Network


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

Hotspot


Dive into the research topics where Lene B. Køhler is active.

Publication


Featured researches published by Lene B. Køhler.


Journal of Neurochemistry | 2004

An NCAM-derived FGF-receptor agonist, the FGL-peptide, induces neurite outgrowth and neuronal survival in primary rat neurons.

Johanne Louise Neiiendam; Lene B. Køhler; Claus Christensen; Shizhong Li; Martin V. Pedersen; Dorte Kornerup Ditlevsen; Martin Kirkegaard Kornum; Vladislav V. Kiselyov; Vladimir Berezin; Elisabeth Bock

The Neural Cell Adhesion Molecule (NCAM) plays a crucial role in development of the central nervous system regulating cell migration, differentiation and synaptogenesis. NCAM mediates cell–cell adhesion through homophilic NCAM binding, subsequently resulting in activation of the fibroblast growth factor receptor (FGFR). NCAM‐mediated adhesion leads to activation of various intracellular signal transduction pathways, including the Ras‐mitogen activated protein kinase (MAPK) and the phosphatidylinositol‐3‐kinase (PI3K)‐Akt pathways. A synthetic peptide derived from the second fibronectin type III module of NCAM, the FGL peptide, binds to and induces phosphorylation of FGFR without prior homophilic NCAM binding. We here present evidence that this peptide is able to mimic NCAM heterophilic binding to the FGFR by inducing neuronal differentiation as reflected by neurite outgrowth through a direct interaction with FGFR in primary cultures of three different neuronal cell types all expressing FGFR subtype 1: dopaminergic, hippocampal and cerebellar granule neurons. Moreover, we show that the FGL peptide promotes neuronal survival upon induction of cell death in the same three cell types. The effects of the FGL peptide are shown to depend on activation of FGFR and the MAPK and PI3K intracellular signalling pathways, all three kinases being necessary for the effects of FGL on neurite outgrowth and neuronal survival.


Journal of Neurochemistry | 2003

The role of phosphatidylinositol 3-kinase in neural cell adhesion molecule-mediated neuronal differentiation and survival.

Dorte Kornerup Ditlevsen; Lene B. Køhler; Martin V. Pedersen; Michael Risell; Kateryna Kolkova; Morten Meyer; Vladimir Berezin; Elisabeth Bock

The neural cell adhesion molecule, NCAM, is known to stimulate neurite outgrowth from primary neurones and PC12 cells presumably through signalling pathways involving the fibroblast growth factor receptor (FGFR), protein kinase A (PKA), protein kinase C (PKC), the Ras‐mitogen activated protein kinase (MAPK) pathway and an increase in intracellular Ca2+ levels. Stimulation of neurones with the synthetic NCAM‐ligand, C3, induces neurite outgrowth through signalling pathways similar to the pathways activated through physiological, homophilic NCAM‐stimulation. We present here data indicating that phosphatidylinositol 3‐kinase (PI3K) is required for NCAM‐mediated neurite outgrowth from PC12‐E2 cells and from cerebellar and dopaminergic neurones in primary culture, and that the thr/ser kinase Akt/protein kinase B (PKB) is phosphorylated downstream of PI3K after stimulation with C3. Moreover, we present data indicating a survival‐promoting effect of NCAM‐stimulation by C3 on cerebellar and dopaminergic neurones induced to undergo apoptosis. This protective effect of C3 included an inhibition of both DNA‐fragmentation and caspase‐3 activation. The survival‐promoting effect of NCAM‐stimulation was also shown to be dependent on PI3K.


Brain Research | 2003

The role of metallothionein II in neuronal differentiation and survival.

Lene B. Køhler; Vladimir Berezin; Elisabeth Bock; Milena Penkowa

Metallothionein I and II (MT-I+II) are antioxidant and tissue protective factors. We have previously shown that MT-I+II prevent oxidative stress and apoptotic cell death and are of therapeutic value in brain inflammation. However, MT-I+II are expressed in glia and it remains to be elucidated if MT-I+II can affect neurons directly. It is likely that MT isoforms could be beneficial also during neurodegenerative disorders. In this study, we have examined if MT-II affects survival and neurite extension of dopaminergic and hippocampal neurons. We show for the first time that MT-II treatment can significantly stimulate neurite extension from both dopaminergic and hippocampal neurons. Moreover, MT-II treatment significantly increases survival of dopaminergic neurons exposed to 6-hydroxydopamine (6-OHDA) and protects significantly hippocampal neurons from amyloid beta-peptide-induced neurotoxicity. Accordingly, treatment with MT-II may be of therapeutic value in neurodegenerative disorders.


Brain | 2010

Neuroprotective properties of a novel, non-haematopoietic agonist of the erythropoietin receptor

Stanislava Pankratova; Darya Kiryushko; Katrin Sonn; Vladislav Soroka; Lene B. Køhler; Mette Rathje; Bing Gu; Kamil Gotfryd; Ole Clausen; Alexander Zharkovsky; Elisabeth Bock; Vladimir Berezin

Erythropoietin, a member of the type 1 cytokine superfamily, controls proliferation and differentiation of erythroid progenitor cells through binding to and dimerization of the erythropoietin receptor. Both erythropoietin and its receptor are also expressed in the central nervous system, where they are involved in tissue protection. However, the use of erythropoietin as a neuroprotective agent may be hampered by its erythropoietic activity. Therefore, developing non-haematopoietic erythropoietin mimetics is important. Based on the crystal structure of the complex of erythropoietin and its receptor, we designed a peptide, termed Epotris, corresponding to the C α-helix region (amino-acid residues 92-111) of human erythropoietin. The peptide specifically bound to the erythropoietin receptor and promoted neurite outgrowth and survival of primary neurons with the same efficiency as erythropoietin, but with 10(3)-fold lower potency. Knockdown of the erythropoietin receptor or interference with its downstream signalling inhibited the Epotris-induced neuritogenic and pro-survival effect. Similarly to erythropoietin, Epotris penetrated the blood-brain barrier. Moreover, treatment with the peptide attenuated seizures, decreased mortality and reduced neurodegeneration in an in vivo model of kainic acid-induced neurotoxicity. In contrast to erythropoietin, Epotris did not stimulate erythropoiesis upon chronic administration. Thus, Epotris is a novel neuroprotective non-haematopoietic erythropoietin mimetic that may offer new opportunities for the treatment of neurological disorders.


Journal of Neuroscience Research | 2004

Neuritogenic and survival‐promoting effects of the P2 peptide derived from a homophilic binding site in the neural cell adhesion molecule

Martin V. Pedersen; Lene B. Køhler; Dorte Kornerup Ditlevsen; Shizong Li; Vladimir Berezin; Elisabeth Bock

The neural cell adhesion molecule (NCAM) plays a pivotal role in neural development, regeneration, and plasticity. NCAM mediates adhesion and subsequent signal transduction through NCAM–NCAM binding. Recently, a peptide ligand termed P2 corresponding to a 12‐amino‐acid sequence in the FG loop of the second Ig domain of NCAM was shown to mimic NCAM homophilic binding as reflected by induction of neurite outgrowth in hippocampal neurons. We demonstrate here that in concentrations between 0.1 and 10 μM, P2 also induced neuritogenesis in primary dopaminergic and cerebellar neurons. Furthermore, it enhanced the survival rate of cerebellar neurons although not of mesencephalic dopaminergic neurons. Moreover, our data indicate that the protective effect of P2 in cerebellar neurons was due to an inhibition of the apoptotic process, in that caspase‐3 activity and the level of DNA fragmentation were lowered by P2. Finally, treatment of neurons with P2 resulted in phosphorylation of the ser/thr kinase Akt. Thus, a small peptide mimicking homophilic NCAM interaction is capable of inducing differentiation as reflected by neurite outgrowth in several neuronal cell types and inhibiting apoptosis in cerebellar granule neurons.


Journal of Neuroscience Research | 2004

The Mts1/S100A4 protein is a neuroprotectant

Martin V. Pedersen; Lene B. Køhler; Mariam Grigorian; Vera Novitskaya; Elisabeth Bock; Eugine Lukanidin; Vladimir Berezin

Mts1 (S100A4) is a calcium‐binding protein of the EF‐hand type, belonging to the S100 family of proteins. The mts1/S100A4 gene was originally isolated from tumor cell lines, and the protein is believed to play an important role in tumor progression. More recently, oligomeric, but not dimeric, forms of Mts1 have been shown to have a neuritogenic effect when added extracellularly to hippocampal neurons. Here we show increased neurite outgrowth in two other cell types, dopaminergic and cerebellar neurons, in response to treatment with Mts1 oligomers. Moreover, we demonstrate that Mts1 acts as a neuroprotectant in primary cerebellar, dopaminergic, and hippocampal neurons induced to undergo cell death. Interestingly, the survival of the cerebellar and hippocampal neurons increased as a result of treatment with Mts1 not only in oligomeric form but also—although to a lesser extent—in dimeric form. The inhibition of death in cerebellar neurons by Mts1 was accompanied by an inhibition of DNA fragmentation, but Mts1 did not affect the activity of caspases‐3 and ‐6. In hippocampal neurons, cell death induced by the amyloid‐β peptide (Aβ25–35) was characterized by an increase in caspase‐3 and ‐6 activity, but no DNA fragmentation was observed. As in cerebellar neurons, the induced increase in caspase activity in hippocampal neurons was not affected by Mts1.


Journal of Neuroscience Research | 2007

Cyclic guanosine monophosphate signalling pathway plays a role in neural cell adhesion molecule‐mediated neurite outgrowth and survival

Dorte Kornerup Ditlevsen; Lene B. Køhler; Vladimir Berezin; Elisabeth Bock

The neural cell adhesion molecule (NCAM) plays a crucial role in neuronal development, regeneration, and synaptic plasticity associated with learning and memory consolidation. Homophilic binding of NCAM leads to neurite extension and neuroprotection in various types of primary neurons through activation of a complex network of signalling cascades, including fibroblast growth factor receptor, Src‐family kinases, the mitogen‐activated protein kinase pathway, protein kinase C, phosphatidylinositol‐3 kinase, and an increase in intracellular Ca2+. Here we present data indicating an involvement of cyclic GMP in NCAM‐mediated neurite outgrowth in both hippocampal and dopaminergic neurons and in NCAM‐mediated neuroprotection of dopaminergic neurons. In addition, evidence is presented suggesting that NCAM mediates activation of cGMP via synthesis of nitric oxide (NO) by NO synthase (NOS) and activation of soluble guanylyl cyclase by NO, leading to an increased synthesis of cGMP and activation by cGMP of protein kinase G.


Journal of Neurochemistry | 2007

Fibroblast growth factor-derived peptides : functional agonists of the fibroblast growth factor receptor

Shizhong Li; Claus Christensen; Vladislav V. Kiselyov; Lene B. Køhler; Elisabeth Bock; Vladimir Berezin

A series of peptides, termed dekafins, were derived from the β10–β11 loop regions of fibroblast growth factors (FGFs) 1, 2, 3, 5, 6, 8, 9, 10, and 17. The dekafins share a homologous amino acid sequence similar to a sequence in the first fibronectin type III module of the neural cell adhesion molecule. All dekafins were shown by surface plasmon resonance analysis to bind fibroblast growth factor receptor (FGFR)1‐IIIc‐Ig2–3 and FGFR2‐IIIb‐Ig2–3, respectively, with Kd values of approximately 10−7 to 10−8 mol/L. Binding of dekafin1 to FGFR1‐IIIc‐Ig2–3 was inhibited by a heparin analog, sucrose octasulfate, indicating that heparin sulfate moiety can modulate dekafin binding to FGFRs. Treatment of transcription and mRNA export (TREX) cells permanently expressing Strep‐tag‐labeled FGFR1‐IIIc with dekafins resulted in receptor phosphorylation. FGF1‐induced FGFR1‐IIIc phosphorylation was inhibited by dekafin1 and 10 in high concentrations, indicating that dekafins are FGFR partial agonists. The dekafins induced neuronal differentiation as reflected by neurite outgrowth from cerebellar granule neurons, an effect that was abolished by SU5402, a specific inhibitor of the FGFR tyrosine kinase, and by inositolhexaphosphate, an extracellularly acting FGFR antagonist. Some, but not all, dekafins were capable of promoting survival of cerebellar granule neurons induced to undergo apoptosis. Thus, the dekafins are functional FGFR agonists with apparent therapeutic potential.


Journal of Neurochemistry | 2008

NCAM-derived peptides function as agonists for the fibroblast growth factor receptor

Stine Maria Hansen; Lene B. Køhler; Shizhong Li; Vladislav V. Kiselyov; Claus Christensen; Sylwia Owczarek; Elisabeth Bock; Vladimir Berezin

The neural cell adhesion molecule (NCAM) directly interacts with the fibroblast growth factor receptor (FGFR). Both fibronectin type III (FN3) modules of NCAM are involved in this interaction. One of the NCAM–FGFR contact sites has been localized recently to the upper N‐terminal part of the second NCAM FN3 module encompassing the F and G β‐strands and the interconnecting loop region. Here, we investigated whether any of the six putative strand‐loop‐strand regions in the first NCAM FN3 module are involved in FGFR interactions. Peptide sequences encompassing these regions, termed encamins, were synthesized and tested for their ability to bind and activate FGFR. Encamins localized to the N‐terminal part of the first FN3 module did not interact with FGFR, whereas encamins localized to the C‐terminal part, termed EncaminA, C and E, bound to and activated FGFR. The encamins induced FGFR‐dependent neurite outgrowth, and EncaminC and E promoted neuronal survival and enhanced pre‐synaptic function. In conclusion, the interaction between NCAM and FGFR probably involves multiple contact sites at an interface formed by the two NCAM FN3 modules and FGFR, and encamins could constitute important pharmacological tools for the study of specific functional aspects of NCAM, including neuroprotection and modulation of plasticity.


PLOS ONE | 2011

A Peptide Mimetic Targeting Trans-Homophilic NCAM Binding Sites Promotes Spatial Learning and Neural Plasticity in the Hippocampus

Igor Kraev; Christian Henneberger; Clara Rossetti; Lisa Conboy; Lene B. Køhler; Martina Fantin; Alistair Jennings; César Venero; Victor I. Popov; Dmitri A. Rusakov; Michael G. Stewart; Elisabeth Bock; Vladimir Berezin; Carmen Sandi

The key roles played by the neural cell adhesion molecule (NCAM) in plasticity and cognition underscore this membrane protein as a relevant target to develop cognitive-enhancing drugs. However, NCAM is a structurally and functionally complex molecule with multiple domains engaged in a variety of actions, which raise the question as to which NCAM fragment should be targeted. Synthetic NCAM mimetic peptides that mimic NCAM sequences relevant to specific interactions allow identification of the most promising targets within NCAM. Recently, a decapeptide ligand of NCAM—plannexin, which mimics a homophilic trans-binding site in Ig2 and binds to Ig3—was developed as a tool for studying NCAMs trans-interactions. In this study, we investigated plannexins ability to affect neural plasticity and memory formation. We found that plannexin facilitates neurite outgrowth in primary hippocampal neuronal cultures and improves spatial learning in rats, both under basal conditions and under conditions involving a deficit in a key plasticity-promoting posttranslational modification of NCAM, its polysialylation. We also found that plannexin enhances excitatory synaptic transmission in hippocampal area CA1, where it also increases the number of mushroom spines and the synaptic expression of the AMPAR subunits GluA1 and GluA2. Altogether, these findings provide compelling evidence that plannexin is an important facilitator of synaptic functional, structural and molecular plasticity in the hippocampal CA1 region, highlighting the fragment in NCAMs Ig3 module where plannexin binds as a novel target for the development of cognition-enhancing drugs.

Collaboration


Dive into the Lene B. Køhler'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

Shizhong Li

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bing Gu

University of Copenhagen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge