Network


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

Hotspot


Dive into the research topics where Sune Skeldal is active.

Publication


Featured researches published by Sune Skeldal.


Thrombosis and Haemostasis | 2004

Plasminogen activator inhibitor-1 and tumour growth, invasion, and metastasis

Michelle K.V. Durand; Julie Støve Bødker; Anni Christensen; Daniel M. Dupont; Martin Hansen; Jan K. Jensen; Signe Kjelgaard; Lisa Mathiasen; Katrine E. Pedersen; Sune Skeldal; Troels Wind; Peter A. Andreasen

In recent decades, evidence has been accumulating showing the important role of urokinase-type plasminogen activator (uPA) in growth, invasion, and metastasis of malignant tumours. The evidence comes from results with animal tumour models and from the observation that a high level of uPA in human tumours is associated with a poor patient prognosis. It therefore initially came as a surprise that a high tumour level of the uPA inhibitor plasminogen activator inhibitor-1 (PAI-1) is also associated with a poor prognosis, the PAI-1 level in fact being one of the most informative biochemical prognostic markers. We review here recent investigations into the possible tumour biological role of PAI-1, performed by animal tumour models, histological examination of human tumours, and new knowledge about the molecular interactions of PAI-1 possibly underlying its tumour biological functions. The exact tumour biological functions of PAI-1 remain uncertain but PAI-1 seems to be multifunctional as PAI-1 is expressed by multiple cell types and has multiple molecular interactions. The potential utilisation of PAI-1 as a target for anti-cancer therapy depends on further mapping of these functions.


BioEssays | 2011

Proteolytic processing of the p75 neurotrophin receptor: A prerequisite for signalling?: Neuronal life, growth and death signalling are crucially regulated by intra-membrane proteolysis and trafficking of p75(NTR)

Sune Skeldal; Dusan Matusica; Anders Nykjaer; Elizabeth J. Coulson

The common neurotrophin receptor (p75NTR) regulates various functions in the developing and adult nervous system. Cell survival, cell death, axonal and growth cone retraction, and regulation of the cell cycle can be regulated by p75NTR‐mediated signals following activation by either mature or pro‐neurotrophins and in combination with various co‐receptors, including Trk receptors and sortilin. Here, we review the known functions of p75NTR by cell type, receptor‐ligand combination, and whether regulated intra‐membrane proteolysis of p75NTR is required for signalling. We highlight that the generation of the intracellular domain fragment of p75NTR is associated with many of the receptor functions, regardless of its ligand and co‐receptor interactions.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Structural basis for endosomal trafficking of diverse transmembrane cargos by PX-FERM proteins.

Rajesh Ghai; Andrea Bugarcic; Huadong Liu; Suzanne J. Norwood; Sune Skeldal; Elizabeth J. Coulson; Shawn S.-C. Li; Rohan D. Teasdale; Brett M. Collins

Transit of proteins through the endosomal organelle following endocytosis is critical for regulating the homeostasis of cell-surface proteins and controlling signal transduction pathways. However, the mechanisms that control these membrane-transport processes are poorly understood. The Phox-homology (PX) domain-containing proteins sorting nexin (SNX) 17, SNX27, and SNX31 have emerged recently as key regulators of endosomal recycling and bind conserved Asn-Pro-Xaa-Tyr–sorting signals in transmembrane cargos via an atypical band, 4.1/ezrin/radixin/moesin (FERM) domain. Here we present the crystal structure of the SNX17 FERM domain bound to the sorting motif of the P-selectin adhesion protein, revealing both the architecture of the atypical FERM domain and the molecular basis for recognition of these essential sorting sequences. We further show that the PX-FERM proteins share a promiscuous ability to bind a wide array of putative cargo molecules, including receptor tyrosine kinases, and propose a model for their coordinated molecular interactions with membrane, cargo, and regulatory proteins.


Nanotechnology | 2009

The influence of glancing angle deposited nano-rough platinum surfaces on the adsorption of fibrinogen and the proliferation of primary human fibroblasts

Alireza Dolatshahi-Pirouz; Cristian Pablo Pennisi; Sune Skeldal; Morten Foss; Jacques Chevallier; Vladimir Zachar; Peter A. Andreasen; Ken Yoshida; Flemming Besenbacher

We have used the glancing angle deposition (GLAD) method as a simple and fast method to generate nano-rough surfaces for protein adsorption experiments and cell assays. The surface roughness and the detailed geometrical surface morphology of the thin films were characterized by atomic force microscopy (AFM) and scanning electron microscopy (SEM). As the GLAD deposition angle approaches grazing incidence, sharp and whisker-like columnar protrusions are formed. Smaller and less sharp surface features appear for the thin films synthesized at higher deposition angles. By changing the GLAD deposition angle together with the total amount of mass deposited per area on the respective surfaces, the size of the surface features can be varied on the nanoscale. Using the GLAD topographies as model surfaces, we have investigated the influence of the nano-roughness on fibrinogen adsorption and on the proliferation of primary human fibroblasts. It is found that fibrinogen, an important blood protein, preferentially adheres on the whisker-like nano-rough substrates in comparison to a flat surface. Furthermore, the proliferation of the human fibroblasts is significantly reduced on the nano-rough substrates. These results demonstrate that the GLAD technique can be used to fabricate nano-rough surface morphologies that significantly influence both protein and cellular adhesion to surfaces and are therefore well suited for biological assays.


Neuron | 2014

SorCS2 Regulates Dopaminergic Wiring and Is Processed into an Apoptotic Two-Chain Receptor in Peripheral Glia

Simon Glerup; Ditte Olsen; Christian Bjerggaard Vaegter; Camilla Gustafsen; Susanne S. Sjoegaard; Guido Hermey; Mads Kjolby; Simon Molgaard; Maj Ulrichsen; Simon Boggild; Sune Skeldal; Anja Nawarecki Fjorback; Jens R. Nyengaard; Jan Jacobsen; Dirk Bender; Carsten R. Bjarkam; Esben S. Sørensen; Ernst-Martin Füchtbauer; Gregor Eichele; Peder Madsen; Thomas E. Willnow; Claus Munck Petersen; Anders Nykjaer

Balancing trophic and apoptotic cues is critical for development and regeneration of neuronal circuits. Here we identify SorCS2 as a proneurotrophin (proNT) receptor, mediating both trophic and apoptotic signals in conjunction with p75(NTR). CNS neurons, but not glia, express SorCS2 as a single-chain protein that is essential for proBDNF-induced growth cone collapse in developing dopaminergic processes. SorCS2- or p75(NTR)-deficient in mice caused reduced dopamine levels and metabolism and dopaminergic hyperinnervation of the frontal cortex. Accordingly, both knockout models displayed a paradoxical behavioral response to amphetamine reminiscent of ADHD. Contrary, in PNS glia, but not in neurons, proteolytic processing produced a two-chain SorCS2 isoform that mediated proNT-dependent Schwann cell apoptosis. Sciatic nerve injury triggered generation of two-chain SorCS2 in p75(NTR)-positive dying Schwann cells, with apoptosis being profoundly attenuated in Sorcs2(-/-) mice. In conclusion, we have demonstrated that two-chain processing of SorCS2 enables neurons and glia to respond differently to proneurotrophins.


FEBS Journal | 2006

Binding areas of urokinase-type plasminogen activator-plasminogen activator inhibitor-1 complex for endocytosis receptors of the low-density lipoprotein receptor family, determined by site-directed mutagenesis.

Sune Skeldal; Jakob Vejby Larsen; Katrine E. Pedersen; Helle H. Petersen; Rikke Egelund; Anni Christensen; Jan K. Jensen; Jørgen Gliemann; Peter A. Andreasen

Some endocytosis receptors related to the low‐density lipoprotein receptor, including low‐density lipoprotein receptor‐related protein‐1A, very‐low‐density lipoprotein receptor, and sorting protein‐related receptor, bind protease‐inhibitor complexes, including urokinase‐type plasminogen activator (uPA), plasminogen activator inhibitor‐1 (PAI‐1), and the uPA–PAI‐1 complex. The unique capacity of these receptors for high‐affinity binding of many structurally unrelated ligands renders mapping of receptor‐binding surfaces of serpin and serine protease ligands a special challenge. We have mapped the receptor‐binding area of the uPA–PAI‐1 complex by site‐directed mutagenesis. Substitution of a cluster of basic residues near the 37‐loop and 60‐loop of uPA reduced the receptor‐binding affinity of the uPA–PAI‐1 complex approximately twofold. Deletion of the N‐terminal growth factor domain of uPA reduced the affinity 2–4‐fold, depending on the receptor, and deletion of both the growth factor domain and the kringle reduced the affinity sevenfold. The binding affinity of the uPA–PAI‐1 complex to the receptors was greatly reduced by substitution of basic and hydrophobic residues in α‐helix D and α‐helix E of PAI‐1. The localization of the implicated residues in the 3D structures of uPA and PAI‐1 shows that they form a continuous receptor‐binding area spanning the serpin as well as the A‐chain and the serine protease domain of uPA. Our results suggest that the 10–100‐fold higher affinity of the uPA–PAI‐1 complex compared with the free components depends on the bonus effect of bringing the binding areas on uPA and PAI‐1 together on the same binding entity.


Journal of Biological Chemistry | 2012

Mapping of the interaction site between sortilin and the p75 neurotrophin receptor reveals a regulatory role for the sortilin intracellular domain in p75 neurotrophin receptor shedding and apoptosis.

Sune Skeldal; Alex M. Sykes; Simon Glerup; Dusan Matusica; Nickless Palstra; Henri Autio; Zoran Boskovic; Peder Madsen; Eero Castrén; Anders Nykjaer; Elizabeth J. Coulson

Background: Sortilin and p75NTR induce neuronal apoptosis by binding pro-neurotrophins during development and following neuronal injury. Results: Sortilin interacts with an extracellular juxtamembrane 23-amino acid sequence of p75NTR. Conclusion: Despite binding being mediated through extracellular interactions, the intracellular domain of sortilin regulates p75NTR shedding and apoptosis. Significance: Mapping may allow design of compounds inhibiting neuronal cell death by blocking the interaction between sortilin and p75NTR. Neurotrophins comprise a group of neuronal growth factors that are essential for the development and maintenance of the nervous system. However, the immature pro-neurotrophins promote apoptosis by engaging in a complex with sortilin and the p75 neurotrophin receptor (p75NTR). To identify the interaction site between sortilin and p75NTR, we analyzed binding between chimeric receptor constructs and truncated p75NTR variants by co-immunoprecipitation experiments, surface plasmon resonance analysis, and FRET. We found that complex formation between sortilin and p75NTR relies on contact points in the extracellular domains of the receptors. We also determined that the interaction critically depends on an extracellular juxtamembrane 23-amino acid sequence of p75NTR. Functional studies further revealed an important regulatory function of the sortilin intracellular domain in p75NTR-regulated intramembrane proteolysis and apoptosis. Thus, although the intracellular domain of sortilin does not contribute to p75NTR binding, it does regulate the rates of p75NTR cleavage, which is required to mediate pro-neurotrophin-stimulated cell death.


Journal of Biological Chemistry | 2013

An intracellular domain fragment of the p75 neurotrophin receptor (p75(NTR)) enhances tropomyosin receptor kinase A (TrkA) receptor function.

Dusan Matusica; Sune Skeldal; Alex M. Sykes; Nickless Palstra; Aanchal Sharma; Elizabeth J. Coulson

Background: The mechanism by which the p75 neurotrophin receptor (p75NTR) and TrkA interact to enhance neurotrophin signaling is unknown. Results: The p75NTR intracellular domain fragment, p75ICD, but not full-length p75NTR enhanced NGF binding to TrkA and neurite outgrowth. Conclusion: The results suggest that p75ICD causes a conformational change within the extracellular domain of TrkA. Significance: The findings challenge our current understanding of how p75NTR enhances neurotrophic activity. Facilitation of nerve growth factor (NGF) signaling by the p75 neurotrophin receptor (p75NTR) is critical for neuronal survival and differentiation. However, the interaction between p75NTR and TrkA receptors required for this activity is not understood. Here, we report that a specific 29-amino acid peptide derived from the intracellular domain fragment of p75NTR interacts with and potentiates binding of NGF to TrkA-expressing cells, leading to increased neurite outgrowth in sympathetic neurons as a result of enhanced Erk1/2 and Akt signaling. An endogenous intracellular domain fragment of p75NTR (p75ICD) containing these 29 amino acids is produced by regulated proteolysis of the full-length receptor. We demonstrate that generation of this fragment is a requirement for p75NTR to facilitate TrkA signaling in neurons and propose that the juxtamembrane region of p75ICD acts to cause a conformational change within the extracellular domain of TrkA. This finding provides new insight into the mechanism by which p75NTR and TrkA interact to enhance neurotrophic signaling.


Journal of Biological Chemistry | 2012

The effects of transmembrane sequence and dimerization on cleavage of the p75 neurotrophin receptor by γ-secretase.

Alex M. Sykes; Nickless Palstra; Daniel Abankwa; Justine M. Hill; Sune Skeldal; Dusan Matusica; Prahatha Venkatraman; John F. Hancock; Elizabeth J. Coulson

Background: p75 neurotrophin receptor (p75NTR) signaling is modulated by dimerization and regulated intramembrane proteolysis (RIP). Results: Transmembrane sequence and TrkA but not ligands regulate p75NTR homodimerization and γ-secretase cleavage. Conclusion: Although γ-secretase does not require a dimeric substrate, p75NTR dimerization facilitates RIP. Significance: Structural change mediated by homo- and heterodimerization is more important than ligand for inducing RIP of p75NTR. Cleavage of transmembrane receptors by γ-secretase is the final step in the process of regulated intramembrane proteolysis (RIP) and has a significant impact on receptor function. Although relatively little is known about the molecular mechanism of γ-secretase enzymatic activity, it is becoming clear that substrate dimerization and/or the α-helical structure of the substrate can regulate the site and rate of γ-secretase activity. Here we show that the transmembrane domain of the pan-neurotrophin receptor p75NTR, best known for regulating neuronal death, is sufficient for its homodimerization. Although the p75NTR ligands NGF and pro-NGF do not induce homerdimerization or RIP, homodimers of p75NTR are γ-secretase substrates. However, dimerization is not a requirement for p75NTR cleavage, suggesting that γ-secretase has the ability to recognize and cleave each receptor molecule independently. The transmembrane cysteine 257, which mediates covalent p75NTR interactions, is not crucial for homodimerization, but this residue is required for normal rates of γ-secretase cleavage. Similarly, mutation of the residues alanine 262 and glycine 266 of an AXXXG dimerization motif flanking the γ-secretase cleavage site within the p75NTR transmembrane domain alters the orientation of the domain and inhibits γ-secretase cleavage of p75NTR. Nonetheless, heteromer interactions of p75NTR with TrkA increase full-length p75NTR homodimerization, which in turn potentiates the rate of γ-cleavage following TrkA activation independently of rates of α-cleavage. These results provide support for the idea that the helical structure of the p75NTR transmembrane domain, which may be affected by co-receptor interactions, is a key element in γ-secretase-catalyzed cleavage.


FEBS Letters | 2004

Construction of a plasminogen activator inhibitor-1 variant without measurable affinity to vitronectin but otherwise normal.

Jan K. Jensen; Michelle K.V. Durand; Sune Skeldal; Daniel M. Dupont; Julie Støve Bødker; Troels Wind; Peter A. Andreasen

Vitronectin (VN) and plasminogen activator inhibitor‐1 (PAI‐1) have important functional interactions: VN stabilises the protease inhibitory activity of PAI‐1 and PAI‐1 inhibits binding of adhesion receptors to VN. Having previously mapped the PAI‐1 binding area for VN, we have now constructed a PAI‐1 variant, R103A‐M112A‐Q125A, without measurable affinity to VN, but with full protease inhibitory activity and endocytosis receptor binding. As a tool for evaluating the physiological and pathophysiological functions of the PAI‐1–VN interaction, our new variant is far superior to the previously widely used PAI‐1 variant Q125K, which we have found possesses an only about 10‐fold reduced affinity to VN.

Collaboration


Dive into the Sune Skeldal's collaboration.

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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge