target in the disease. Here, two-photon microscopy using ion-sensitive dyes revealed that within the complex cellular network of living brain tissue, proteolipid protein (PLP)-specific T cells and T cells recognizing the nonmurine antigen ovalbumin (OVA) directly and independently of the major histocompatibility complex (MHC) contact neurons in which they induce calcium oscillations. T cell contact finally resulted in a lethal increase in neuronal calcium levels. This could be prevented by blocking both perforin and glutamate receptors. For the first time, our data provide direct insight into the activity of T cells in the living brain and their detrimental impact on neurons.


Nature Medicine | 2009

Activation of kinin receptor B1 limits encephalitogenic T lymphocyte recruitment to the central nervous system

Ulf Schulze-Topphoff; Alexandre Prat; Timour Prozorovski; Volker Siffrin; Magdalena Paterka; Josephine Herz; Ivo Bendix; Igal Ifergan; Ines Schadock; Marcelo A. Mori; Jack van Horssen; Friederike Schröter; Alina Smorodchenko; May H. Han; Michael Bader; Lawrence Steinman; Orhan Aktas; Frauke Zipp

Previous proteomic and transcriptional analyses of multiple sclerosis lesions revealed modulation of the renin-angiotensin and the opposing kallikrein-kinin pathways. Here we identify kinin receptor B1 (Bdkrb1) as a specific modulator of immune cell entry into the central nervous system (CNS). We demonstrate that the Bdkrb1 agonist R838 (Sar-[D-Phe]des-Arg9-bradykinin) markedly decreases the clinical symptoms of experimental autoimmune encephalomyelitis (EAE) in SJL mice, whereas the Bdkrb1 antagonist R715 (Ac-Lys-[D-βNal7, Ile8]des-Arg9-bradykinin) resulted in earlier onset and greater severity of the disease. Bdkrb1-deficient (Bdkrb1−/−) C57BL/6 mice immunized with a myelin oligodendrocyte glycoprotein fragment, MOG35–55, showed more severe disease with enhanced CNS-immune cell infiltration. The same held true for mixed bone marrow–chimeric mice reconstituted with Bdkrb1−/− T lymphocytes, which showed enhanced T helper type 17 (TH17) cell invasion into the CNS. Pharmacological modulation of Bdkrb1 revealed that in vitro migration of human TH17 lymphocytes across blood-brain barrier endothelium is regulated by this receptor. Taken together, these results suggest that the kallikrein-kinin system is involved in the regulation of CNS inflammation, limiting encephalitogenic T lymphocyte infiltration into the CNS, and provide evidence that Bdkrb1 could be a new target for the treatment of chronic inflammatory diseases such as multiple sclerosis.


The EMBO Journal | 2014

FGF23 promotes renal calcium reabsorption through the TRPV5 channel

Olena Andrukhova; Alina Smorodchenko; Monika Egerbacher; Carmen Streicher; Ute Zeitz; Regina Goetz; Victoria Shalhoub; Moosa Mohammadi; Elena E. Pohl; Beate Lanske; Reinhold G. Erben

αKlotho is thought to activate the epithelial calcium channel Transient Receptor Potential Vanilloid‐5 (TRPV5) in distal renal tubules through its putative glucuronidase/sialidase activity, thereby preventing renal calcium loss. However, αKlotho also functions as the obligatory co‐receptor for fibroblast growth factor‐23 (FGF23), a bone‐derived phosphaturic hormone. Here, we show that renal calcium reabsorption and renal membrane abundance of TRPV5 are reduced in Fgf23 knockout mice, similar to what is seen in αKlotho knockout mice. We further demonstrate that αKlotho neither co‐localizes with TRPV5 nor is regulated by FGF23. Rather, apical membrane abundance of TRPV5 in renal distal tubules and thus renal calcium reabsorption are regulated by FGF23, which binds the FGF receptor‐αKlotho complex and activates a signaling cascade involving ERK1/2, SGK1, and WNK4. Our data thereby identify FGF23, not αKlotho, as a calcium‐conserving hormone in the kidney.


Stem Cells | 2007

Neural cell adhesion molecule polysialylation enhances the sensitivity of embryonic stem cell-derived neural precursors to migration guidance cues.

Tamara Glaser; Claudia Brose; Isabelle Franceschini; Katja Hamann; Alina Smorodchenko; Frauke Zipp; Monique Dubois-Dalcq; Oliver Brüstle

The development of stem cell‐based neural repair strategies requires detailed knowledge on the interaction of migrating donor cells with the host brain environment. Here we report that overexpression of polysialic acid (PSA), a carbohydrate polymer attached to the neural cell adhesion molecule (NCAM), in embryonic stem (ES) cell‐derived glial precursors (ESGPs) strikingly modifies their migration behavior in response to guidance cues. ESGPs transduced with a retrovirus encoding the polysialyltransferase STX exhibit enhanced migration in monolayer cultures and an increased penetration of organotypic slice cultures. Chemotaxis assays show that overexpression of PSA results in an enhanced chemotactic migration toward gradients of a variety of chemoattractants, including fibroblast growth factor 2 (FGF2), platelet‐derived growth factor, and brain‐derived neurotrophic factor (BDNF), and that this effect is mediated via the phosphatidylinositol 3′‐kinase (PI3K) pathway. Moreover, PSA‐overexpressing ESGPs also exhibit an enhanced chemotactic response to tissue explants derived from different brain regions. The effect of polysialylation on directional migration is preserved in vivo. Upon transplantation into the adult striatum, PSA‐overexpressing but not control cells display a targeted migration toward the subventricular zone. On the basis of these data, we propose that PSA plays a crucial role in modulating the ability of migrating precursor cells to respond to regional guidance cues within the brain tissue.


European Journal of Neuroscience | 2007

CNS-irrelevant T-cells enter the brain, cause blood–brain barrier disruption but no glial pathology

Alina Smorodchenko; Jens Wuerfel; Elena E. Pohl; Johannes Vogt; Eva Tysiak; Robert Glumm; Sven Hendrix; Robert Nitsch; Frauke Zipp; Carmen Infante-Duarte

Invasion of autoreactive T‐cells and alterations of the blood–brain barrier (BBB) represent early pathological manifestations of multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). Non‐CNS‐specific T‐cells are also capable of entering the CNS. However, studies investigating the spatial pattern of BBB alterations as well as the exact localization and neuropathological consequences of transferred non‐CNS‐specific cells have been thus far lacking. Here, we used magnetic resonance imaging and multiphoton microscopy, as well as histochemical and high‐precision unbiased stereological analyses to compare T‐cell transmigration, localization, persistence, relation to BBB disruption and subsequent effects on CNS tissue in a model of T‐cell transfer of ovalbumin (OVA)‐ and proteolipid protein (PLP)‐specific T‐cells. BBB alterations were present in both EAE‐mice and mice transferred with OVA‐specific T‐cells. In the latter case, BBB alterations were less pronounced, but the pattern of initial cell migration into the CNS was similar for both PLP‐ and OVA‐specific cells [mean (SEM), 95 × 103 (7.6 × 103) and 88 × 103 (18 × 103), respectively]. Increased microglial cell density, astrogliosis and demyelination were, however, observed exclusively in the brain of EAE‐mice. While mice transferred with non‐neural‐specific cells showed similar levels of rhodamine‐dextran extravasation in susceptible brain regions, EAE‐mice presented huge BBB disruption in brainstem and moderate leakage in cerebellum. This suggests that antigen specificity and not the absolute number of infiltrating cells determine the magnitude of BBB disruption and glial pathology.


Biochimica et Biophysica Acta | 2009

Comparative analysis of uncoupling protein 4 distribution in various tissues under physiological conditions and during development.

Alina Smorodchenko; Anne Rupprecht; Irina Sarilova; Olaf Ninnemann; Anja U. Bräuer; Kristin Franke; Stefan Schumacher; Sandra Techritz; Robert Nitsch; Markus Schuelke; Elena E. Pohl

UCP4 is a member of the mitochondrial uncoupling protein subfamily and one of the three UCPs (UCP2, UCP4, UCP5), associated with the nervous system. Its putative functions include thermogenesis, attenuation of reactive oxidative species (ROS), regulation of mitochondrial calcium concentration and involvement in cell differentiation and apoptosis. Here we investigate UCP4s subcellular, cellular and tissue distribution, using an antibody designed specially for this study, and discuss the findings in terms of the proteins possible functions. Western blot and immunohistochemistry data confirmed that UCP4 is expressed predominantly in the central nervous system (CNS), as previously shown at mRNA level. No protein was found in heart, spleen, stomach, intestine, lung, thymus, muscles, adrenal gland, testis and liver. The reports revealing UCP4 mRNA in kidney and white adipose tissue were not confirmed at protein level. The amount of UCP4 varies in the mitochondria of different brain regions, with the highest protein content found in cortex. We show that UCP4 is present in fetal murine brain tissue as early as embryonic days 12-14 (E12-E14), which coincides with the beginning of neuronal differentiation. The UCP4 content in mitochondria decreases as the age of mice increases. UCP4 preferential expression in neurons and its developmental expression pattern under physiological conditions may indicate a specific protein function, e.g. in neuronal cell differentiation.


Cytometry Part A | 2004

LAMP-1 and LAMP-2, but not LAMP-3, are reliable markers for activation-induced secretion of human mast cells

Andreas Grützkau; Alina Smorodchenko; Undine Lippert; Loreen Kirchhof; Metin Artuc; Beate M. Henz

Mast cells are resident tissue cells that induce anaphylactic reactions by rapidly releasing mediators after antigen‐mediated cross‐linking of immunoglobulin E receptors. In the similarly active peripheral blood basophilic leukocyte, lysosome‐associated membrane protein 3 (LAMP‐3; CD63) has been described as an activation marker, but LAMPs have not been investigated in normal tissue mast cells.

Collaboration


Dive into the Alina Smorodchenko's collaboration.

Top Co-Authors

Avatar

Anne Rupprecht

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karolina E. Hilse

University of Veterinary Medicine Vienna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Reinhold G. Erben

University of Veterinary Medicine Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge