Network


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

Hotspot


Dive into the research topics where Raphael Kopan is active.

Publication


Featured researches published by Raphael Kopan.


Cell | 2009

The Canonical Notch Signaling Pathway: Unfolding the Activation Mechanism

Raphael Kopan; Ma. Xenia G. Ilagan

Notch signaling regulates many aspects of metazoan development and tissue renewal. Accordingly, the misregulation or loss of Notch signaling underlies a wide range of human disorders, from developmental syndromes to adult-onset diseases and cancer. Notch signaling is remarkably robust in most tissues even though each Notch molecule is irreversibly activated by proteolysis and signals only once without amplification by secondary messenger cascades. In this Review, we highlight recent studies in Notch signaling that reveal new molecular details about the regulation of ligand-mediated receptor activation, receptor proteolysis, and target selection.


Nature | 1999

A presenilin-1-dependent |[gamma]|-secretase-like protease mediates release of Notch intracellular domain

Bart De Strooper; Wim Annaert; Philippe Cupers; Paul Saftig; Katleen Craessaerts; Jeff S. Mumm; Eric H. Schroeter; Vincent Schrijvers; Michael S. Wolfe; William J. Ray; Alison Goate; Raphael Kopan

Signalling through the receptor protein Notch, which is involved in crucial cell-fate decisions during development, requires ligand-induced cleavage of Notch. This cleavage occurs within the predicted transmembrane domain, releasing the Notch intracellular domain (NICD), and is reminiscent of γ-secretase-mediated cleavage of β-amyloid precursor protein (APP), a critical event in the pathogenesis of Alzheimers disease. A deficiency in presenilin-1 (PS1) inhibits processing of APP by γ-secretase in mammalian cells, and genetic interactions between Notch and PS1 homologues in Caenorhabditis elegans indicate that the presenilins may modulate the Notch signalling pathway. Here we report that, in mammalian cells, PS1 deficiency also reduces the proteolytic release of NICD from a truncated Notch construct, thus identifying the specific biochemical step of the Notch signalling pathway that is affected by PS1. Moreover, several γ-secretase inhibitors block this same step in Notch processing, indicating that related protease activities are responsible for cleavage within the predicted transmembrane domains of Notch and APP. Thus the targeting of γ-secretase for the treatment of Alzheimers disease may risk toxicity caused by reduced Notch signalling.


Nature | 1998

Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain

Eric H. Schroeter; Jeffrey A. Kisslinger; Raphael Kopan

Notch proteins are ligand-activated transmembrane receptors involved in cell-fate selection throughout development. No known enzymatic activity is contained within Notch and the molecular mechanism by which it transduces signals across the cell membrane is poorly understood. In many instances, Notch activation results in transcriptional changes in the nucleus through an association with members of the CSL family of DNA-binding proteins (where CSL stands for CBF1, Su(H), Lag-1). As Notch is located in the plasma membrane and CSL is a nuclear protein, two models have been proposed to explain how they interact (Fig. 1) . The first suggests that the two interact transiently at the membrane,. The second postulates that Notch is cleaved by a protease, enabling the cleaved fragment to enter the nucleus,. Here we show that signalling by a constitutively active membrane-bound Notch-1 protein requires the proteolytic release of the Notch intracellular domain (NICD), which interacts preferentially with CSL. Very small amounts of NICD are active, explaining why it is hard to detect in the nucleus in vivo. We also show that it is ligand binding that induces release of NICD.


Molecular Cell | 2000

A Ligand-Induced Extracellular Cleavage Regulates γ-Secretase-like Proteolytic Activation of Notch1

Jeff S. Mumm; Eric H. Schroeter; Meera T. Saxena; Adam Griesemer; Xiaolin Tian; D.J Pan; William J. Ray; Raphael Kopan

Gamma-secretase-like proteolysis at site 3 (S3), within the transmembrane domain, releases the Notch intracellular domain (NICD) and activates CSL-mediated Notch signaling. S3 processing occurs only in response to ligand binding; however, the molecular basis of this regulation is unknown. Here we demonstrate that ligand binding facilitates cleavage at a novel site (S2), within the extracellular juxtamembrane region, which serves to release ectodomain repression of NICD production. Cleavage at S2 generates a transient intermediate peptide termed NEXT (Notch extracellular truncation). NEXT accumulates when NICD production is blocked by point mutations or gamma-secretase inhibitors or by loss of presenilin 1, and inhibition of NEXT eliminates NICD production. Our data demonstrate that S2 cleavage is a ligand-regulated step in the proteolytic cascade leading to Notch activation.


Nature Reviews Molecular Cell Biology | 2004

γ-Secretase: proteasome of the membrane?

Raphael Kopan; Ma. Xenia G. Ilagan

γ-Secretase — a protease that cleaves within the membrane — was first recognized for its role in the production of amyloidogenic Aβ peptides, but was subsequently found to mediate Notch signalling by releasing the Notch-receptor intracellular domain. Many other γ-secretase substrates have recently been identified, which indicates a broader biological function for this unusual protease. Emerging evidence implies that whereas some intracellular cleavage products of γ-secretase function as signalling molecules, others might simply be intermediates that are destined for degradation.


Developmental Cell | 2010

Patterning a Complex Organ: Branching Morphogenesis and Nephron Segmentation in Kidney Development

Frank Costantini; Raphael Kopan

The two major components of the kidney, the collecting system and the nephron, have different developmental histories. The collecting system arises by the reiterated branching of a simple epithelial tube, while the nephron forms from a cloud of mesenchymal cells that coalesce into epithelial vesicles. Each develops into a morphologically complex and highly differentiated structure, and together they provide essential filtration and resorption functions. In this review, we will consider their embryological origin and the genes controlling their morphogenesis, patterning, and differentiation, with a focus on recent advances in several areas.


Nature Medicine | 2008

Notch signaling maintains bone marrow mesenchymal progenitors by suppressing osteoblast differentiation

Matthew J. Hilton; Xiaolin Tu; Ximei Wu; Shuting Bai; Haibo Zhao; Tatsuya Kobayashi; Henry M. Kronenberg; Steven L. Teitelbaum; F. Patrick Ross; Raphael Kopan; Fanxin Long

Postnatal bone marrow houses mesenchymal progenitor cells that are osteoblast precursors. These cells have established therapeutic potential, but they are difficult to maintain and expand in vitro, presumably because little is known about the mechanisms controlling their fate decisions. To investigate the potential role of Notch signaling in osteoblastogenesis, we used conditional alleles to genetically remove components of the Notch signaling system during skeletal development. We found that disruption of Notch signaling in the limb skeletogenic mesenchyme markedly increased trabecular bone mass in adolescent mice. Notably, mesenchymal progenitors were undetectable in the bone marrow of mice with high bone mass. As a result, these mice developed severe osteopenia as they aged. Moreover, Notch signaling seemed to inhibit osteoblast differentiation through Hes or Hey proteins, which diminished Runx2 transcriptional activity via physical interaction. These results support a model wherein Notch signaling in bone marrow normally acts to maintain a pool of mesenchymal progenitors by suppressing osteoblast differentiation. Thus, mesenchymal progenitors may be expanded in vitro by activating the Notch pathway, whereas bone formation in vivo may be enhanced by transiently suppressing this pathway.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Loss of leucine-rich repeat kinase 2 causes impairment of protein degradation pathways, accumulation of α-synuclein, and apoptotic cell death in aged mice

Youren Tong; Hiroo Yamaguchi; Emilie Giaime; Scott Boyle; Raphael Kopan; Raymond J. Kelleher; Jie Shen

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinsons disease. LRRK2 is a large protein containing a small GTPase domain and a kinase domain, but its physiological role is unknown. To identify the normal function of LRRK2 in vivo, we generated two independent lines of germ-line deletion mice. The dopaminergic system of LRRK2−/− mice appears normal, and numbers of dopaminergic neurons and levels of striatal dopamine are unchanged. However, LRRK2−/− kidneys, which suffer the greatest loss of LRRK compared with other organs, develop striking accumulation and aggregation of α-synuclein and ubiquitinated proteins at 20 months of age. The autophagy–lysosomal pathway is also impaired in the absence of LRRK2, as indicated by accumulation of lipofuscin granules as well as altered levels of LC3-II and p62. Furthermore, loss of LRRK2 dramatically increases apoptotic cell death, inflammatory responses, and oxidative damage. Collectively, our findings show that LRRK2 plays an essential and unexpected role in the regulation of protein homeostasis during aging, and suggest that LRRK2 mutations may cause Parkinsons disease and cell death via impairment of protein degradation pathways, leading to α-synuclein accumulation and aggregation over time.


Nature | 2000

Embryonic lethality in mice homozygous for a processing-deficient allele of Notch1.

Stacey S. Huppert; Anh Le; Eric H. Schroeter; Jeff S. Mumm; Meera T. Saxena; Laurie A. Milner; Raphael Kopan

The Notch genes encode single-pass transmembrane receptors that transduce the extracellular signals responsible for cell fate determination during several steps of metazoan development. The mechanism by which extracellular signals affect gene transcription and ultimately cell fate decisions is beginning to emerge for the Notch signalling pathway. One paradigm is that ligand binding to Notch triggers a Presenilin1-dependent proteolytic release of the Notch intracellular domain from the membrane, resulting in low amounts of Notch intracellular domain which form a nuclear complex with CBF1/Su(H)/Lag1 to activate transcription of downstream targets. Not all observations clearly support this processing model, and the most rigorous test of it is to block processing in vivo and then determine the ability of unprocessed Notch to signal. Here we report that the phenotypes associated with a single point mutation at the intramembranous processing site of Notch1, Val1,744→Gly, resemble the null Notch1 phenotype. Our results show that efficient intramembranous processing of Notch1 is indispensable for embryonic viability and proper early embryonic development in vivo.


Journal of Biological Chemistry | 1999

A loss of function mutation of presenilin-2 interferes with amyloid beta-peptide production and notch signaling.

Harald Steiner; Karen Duff; Anja Capell; Helmut Romig; Melissa G. Grim; Sarah Lincoln; John Hardy; Xin Yu; Melanie Picciano; Katja Fechteler; Martin Citron; Raphael Kopan; Brigitte Pesold; Simone Keck; Miriam Baader; Taisuke Tomita; Takeshi Iwatsubo; Ralf Baumeister; Christian Haass

Presenilin-1 (PS1) facilitates γ-secretase cleavage of the β-amyloid precursor protein and the intramembraneous cleavage of Notch1. Although Alzheimer’s disease-associated mutations in the homologous presenilin (PS2) gene elevate amyloid β-peptide (Aβ42) production like PS1 mutations, here we demonstrate that a gene ablation of PS2 (unlike that of PS1) in mice does not result in a severe phenotype resembling that of Notch-ablated animals. To investigate the amyloidogenic function of PS2 more directly, we mutagenized a conserved aspartate at position 366 to alanine, because the corresponding residue of PS1 is known to be required for its amyloidogenic function. Cells expressing the PS2 D366A mutation exhibit significant deficits in proteolytic processing of β-amyloid precursor protein indicating a defect in γ-secretase activity. The reduced γ-secretase activity results in the almost complete inhibition of Aβ and p3 production in cells stably expressing PS2 D366A, whereas cells overexpressing the wild-type PS2 cDNA produce robust levels of Aβ and p3. Using highly sensitive in vivo assays, we demonstrate that the PS2 D366A mutation not only blocks γ-secretase activity but also inactivates PS2 activity in Notch signaling by inhibiting the proteolytic release of the cytoplasmic Notch1 domain. These data suggest that PS2 is functionally involved in Aβ production and Notch signaling by facilitating similar proteolytic cleavages.

Collaboration


Dive into the Raphael Kopan's collaboration.

Top Co-Authors

Avatar

Zhenyi Liu

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Ma. Xenia G. Ilagan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric H. Schroeter

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Stacey S. Huppert

Cincinnati Children's Hospital Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hui-Teng Cheng

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Jeff S. Mumm

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Ahu Turkoz

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Alison Goate

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge