Network


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

Hotspot


Dive into the research topics where Camilla Gustafsen is active.

Publication


Featured researches published by Camilla Gustafsen.


Molecular and Cellular Biology | 2007

Sorting by the Cytoplasmic Domain of the Amyloid Precursor Protein Binding Receptor SorLA

Morten Nielsen; Camilla Gustafsen; Peder Madsen; Jens R. Nyengaard; Guido Hermey; Oddmund Bakke; Muriel Mari; Peter Schu; Regina Pohlmann; André Dennes; Claus Munck Petersen

ABSTRACT SorLA/LR11 (250 kDa) is the largest and most composite member of the Vps10p-domain receptors, a family of type 1 proteins preferentially expressed in neuronal tissue. SorLA binds several ligands, including neurotensin, platelet-derived growth factor-bb, and lipoprotein lipase, and via complex-formation with the amyloid precursor protein it downregulates generation of Alzheimers disease-associated Aβ-peptide. The receptor is mainly located in vesicles, suggesting a function in protein sorting and transport. Here we examined SorLAs trafficking using full-length and chimeric receptors and find that its cytoplasmic tail mediates efficient Golgi body-endosome transport, as well as AP-2 complex-dependent endocytosis. Functional sorting sites were mapped to an acidic cluster-dileucine-like motif and to a GGA binding site in the C terminus. Experiments in permanently or transiently AP-1 μ1-chain-deficient cells established that the AP-1 adaptor complex is essential to SorLAs transport between Golgi membranes and endosomes. Our results further implicate the GGA proteins in SorLA trafficking and provide evidence that SNX1 and Vps35, as parts of the retromer complex or possibly in a separate context, are engaged in retraction of the receptor from endosomes.


Cell Metabolism | 2014

The Hypercholesterolemia-Risk Gene SORT1 Facilitates PCSK9 Secretion

Camilla Gustafsen; Mads Kjolby; Mette Nyegaard; Manuel Mattheisen; Henriette N. Buttenschøn; Ole Mors; Jacob F. Bentzon; Peder Madsen; Anders Nykjaer; Simon Glerup

Circulating PCSK9 destines low-density lipoprotein receptor for degradation in lysosomes, resulting in increased LDL cholesterol. Accordingly, it is an attractive drug target for hypercholesterolemia, and results from clinical trials are promising. While the physiological role of PCSK9 in cholesterol metabolism is well described, its complex mechanism of action remains poorly understood, although it is known to depend on intracellular trafficking. We here identify sortilin, encoded by the hypercholesterolemia-risk gene SORT1, as a high-affinity sorting receptor for PCSK9. Sortilin colocalizes with PCSK9 in the trans-Golgi network and facilitates its secretion from primary hepatocytes. Accordingly, sortilin-deficient mice display decreased levels of circulating PCSK9, while sortilin overexpression in the liver confers increased plasma PCSK9. Furthermore, circulating PCSK9 and sortilin were positively correlated in a human cohort of healthy individuals, suggesting that sortilin is involved in PCSK9 secretion in humans. Taken together, our findings establish sortilin as a critical regulator of PCSK9 activity.


The Journal of Neuroscience | 2013

Sortilin and SorLA Display Distinct Roles in Processing and Trafficking of Amyloid Precursor Protein

Camilla Gustafsen; Simon Glerup; Lone Tjener Pallesen; Ditte Olsen; Olav M. Andersen; Anders Nykjaer; Peder Madsen; Claus Munck Petersen

The development and progression of Alzheimers disease is linked to excessive production of toxic amyloid-β peptide, initiated by β-secretase cleavage of the amyloid precursor protein (APP). In contrast, soluble APPα (sAPPα) generated by the α-secretase is known to stimulate dendritic branching and enhance synaptic function. Regulation of APP processing, and the shift from neurotrophic to neurotoxic APP metabolism remains poorly understood, but the cellular localization of APP and its interaction with various receptors is considered important. We here identify sortilin as a novel APP interaction partner. Like the related APP receptor SorLA, sortilin is highly expressed in the CNS, but whereas SorLA mainly colocalizes with APP in the soma, sortilin interacts with APP in neurites. The presence of sortilin promotes α-secretase cleavage of APP, unlike SorLA, which inhibits the generation of all soluble products. Also, sortilin and SorLA both bind and mediate internalization of sAPP but to different cellular compartments. The interaction involves the 6A domain of APP, present in both neuronal and non-neuronal APP isoforms. This is important as sAPP receptors described so far only bind the non-neuronal isoforms, leaving SorLA and sortilin as the only receptors for sAPP generated by neurons. Together, our findings establish sortilin, as a novel APP interaction partner that influences both production and cellular uptake of sAPP.


The Journal of Neuroscience | 2013

The Pro-Neurotrophin Receptor Sortilin Is a Major Neuronal Apolipoprotein E Receptor for Catabolism of Amyloid-β Peptide in the Brain

Anne Sophie Carlo; Camilla Gustafsen; Guido Mastrobuoni; Morten Nielsen; Tilman Burgert; Daniela Hartl; Michael Rohe; Anders Nykjaer; Joachim Herz; Joerg Heeren; Stefan Kempa; Claus Munck Petersen; Thomas E. Willnow

Apolipoprotein E (APOE) is the major risk factor for sporadic Alzheimers disease. Among other functions, APOE is proposed to sequester neurotoxic amyloid-β (Aβ) peptides in the brain, delivering them to cellular catabolism via neuronal APOE receptors. Still, the receptors involved in this process remain controversial. Here, we identified the pro-neurotrophin receptor sortilin as major endocytic pathway for clearance of APOE/Aβ complexes in neurons. Sortilin binds APOE with high affinity. Lack of receptor expression in mice results in accumulation of APOE and of Aβ in the brain and in aggravated plaque burden. Also, primary neurons lacking sortilin exhibit significantly impaired uptake of APOE/Aβ complexes despite proper expression of other APOE receptors. Despite higher than normal brain APOE levels, sortilin-deficient animals display anomalies in brain lipid metabolism (e.g., accumulation of sulfatides) seen in APOE-deficient mice, indicating functional deficiency in cellular APOE uptake pathways. Together, our findings identified sortilin as an essential neuronal pathway for APOE-containing lipoproteins in vivo and suggest an intriguing link between Aβ catabolism and pro-neurotrophin signaling converging on this receptor.


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.


Traffic | 2008

Different Motifs Regulate Trafficking of SorCS1 Isoforms

Morten Nielsen; Sady J. Keat; Jida W. Hamati; Peder Madsen; Jakob J. Gutzmann; Arne Engelsberg; Karen M. Pedersen; Camilla Gustafsen; Anders Nykjaer; Jørgen Gliemann; Irm Hermans-Borgmeyer; Dietmar Kuhl; Claus Munck Petersen; Guido Hermey

The type I transmembrane protein SorCS1 is a member of the Vps10p‐domain receptor family comprised of Sortilin, SorLA and SorCS1, ‐2 and ‐3. Current information indicates that Sortilin and SorLA mediate intracellular protein trafficking and sorting, but little is known about the cellular functions of the SorCS subgroup. SorCS1 binds platelet‐derived growth factor‐BB (PDGF‐BB) and is expressed in isoforms differing only in their cytoplasmic domains. Here, we identify two novel isoforms of mouse SorCS1 designated m‐SorCS1c and ‐d. In situ hybridization revealed a combinatorial expression pattern of the variants in brain and embryonic tissues. We demonstrate that among the mouse variants, only SorCS1c mediates internalization and that the highly conserved SorCS1c is internalized through a canonical tyrosine‐based motif. In contrast, human SorCS1a, whose cytoplasmic domain is completely different from mouse SorCS1a, is internalized through a DXXLL motif. We report that the human SorCS1a cytoplasmic domain interacts with the αC/σ2 subunits of the adaptor protein (AP)‐2 complex, and internalization of human SorCS1a and ‐c is mediated by AP‐2. Our results suggest that the endocytic isoforms target internalized cargo to lysosomes but are not engaged in Golgi–endosomal transport to a significant degree.


Cell Reports | 2013

SorLA Controls Neurotrophic Activity by Sorting of GDNF and Its Receptors GFRα1 and RET

Simon Glerup; Maria Lume; Ditte Olsen; Jens R. Nyengaard; Christian Bjerggaard Vaegter; Camilla Gustafsen; Erik Ilsø Christensen; Mads Kjolby; Anders Hay-Schmidt; Dirk Bender; Peder Madsen; Mart Saarma; Anders Nykjaer; Claus Munck Petersen

Glial cell-line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor that has reached clinical trials for Parkinsons disease. GDNF binds to its coreceptor GFRα1 and signals through the transmembrane receptor tyrosine kinase RET, or RET independently through NCAM or syndecan-3. Whereas the GDNF signaling cascades are well described, cellular turnover and trafficking of GDNF and its receptors remain poorly characterized. Here, we find that SorLA acts as sorting receptor for the GDNF/GFRα1 complex, directing it from the cell surface to endosomes. Through this mechanism, GDNF is targeted to lysosomes and degraded while GFRα1 recycles, creating an efficient GDNF clearance pathway. The SorLA/GFRα1 complex further targets RET for endocytosis but not for degradation, affecting GDNF-induced neurotrophic activities. SorLA-deficient mice display elevated GDNF levels, altered dopaminergic function, marked hyperactivity, and reduced anxiety, all of which are phenotypes related to abnormal GDNF activity. Taken together, these findings establish SorLA as a critical regulator of GDNF activity in the CNS.


European Journal of Neuroscience | 2011

Proneurotrophin-3 may induce Sortilin dependent death in inner ear neurons

Jacob Tauris; Camilla Gustafsen; Erik Ilsø Christensen; Pernille Jansen; Anders Nykjaer; Jens R. Nyengaard; Kenneth K. Teng; Elisabeth Schwarz; Therese Ovesen; Peder Madsen; Claus Munck Petersen

The precursor of the neurotrophin (NT) nerve growth factor (NGF) (proNGF) serves physiological functions distinct from its mature counterpart as it induces neuronal apoptosis through activation of a p75 NT receptor (p75NTR) and Sortilin death‐signalling complex. The NTs brain‐derived nerve growth factor (BDNF) and NT3 provide essential trophic support to auditory neurons. Injury to the NT‐secreting cells in the inner ear is followed by irreversible degeneration of spiral ganglion neurons with consequences such as impaired hearing or deafness. Lack of mature NTs may explain the degeneration of spiral ganglion neurons, but another mechanism is possible as unprocessed proNTs released from the injured cells may contribute to the degeneration by induction of apoptosis. Recent studies demonstrate that proBDNF, like proNGF, is a potent inducer of Sortilin:p75NTR‐mediated apoptosis. In addition, a coincident upregulation of proBDNF and p75NTR has been observed in degenerating spiral ganglion neurons, but the Sortilin expression in the inner ear is unresolved. Here we demonstrate that Sortilin and p75NTR are coexpressed in neurons of the neonatal inner ear. Furthermore, we establish that proNT3 exhibits high‐affinity binding to Sortilin and has the capacity to enhance cell surface Sortilin:p75NTR complex formation as well as to mediate apoptosis in neurons coexpressing p75NTR and Sortilin. Based on the examination of wildtype and Sortilin‐deficient mouse embryos, Sortilin does not significantly influence the developmental selection of spiral ganglion neurons. However, our results suggest that proNT3 and proBDNF may play important roles in the response to noise‐induced injuries or ototoxic damage via the Sortilin:p75NTR death‐signalling complex.


Traffic | 2010

GGA autoinhibition revisited

Jacob Flyvholm Cramer; Camilla Gustafsen; Manja A. Behrens; Cristiano L. P. Oliveira; Jan Skov Pedersen; Peder Madsen; Claus Munck Petersen; Søren Thirup

The cytosolic adaptors GGA1‐3 mediate sorting of transmembrane proteins displaying a C‐terminal acidic dileucine motif (DXXLL) in their cytosolic domain. GGA1 and GGA3 contain similar but intrinsic motifs that are believed to serve as autoinhibitory sites activated by the phosphorylation of a serine positioned three residues upstream of the DXXLL motif. In the present study, we have subjected the widely acknowledged concept of GGA1 autoinhibition to a thorough structural and functional examination. We find that (i) the intrinsic motif of GGA1 is inactive, (ii) only C‐terminal DXXLL motifs constitute active GGA binding sites, (iii) while aspartates and phosphorylated serines one or two positions upstream of the DXXLL motif increase GGA1 binding, phosphoserines further upstream have little or no influence and (iv) phosphorylation of GGA1 does not affect its conformation or binding to Sortilin and SorLA. Taken together, our findings seem to refute the functional significance of GGA autoinhibition in particular and of intrinsic GGA binding motifs in general.


Journal of Biological Chemistry | 2015

SorLA Complement-type Repeat Domains Protect the Amyloid Precursor Protein against Processing

Arnela Mehmedbasic; Sofie K. Christensen; Jonas Nilsson; Ulla Rüetschi; Camilla Gustafsen; Annemarie Svane Aavild Poulsen; Rikke W. Rasmussen; Anja Nawarecki Fjorback; Göran Larson; Olav M. Andersen

Background: SorLA binds APP and decreases the production of Aβ; however, the molecular mechanisms controlling these processes are poorly understood. Results: We identified CR(5–8) as an APP-binding site in SorLA. Conclusion: The CR-cluster is essential for the SorLA-dependent decrease in APP proteolysis. Significance: Details regarding the function of SorLA in APP metabolism might lead to an understanding of the genetic association of SorLA with Alzheimer disease. SorLA is a neuronal sorting receptor that is genetically associated with Alzheimer disease. SorLA interacts directly with the amyloid precursor protein (APP) and affects the processing of the precursor, leading to a decreased generation of the amyloid-β peptide. The SorLA complement-type repeat (CR) domains associate in vitro with APP, but the precise molecular determinants of SorLA·APP complex formation and the mechanisms responsible for the effect of binding on APP processing have not yet been elucidated. Here, we have generated protein expression constructs for SorLA devoid of the 11 CR-domains and for two SorLA mutants harboring substitutions of the fingerprint residues in the central CR-domains. We generated SH-SY5Y cell lines that stably express these SorLA variants to study the binding and processing of APP using co-immunoprecipitation and Western blotting/ELISAs, respectively. We found that the SorLA CR-cluster is essential for interaction with APP and that deletion of the CR-cluster abolishes the protection against APP processing. Mutation of identified fingerprint residues in the SorLA CR-domains leads to changes in the O-linked glycosylation of APP when expressed in SH-SY5Y cells. Our results provide novel information on the mechanisms behind the influence of SorLA activity on APP metabolism by controlling post-translational glycosylation in the Golgi, suggesting new strategies against amyloidogenesis in Alzheimer disease.

Collaboration


Dive into the Camilla Gustafsen'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

Thomas E. Willnow

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge