Network


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

Hotspot


Dive into the research topics where Boris Klementiev is active.

Publication


Featured researches published by Boris Klementiev.


Neuroscience | 2007

A neural cell adhesion molecule–derived peptide reduces neuropathological signs and cognitive impairment induced by Aβ25-35

Boris Klementiev; Tatiana Novikova; V. Novitskaya; Peter S. Walmod; Oksana Dmytriyeva; B. Pakkenberg; Vladimir Berezin; Elisabeth Bock

Abstract By means of i.c.v. administration of preaggregated oligomeric β-amyloid (Aβ) 25-35 peptide it was possible in rats to generate neuropathological signs related to those of early stages of Alzheimer’s disease (AD). Aβ 25-35 -administration induced the deposition of endogenously produced amyloid protein. Furthermore, quantitative immunohistochemistry demonstrated time-related statistically significant increases in amyloid immunoreactivity, tau phosphorylation, microglial activation, and astrocytosis, and stereological investigations demonstrated statistically significant increased neuronal cell death and brain atrophy in response to Aβ 25-35 . Finally, the Aβ 25-35 -administration led to a reduced short-term memory as determined by the social recognition test. A synthetic peptide termed FGL derived from the neural cell adhesion molecule (NCAM) was able to prevent or, if already manifest, strongly reduce all investigated signs of Aβ 25-35 -induced neuropathology and cognitive impairment. The FGL peptide was recently demonstrated to be able to cross the blood–brain-barrier. Accordingly, we found that the beneficial effects of FGL were achieved not only by intracisternal, but also by intranasal and s.c. administration of the peptide. Furthermore, FGL-treatment was shown to inhibit the activity of GSK3β, a kinase implicated in signaling regulating cell survival, tau phosphorylation and the processing of the amyloid precursor protein (APP). Thus, the peptide induced a statistically significant increase in the fraction of GSK3β phosphorylated on the Ser9-position, a posttranslational modification known to inhibit the activity of the kinase. Hence, the mode of action of FGL with respect to the preventive and curative effects on Aβ 25-35 -induced neuropathological manifestations and cognitive impairment involves the modulation of intracellular signal-transduction mediated through GSK3β.


Neuroscience | 2006

A neural cell adhesion molecule–derived fibroblast growth factor receptor agonist, the FGL-peptide, promotes early postnatal sensorimotor development and enhances social memory retention

Thomas Secher; V. Novitskaia; Vladimir Berezin; Elisabeth Bock; Birte Glenthøj; Boris Klementiev

The neural cell adhesion molecule (NCAM) belongs to the immunoglobulin (Ig) superfamily and is composed extracellularly of five Ig-like and two fibronectin type III (F3) modules. It plays a pivotal role in neuronal development and synaptic plasticity. NCAM signals via a direct interaction with the fibroblast growth factor receptor (FGFR). A 15-amino-acid long peptide, the FG loop (FGL) peptide, that is derived from the second F3 module of NCAM has been found to activate FGFR1. We here report that the FGL peptide, when administered intranasally to newborn rats, accelerated early postnatal development of coordination skills. In adult animals s.c. administration of FGL resulted in a prolonged retention of social memory. We found that FGL rapidly penetrated into the blood and cerebrospinal fluid after both intranasal and s.c. administration and remained detectable in the fluids for up to 5 hours.


European Journal of Neuroscience | 2005

A synthetic NCAM-derived peptide, FGL, protects hippocampal neurons from ischemic insult both in vitro and in vivo.

G. G. Skibo; Iryna V. Lushnikova; Kyrill Y. Voronin; Oksana Dmitrieva; Tatiana Novikova; Boris Klementiev; Elisabetta Vaudano; Vladimir Berezin; Elisabeth Bock

There is a major unmet need for development of innovative strategies for neuroprotection against ischemic brain injury. Here we show that FGL, a neural cell adhesion molecule (NCAM)‐derived peptide binding to and inducing phosphorylation of the fibroblast growth factor receptor (FGFR), acts neuroprotectively after an ischemic insult both in vitro and in vivo. The neuroprotective activity of FGL was tested in vitro on dissociated rat hippocampal neurons and hippocampal slice cultures, using a protocol of oxygen–glucose deprivation (OGD). FGL protected hippocampal neurons from damage and maintained or restored their metabolic and presynaptic activity, both if employed as a pretreatment alone to OGD, and if only applied after the insult. In vivo 24 h pretreatment with a single suboccipital injection of FGL significantly protected hippocampal CA1 neurons from death in a transient global ischemia model in the gerbil. We conclude that FGL promotes neuronal survival after ischemic brain injury.


European Journal of Neuroscience | 2008

The NCAM-derived P2 peptide facilitates recovery of cognitive and motor function and ameliorates neuropathology following traumatic brain injury.

Boris Klementiev; Tatiana Novikova; Irina Korshunova; Vladimir Berezin; Elisabeth Bock

The neural cell adhesion molecule (NCAM) plays a crucial role during development and regeneration of the nervous system, mediating neuronal differentiation, survival and plasticity. Moreover, NCAM regulates learning and memory. A peptide termed P2, corresponding to a 12‐amino‐acid sequence in the second immunoglobulin (Ig)‐like module of NCAM, represents the natural cis‐binding site for the first NCAM Ig module. The P2 peptide targets NCAM, thereby inducing a number of intracellular signaling events leading to the stimulation of neurite outgrowth and promotion of neuronal survival in vitro. The present study evaluated the effect of the P2 peptide on functional and histological outcomes following traumatic brain injury inflicted by a cortical cryogenic lesion. Lesioned rats were injected subcutaneously with P2 peptide, 5 mg/kg daily for 15 days beginning 2 h after injury. This treatment significantly improved postlesion recovery of motor and cognitive function, reduced neuronal degeneration, protected cells against oxidative stress, and increased reactive astrogliosis and neuronal plasticity in the sublesional area. P2 appeared rapidly in blood and cerebrospinal fluid after subcutaneous administration and remained detectable in blood for up to 5 h. The results suggest that P2 has therapeutic potential for the treatment of traumatic brain injury.


International Journal of Developmental Neuroscience | 2002

A peptide agonist of the neural cell adhesion molecule (NCAM), C3, protects against developmental defects induced by a teratogen pyrimethamine.

Boris Klementiev; Natalia Bichevaja; Tatiana Novikova; Nikolaj Chebotar; Elisabeth Bock; Vladimir Berezin

The neural cell adhesion molecule, NCAM, not only plays an important role in neuronal migration, differentiation and formation of connections in the developing nervous system, but also in the condensation of the mesodermal mesenchyme of the limb bud. Therefore, NCAM may be regarded as a target molecule for preventive strategies aimed at minimizing the effects of teratogens affecting the prenatal development of the nervous system and the skeleton. Treatment of fetuses with the teratogen pyrimethamine results in a reduced body weight, microcephaly and malformations of the hind limbs and forelimbs, e.g. micromelia, brachydactyly and adactyly. We here show that a peptide agonist of NCAM, C3, partly prevented the defects induced by this treatment. Although intra‐amniotic administration of C3 at gestational day 14 had no effect on the pyrimethamine‐induced reduction in body weight, it rescued the deficit in brain weight (microcephaly), partly reversed a decrease in thickness of the cortical plate, and significantly reduced the number of malformed fetuses. In vitro, C3 promoted survival of PC12‐E2 cells treated with pyrimethamine. Since C3 is a peptide mimetic of NCAM, our data strongly suggest that stimulating of NCAM results in neuroprotection in vivo and in vitro.


Neuroscience | 2007

Effects of P2, a peptide derived from a homophilic binding site in the neural cell adhesion molecule on learning and memory in rats.

Larissa Rizhova; Boris Klementiev; Karine Cambon; César Venero; Carmen Sandi; Elena Vershinina; Elisabetta Vaudano; Vladimir Berezin; Elisabeth Bock

The neural cell adhesion molecule (NCAM) plays a pivotal role in neural development, regeneration, synaptic plasticity, and memory processes. P2 is a 12-amino-acid peptide derived from the second immunoglobulin-like (Ig) module of NCAM mediating cis-homophilic interactions between NCAM molecules present on the same cell. P2 is a potent NCAM agonist, capable of promoting neuronal differentiation and survival in vitro. The aim of this study was to assess the effect of P2 on learning and memory. Rats treated with P2 intracerebroventricularly (1 h prior to test) performed significantly better than controls in the reinforced T-maze, a test of spatial working memory. Further, rats treated with P2 exhibited decreased anxiety-like behavior while learning the T-maze task. In the social recognition test, both intracerebroventricular (1 h prior to test) and systemic (1 and 24 h prior to test) P2 treatment enhanced short-term social memory and counteracted (administration 24 h prior test) scopolamine-induced social memory impairment. In contrast, P2 (1 h prior to test) did not significantly improve long-term (24 h) retention of social memory, nor did it have any significant effects on long-term memory evaluated by the Morris water maze (administration between 2 days before training and 5.5 h posttraining). In the open field test, P2 (1 h prior to test) decreased general locomotion and rearing, but did not influence any other anxiety-related behaviors, indicating only a minimal influence on baseline anxiety levels. Taken together, these data indicate that in vivo P2 enhances short-term memory and protects against the amnestic effects of scopolamine, while modulating emotional behavior in a learning or novelty-related task.


Journal of Neuroinflammation | 2014

Anti-inflammatory properties of a novel peptide interleukin 1 receptor antagonist

Boris Klementiev; Shizhong Li; Irina Korshunova; Oksana Dmytriyeva; Stanislava Pankratova; Peter S. Walmod; Laura Kofoed Kjær; Mattias S. Dahllöf; Morten Lundh; Dan Ploug Christensen; Thomas Mandrup-Poulsen; Elisabeth Bock; Vladimir Berezin

BackgroundInterleukin 1 (IL-1) is implicated in neuroinflammation, an essential component of neurodegeneration. We evaluated the potential anti-inflammatory effect of a novel peptide antagonist of IL-1 signaling, Ilantide.MethodsWe investigated the binding of Ilantide to IL-1 receptor type I (IL-1RI) using surface plasmon resonance, the inhibition of Il-1β-induced activation of nuclear factor κB (NF-κB) in HEK-Blue cells that contained an IL-1β-sensitive reporter, the secretion of TNF-α in macrophages, protection against IL-1-induced apoptosis in neonatal pancreatic islets, and the penetration of Ilantide through the blood–brain barrier using competitive enzyme-linked immunosorbent assay (ELISA). We studied the effects of the peptide on social behavior and memory in rat models of lipopolysaccharide (LPS)- and amyloid-induced neuroinflammation, respectively, and its effect in a rat model of experimental autoimmune enchephalomyelitis.ResultsIlantide bound IL-1RI, inhibited the IL-1β-induced activation of NF-κB, and inhibited the secretion of TNF-α in vitro. Ilantide protected pancreatic islets from apoptosis in vitro and reduced inflammation in an animal model of arthritis. The peptide penetrated the blood–brain barrier. It reduced the deficits in social activity and memory in LPS- and amyloid-treated animals and delayed the development of experimental autoimmune enchephalomyelitis.ConclusionsThese findings indicate that Ilantide is a novel and potent IL-1RI antagonist that is able to reduce inflammatory damage in the central nervous system and pancreatic islets.


Neuropharmacology | 2007

Multiple effects of pentyl-4-yn-VPA enantiomers: From toxicity to short-term memory enhancement

Kamil Gotfryd; Sylwia Owczarek; Katrin Hoffmann; Boris Klementiev; Heinz Nau; Vladimir Berezin; Elisabeth Bock; Peter S. Walmod

2-n-Pentyl-4-pentynoic acid (PE-4-yn-VPA) is a derivative of the antiepileptic and mood-stabilizing drug valproic acid (VPA). PE-4-yn-VPA exists as R- and S-enantiomers, the latter being more teratogenic. PE-4-yn-VPA also possesses antiepileptic, antiproliferative, and cell-differentiating properties. Moreover, the less teratogenic enantiomer, R-PE-4-yn-VPA, was recently shown to improve learning and memory. We here present a detailed investigation of the enantioselective properties of PE-4-yn-VPA using a range of in vitro and in vivo assays including measurements of cellular growth and migration, neuronal differentiation and survival, intracellular signal transduction, synaptic plasticity and maturation, and short-term memory as determined by the social recognition test. The results show that the enantiomers of PE-4-yn-VPA largely had similar effects in vitro. However, in all in vitro experiments the more teratogenic enantiomer, S-PE-4-yn-VPA, exhibited a stronger potency than R-PE-4-yn-VPA, and only S-PE-4-yn-VPA had a detrimental effect on cell survival. Interestingly, both the R- and S-enantiomer improved learning and memory. In contrast, the beneficial effect of S-PE-4-yn-VPA on memory was lost by time, whereas the effect of R-PE-4-yn-VPA administration was longer lasting, suggesting that the beneficial effect of the S-enantiomer on memory formation may be counteracted by its detrimental effect on neuronal cell survival.


Neurobiology of Disease | 2012

Neuroprotective and memory enhancing properties of a dual agonist of the FGF receptor and NCAM.

Maj N. Enevoldsen; Artur Kochoyan; Monika Jürgenson; Külli Jaako; Oksana Dmytriyeva; Peter S. Walmod; Jesper D. Nielsen; Janne Nielsen; Shizhong Li; Irina Korshunova; Boris Klementiev; Tatiana Novikova; Alexander Zharkovsky; Vladimir Berezin; Elisabeth Bock

The fibroblast growth factor receptor (FGFR) plays a vital role in the development of the nervous system regulating a multitude of cellular processes. One of the interaction partners of the FGFR is the neural cell adhesion molecule (NCAM), which is known to play an important role in neuronal development, regeneration and synaptic plasticity. Thus, simultaneous activation of FGFR- and NCAM-mediated signaling pathways may be expected to affect processes underlying neurodegenerative diseases. We here report the identification of a peptide compound, Enreptin, capable of interacting with both FGFR and NCAM. We demonstrate that this dual specificity agonist induces phosphorylation of FGFR and differentiation and survival of primary neurons in vitro, and that these effects are inhibited by abrogation of both NCAM and FGFR signaling pathways. Furthermore, Enreptin crosses the blood-brain barrier after subcutaneous administration, enhances long-term memory in normal mice and ameliorates memory deficit in mice with induced brain inflammation. Moreover, Enreptin reduces cognitive impairment and neuronal death induced by Aβ25-35 in a rat model of Alzheimers disease, and reduces the mortality rate and clinical signs of experimental autoimmune encephalomyelitis in rats. Thus, Enreptin is an attractive candidate for the treatment of neurological diseases.


Neuroscience Letters | 2001

Not growth associated protein GAP-43 (B-50), but its fragment GAP-43-3 (B-60) predominates in rat brain during development.

Mark I Mosevitsky; Ekaterina S Konovalova; Natalja K Bitchevaya; Boris Klementiev

The participation of the nerve termini growth associated protein GAP-43 in neurite outgrowth and targeting is well documented. Commonly, besides GAP-43 itself, two big fragments devoid of four (GAP-43-2, IB-50) and of about 40 (GAP-43-3, B-60) N-terminal residues were co-isolated from brain. In adult brain, GAP-43 significantly prevails over the fragments. To find their relative amounts during development, rat brain proteins were isolated on different stages of embryonal and post-natal development and subjected to gel electrophoresis in 0.9 M acetic acid-2.5 M urea system. The bands of GAP-43 protein family were detected on Western blots. We show that in developing brain (until 5th post-natal day), a proteolysis of GAP-43 near Ser(41) that results in GAP-43-3 accumulation is activated. We hypothesize that just the functions that can be performed by the GAP-43 fragments are of importance for developing brain.

Collaboration


Dive into the Boris Klementiev's collaboration.

Top Co-Authors

Avatar

Elisabeth Bock

University of Copenhagen

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicola J. Corbett

Rush University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge