Network


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

Hotspot


Dive into the research topics where Maxim M. Bespalov is active.

Publication


Featured researches published by Maxim M. Bespalov.


Journal of Cell Biology | 2011

Heparan sulfate proteoglycan syndecan-3 is a novel receptor for GDNF, neurturin, and artemin.

Maxim M. Bespalov; Yulia Sidorova; Sarka Tumova; Anni Ahonen-Bishopp; Ana Cathia Magalhães; Evgeny Kulesskiy; Mikhail Paveliev; Claudio Rivera; Heikki Rauvala; Mart Saarma

Syndecan-3 may act alone or as a coreceptor with RET to promote cell spreading, neurite outgrowth, and migration of cortical neurons by GNDF, NRTN, and ARTN.


Journal of Biological Chemistry | 2008

The Structure of the Glial Cell Line-derived Neurotrophic Factor-Coreceptor Complex INSIGHTS INTO RET SIGNALING AND HEPARIN BINDING

Vimal Parkash; Veli-Matti Leppänen; Heidi Virtanen; Jaana M. Jurvansuu; Maxim M. Bespalov; Yulia Sidorova; Pia Runeberg-Roos; Mart Saarma; Adrian Goldman

Glial cell line-derived neurotrophic factor (GDNF), a neuronal survival factor, binds its co-receptor GDNF family receptor α1 (GFRα1) in a 2:2 ratio and signals through the receptor tyrosine kinase RET. We have solved the GDNF2·GFRα12 complex structure at 2.35 Å resolution in the presence of a heparin mimic, sucrose octasulfate. The structure of our GDNF2·GFRα12 complex and the previously published artemin2·GFRα32 complex are unlike in three ways. First, we have experimentally identified residues that differ in the ligand-GFRα interface between the two structures, in particular ones that buttress the key conserved ArgGFRα-Gluligand-ArgGFRα interaction. Second, the flexible GDNF ligand “finger” loops fit differently into the GFRαs, which are rigid. Third, and we believe most importantly, the quaternary structure of the two tetramers is dissimilar, because the angle between the two GDNF monomers is different. This suggests that the RET-RET interaction differs in different ligand2-co-receptor2-RET2 heterohexamer complexes. Consistent with this, we showed that GDNF2·GFRα12 and artemin2·GFRα32 signal differently in a mitogen-activated protein kinase assay. Furthermore, we have shown by mutagenesis and enzyme-linked immunosorbent assays of RET phosphorylation that RET probably interacts with GFRα1 residues Arg-190, Lys-194, Arg-197, Gln-198, Lys-202, Arg-257, Arg-259, Glu-323, and Asp-324 upon both domains 2 and 3. Interestingly, in our structure, sucrose octasulfate also binds to the Arg190-Lys202 region in GFRα1 domain 2. This may explain how GDNF·GFRα1 can mediate cell adhesion and how heparin might inhibit GDNF signaling through RET.


The EMBO Journal | 2004

The structure of GFRα1 domain 3 reveals new insights into GDNF binding and RET activation

Veli-Matti Leppänen; Maxim M. Bespalov; Pia Runeberg-Roos; Ülo Puurand; Andres Merits; Mart Saarma; Adrian Goldman

Glial cell line‐derived neurotrophic factor (GDNF) binds to the GDNF family co‐receptor α1 (GFRα1) and activates RET receptor tyrosine kinase. GFRα1 has a putative domain structure of three homologous cysteine‐rich domains, where domains 2 and 3 make up a central domain responsible for GDNF binding. We report here the 1.8 Å crystal structure of GFRα1 domain 3 showing a new protein fold. It is an all‐α five‐helix bundle with five disulfide bridges. The structure was used to model the homologous domain 2, the other half of the GDNF‐binding fragment, and to construct the first structural model of the GDNF–GFRα1 interaction. Using site‐directed mutagenesis, we identified closely spaced residues, Phe213, Arg224, Arg225 and Ile229, comprising a putative GDNF‐binding surface. Mutating each one of them had slightly different effects on GDNF binding and RET phosphorylation. In addition, the R217E mutant bound GDNF equally well in the presence and absence of RET. Arg217 may thus be involved in the allosteric properties of GFRα1 or in binding RET.


Journal of Biological Chemistry | 2009

Lack of L-iduronic acid in heparan sulfate affects interaction with growth factors and cell signaling.

Juan Jia; Marco Maccarana; Xiao Zhang; Maxim M. Bespalov; Ulf Lindahl; Jin-Ping Li

HSEPI (glucuronyl C5-epimerase) catalyzes the conversion of d-glucuronic acid to l-iduronic acid in heparan sulfate (HS) biosynthesis. Disruption of the Hsepi gene in mice yielded a lethal phenotype with selective organ defects but had remarkably little effect on other organ systems. We have approached the underlying mechanisms by examining the course and effects of FGF2 signaling in a mouse embryonic fibroblast (MEF) cell line derived from the Hsepi−/− mouse. The HS produced by these cells is devoid of l-iduronic acid residues but shows up-regulated N- and 6-O-sulfation compared with wild type (WT) MEF HS. In medium fortified with 10% fetal calf serum, the Hsepi−/− MEFs proliferated and migrated similarly to WT cells. Under starvation conditions, both cell types showed attenuated proliferation and migration that could be restored by the addition of FGF2 to WT cells, whereas Hsepi−/− cells were resistant. Moreover, ERK phosphorylation following FGF2 stimulation was delayed in Hsepi−/− compared with WT cells. Assessment of HS-growth factor interaction by nitrocellulose filter trapping revealed a strikingly aberrant binding property of FGF2 and glia-derived neurotropic factor to Hsepi−/− but not to WT HS. glia-derived neurotropic factor has a key role in kidney development, defective in Hsepi−/− mice. By contrast, Hsepi−/− and WT HS interacted similarly and in conventional mode with FGF10. These findings correlate defective function of growth factors with their mode of HS interaction and may help explain the partly modest organ phenotypes observed after genetic ablation of selected enzymes in HS biosynthesis.


Experimental Neurology | 2009

Heparin-binding determinants of GDNF reduce its tissue distribution but are beneficial for the protection of nigral dopaminergic neurons.

Marjo Piltonen; Maxim M. Bespalov; Dagmar Ervasti; Tero Matilainen; Yulia Sidorova; Heikki Rauvala; Mart Saarma; Pekka T. Männistö

Glial cell line-derived neurotrophic factor (GDNF) protects and repairs dopamine neurons. It binds to GDNF family receptor alpha1 (GFRalpha1) and activates receptor tyrosine kinase. Heparan sulphate proteoglycans (HSPGs) also participate in the signalling of GDNF, though binding to HS may hinder the diffusion of infused GDNF. We assessed the importance of heparin-binding determinants in the neuroprotective effects of GDNF in the 6-OHDA rat model of Parkinsons disease. We utilized a truncated, non-heparin-binding Delta38N-GDNF or combined wtGDNF with heparin-binding growth-associated molecule (HB-GAM, pleiotrophin). Tissue diffusion of wtGDNF+/-HB-GAM and Delta38N-GDNF was also compared. A protective effect against ipsilateral d-amphetamine-induced turning was seen with 10 microg wtGDNF, 17 microg HB-GAM+10 microg wtGDNF or 10 microg Delta38N-GDNF at 8 weeks post lesion. This effect was most pronounced with wtGDNF alone. HB-GAM (17 or 50 microg) also reduced rotational behaviour, but did not protect dopaminergic cells. Otherwise, the survival of TH-positive cells in the substantia nigra correlated with the behavioural data. Although Delta38N-GDNF was more widely distributed than wtGDNF (irrespective of its origin), stable in a brain extract, and potent in mitogen-activated kinase assay, it was inferior in vivo. The results imply that GDNF binding to HSs is needed for the optimum neuroprotective effect.


Antiviral Research | 2016

Discovery of berberine, abamectin and ivermectin as antivirals against chikungunya and other alphaviruses.

Finny S. Varghese; Pasi Kaukinen; Sabine Gläsker; Maxim M. Bespalov; Leena Hanski; Krister Wennerberg; Beate M. Kümmerer; Tero Ahola

Chikungunya virus (CHIKV) is an arthritogenic arbovirus of the Alphavirus genus, which has infected millions of people after its re-emergence in the last decade. In this study, a BHK cell line containing a stable CHIKV replicon with a luciferase reporter was used in a high-throughput platform to screen approximately 3000 compounds. Following initial validation, 25 compounds were chosen as primary hits for secondary validation with wild type and reporter CHIKV infection, which identified three promising compounds. Abamectin (EC50 = 1.5 μM) and ivermectin (EC50 = 0.6 μM) are fermentation products generated by a soil dwelling actinomycete, Streptomyces avermitilis, whereas berberine (EC50 = 1.8 μM) is a plant-derived isoquinoline alkaloid. They inhibited CHIKV replication in a dose-dependent manner and had broad antiviral activity against other alphaviruses--Semliki Forest virus and Sindbis virus. Abamectin and ivermectin were also active against yellow fever virus, a flavivirus. These compounds caused reduced synthesis of CHIKV genomic and antigenomic viral RNA as well as downregulation of viral protein expression. Time of addition experiments also suggested that they act on the replication phase of the viral infectious cycle.


Biochemical Journal | 2005

The first cysteine-rich domain of the receptor GFRα1 stabilizes the binding of GDNF

Heidi Virtanen; Jianmin Yang; Maxim M. Bespalov; Jukka Hiltunen; Veli-Matti Leppänen; Nisse Kalkkinen; Adrian Goldman; Mart Saarma; Pia Runeberg-Roos

The GDNF (glial cell line-derived neurotrophic factor)-binding receptor GFRalpha1 (GDNF family receptor alpha1) is attached to the membrane by a GPI (glycosylphosphatidylinositol) anchor and consists of three cysteine-rich domains. The region corresponding to the second and third domains has been shown previously to participate in ligand binding, and to interact with the transmembrane tyrosine kinase receptor RET. No function has so far been found for the N-terminal, first domain (D1). Here we show that the GPI-anchored full-length receptor binds 125I-GDNF two times more tightly than does a GPI-anchored truncated receptor lacking D1. Scintillation proximity assays with purified receptor proteins also show that the GDNF-binding capacity of the soluble full-length GFRalpha1 is two times higher than the GDNF-binding capacity of the soluble D1-truncated GFRalpha1. As RET stabilizes the binding of GDNF equally well to the full-length and truncated receptors, D1 seems not to be involved in the interaction between GFRalpha1 and RET. Moreover, soluble full-length GFRalpha1 mediates GDNF-promoted neurite outgrowth in PC6-3 cells more efficiently than the soluble truncated GFRalpha1 protein. At low concentrations, the soluble fulllength receptor mediates the phosphorylation of RET more efficiently than the soluble truncated receptor. However, when the receptors are overexpressed on the cell surface as GPI-anchored proteins, or added to the growth medium at high concentrations as soluble proteins, full-length and truncated GFRalpha1 are indistinguishable in GDNF-dependent RET-phosphorylation assays. High levels of the receptors can thus mask a slightly impaired function in the phosphorylation assay. Based on assays with both GPI-anchored and soluble receptors, we therefore conclude that D1 contributes to the optimal function of GFRalpha1 by stabilizing the interaction between GFRalpha1 and GDNF.


Molecular and Cellular Neuroscience | 2010

Persephin signaling through GFRα1: The potential for the treatment of Parkinson's disease

Yulia Sidorova; Kert Mätlik; Mikhail Paveliev; Maria Lindahl; Elisa Piranen; Jeffrey Milbrandt; Urmas Arumäe; Mart Saarma; Maxim M. Bespalov

Neurotrophic factors promote survival, proliferation and differentiation of neurons inducing intracellular signaling via specific receptors. The conventional biochemical methods often fail to reveal full repertoire of neurotrophic factor-receptor interactions because of their limited sensitivity. We evaluated several approaches to study signaling of Glial cell line-Derived Neurotrophic Factor (GDNF) family ligands and found that reporter-gene systems possess exceptionally high sensitivity and a heuristic power to identify novel biologically relevant growth factor-receptor interactions. We identified persephin, a GDNF family member, as a novel ligand for GFRalpha1/RET receptor complex. We confirmed this finding by several independent methods, including neurite outgrowth assay from the explants of sympathetic ganglia expressing Gfralpha1 and Ret mRNA but not persephins conventional receptor GFRalpha4. As the activation of GFRalpha1/RET was shown to rescue dopaminergic neurons, our results suggest the potential of persephin for the treatment of Parkinsons disease.


Antiviral Research | 2017

Obatoclax, saliphenylhalamide and gemcitabine inhibit Zika virus infection in vitro and differentially affect cellular signaling, transcription and metabolism

Maxim M. Bespalov; Jatin Nandania; Aleksandr Ianevski; Vidya Velagapudi; Jef K. De Brabander; Denis E. Kainov; Olli Vapalahti

&NA; An epidemic of Zika virus (ZIKV) infection associated with congenital abnormalities such as microcephaly, is ongoing in the Americas and the Pacific. Currently there are no approved therapies to treat this emerging viral disease. Here, we tested three cell‐directed broad‐spectrum antiviral compounds against ZIKV replication using human retinal pigment epithelial (RPE) cells and a low‐passage ZIKV strain isolated from fetal brain. We found that obatoclax, SaliPhe, and gemcitabine inhibited ZIKV infections at noncytotoxic concentrations. Moreover, all three compounds prevented production of viral RNA and proteins as well as activation of cellular caspase 8, 3 and 7. However, these compounds differentially affected ZIKV‐mediated transcription, translation and posttranslational modifications of cellular factors as well as metabolic pathways indicating that these agents possess different mechanisms of action. Interestingly, combination of obatoclax and SaliPhe at nanomolar concentrations had a synergistic effect against ZIKV infection. Thus, our results provided the foundation for development of broad‐spectrum cell‐directed antivirals or their combinations for treatment of ZIKV and other emerging viral diseases. HighlightsZika virus epidemics is ongoing in the Americas and the Pacific.There are no approved therapies to treat ZIKV infection.Here we identified novel (obatoclax and SaliPhe) inhibitors of ZIKV replication.Our results broaden the spectrum of antiviral activity of these compounds.


Neurobiology of Disease | 2016

Developing therapeutically more efficient Neurturin variants for treatment of Parkinson's disease.

Pia Runeberg-Roos; Elisa Piccinini; Anna-Maija Penttinen; Kert Mätlik; Hanna Heikkinen; Satu Kuure; Maxim M. Bespalov; Johan Peränen; Enrique Garea-Rodriguez; Eberhard Fuchs; Mikko Airavaara; Nisse Kalkkinen; Richard Penn; Mart Saarma

In Parkinsons disease midbrain dopaminergic neurons degenerate and die. Oral medications and deep brain stimulation can relieve the initial symptoms, but the disease continues to progress. Growth factors that might support the survival, enhance the activity, or even regenerate degenerating dopamine neurons have been tried with mixed results in patients. As growth factors do not pass the blood-brain barrier, they have to be delivered intracranially. Therefore their efficient diffusion in brain tissue is of crucial importance. To improve the diffusion of the growth factor neurturin (NRTN), we modified its capacity to attach to heparan sulfates in the extracellular matrix. We present four new, biologically fully active variants with reduced heparin binding. Two of these variants are more stable than WT NRTN in vitro and diffuse better in rat brains. We also show that one of the NRTN variants diffuses better than its close homolog GDNF in monkey brains. The variant with the highest stability and widest diffusion regenerates dopamine fibers and improves the conditions of rats in a 6-hydroxydopamine model of Parkinsons disease more potently than GDNF, which previously showed modest efficacy in clinical trials. The new NRTN variants may help solve the major problem of inadequate distribution of NRTN in human brain tissue.

Collaboration


Dive into the Maxim M. Bespalov's collaboration.

Top Co-Authors

Avatar

Mart Saarma

University of Helsinki

View shared research outputs
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

Maija Wolf

University of Helsinki

View shared research outputs
Top Co-Authors

Avatar

Mika Kontro

University of Helsinki

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge