Network


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

Hotspot


Dive into the research topics where Rolf Kemler is active.

Publication


Featured researches published by Rolf Kemler.


The EMBO Journal | 1997

β‐catenin is a target for the ubiquitin–proteasome pathway

Hermann Aberle; Andreas Bauer; Jörg Stappert; Andreas Kispert; Rolf Kemler

β‐catenin is a central component of the cadherin cell adhesion complex and plays an essential role in the Wingless/Wnt signaling pathway. In the current model of this pathway, the amount of β‐catenin (or its invertebrate homolog Armadillo) is tightly regulated and its steady‐state level outside the cadherin–catenin complex is low in the absence of Wingless/Wnt signal. Here we show that the ubiquitin‐dependent proteolysis system is involved in the regulation of β‐catenin turnover. β‐catenin, but not E‐cadherin, p120cas or α‐catenin, becomes stabilized when proteasome‐mediated proteolysis is inhibited and this leads to the accumulation of multi‐ubiquitinated forms of β‐catenin. Mutagenesis experiments demonstrate that substitution of the serine residues in the glycogen synthase kinase 3β (GSK3β) phosphorylation consensus motif of β‐catenin inhibits ubiquitination and results in stabilization of the protein. This motif in β‐catenin resembles a motif in IκB (inhibitor of NFκB) which is required for the phosphorylation‐dependent degradation of IκB via the ubiquitin–proteasome pathway. We show that ubiquitination of β‐catenin is greatly reduced in Wnt‐expressing cells, providing the first evidence that the ubiquitin–proteasome degradation pathway may act downstream of GSK3β in the regulation of β‐catenin.


The EMBO Journal | 1989

The cytoplasmic domain of the cell adhesion molecule uvomorulin associates with three independent proteins structurally related in different species.

Masayuki Ozawa; H Baribault; Rolf Kemler

Uvomorulin belongs to the group of Ca2+‐dependent cell adhesion molecules, which are integral membrane proteins with several structural features in common. In particular, the cytoplasmic part of these proteins is highly conserved in different species, suggesting a common biological function. To test this assumption we transfected a uvomorulin full‐length cDNA into uvomorulin‐negative mouse NIH 3T3 and L cells. Immunoprecipitations with anti‐uvomorulin antibodies detected, in addition to uvomorulin, three independent proteins of 102, 88 and 80 kd which are of host origin and which form complexes with uvomorulin. Using cDNA constructs coding for uvomorulin with cytoplasmic or extracellular deletions it is shown that the 102, 88 and 80 kd proteins complex with the cytoplasmic domain of uvomorulin. Peptide pattern analysis revealed that these three proteins are identical in different mouse cells. When uvomorulin cDNA was introduced into cell lines from other species, such as human HeLa and avian fibroblasts, the expressed uvomorulin was also associated with endogenous 102, 88 and 80 kd proteins and, moreover, each of these proteins showed structural similarities to the respective mouse molecule. A panel of antibodies specific for known cytoplasmic proteins of mol. wts similar to those of the three proteins did not react with any of the described components. This suggests that the 102, 88 and 80 kd proteins constitute a new group of proteins for which we propose the nomenclature of catenin alpha, beta and gamma respectively. The characterization of these proteins provides a first molecular basis for a possible cytoplasmic anchorage of uvomorulin to the cytoskeleton.


Trends in Genetics | 1993

From cadherins to catenins: cytoplasmic protein interactions and regulation of cell adhesion

Rolf Kemler

Classical cadherins are complexed via their cytoplasmic domains with alpha-, beta- and gamma-catenin. This complex formation links cadherins to the actin filament network and to other transmembrane and cytoplasmic proteins. alpha-Catenin is homologous to vinculin, and beta-catenin to the product of the Drosophila gene armadillo, while gamma-catenin seems to be identical to plakoglobin. Catenins are part of a higher order protein structure that is of crucial importance for the adhesive function of cadherins. A working model of the construction and regulation of this multiprotein interaction is proposed.


Mechanisms of Development | 1996

Nuclear localization of β-catenin by interaction with transcription factor LEF-1

Otmar Huber; Reinhard Korn; John McLaughlin; Mami Ohsugi; Bernhard G. Herrmann; Rolf Kemler

Vertebrate beta-catenin and Drosophila Armadillo share structural similarities suggesting that beta-catenin, like Armadillo, has a developmental signaling function. Both proteins are present as components of cell adherens junctions, but accumulate in the cytoplasm upon Wingless/Wnt signaling. beta-Catenin has axis-inducing properties like Wnt when injected into Xenopus blastomeres, providing evidence for participation of beta-catenin in the Wnt-pathway, but until now no downstream targets for beta-catenin have been identified. Here we demonstrate that beta-catenin binds to the HMG-type transcription factor lymphoid enhancer factor-1 (LEF-1), resulting in a nuclear translocation of beta-catenin both in cultured mouse cells and after ectopic expression of LEF-1 in two-cell mouse embryos. LEF-1/beta-catenin complexes bind to the promoter region of the E-cadherin gene in vitro, suggesting that this interaction could regulate E-cadherin transcription. As shown for beta-catenin, ectopic expression of LEF-1 in Xenopus embryos caused duplication of the body axis, indicating a regulatory role for a LEF-1-like molecule in dorsal mesoderm formation.


Journal of Cellular Biochemistry | 1996

Cadherin-catenin complex: protein interactions and their implications for cadherin function.

Hermann Aberle; Hillel Schwartz; Rolf Kemler

Cadherins comprise a family of calcium‐dependent glycoproteins that function in mediating cell‐cell adhesion in virtually all solid tissues of multicellular organisms. In epithelial cells, E‐cadherin represents a key molecule in the establishment and stabilization of cellular junctions. On the cellular level, E‐cadherin is concentrated at the adherens junction and interacts homophilically with E‐cadherin molecules of adjacent cells. Significant progress has been made in understanding the extra‐ and intracellular interactions of E‐cadherin. Recent success in solving the three‐dimensional structure of an extracellular cadherin domain provides a structural basis for understanding the homophilic interaction mechanism and the calcium requirement of cadherins. According to the crystal structure, individual cadherin molecules cooperate to form a linear cell adhesion zipper. The intracellular anchorage of cadherins is regulated by the dynamic association with cytoplasmic proteins, termed catenins. The cytoplasmic domain of E‐cadherin is complexed with either β‐catenin or plakoglobin (γ‐catenin). β‐catenin and plakoglobin bind directly to α‐catenin, giving rise to two distinct cadherin‐catenin complexes (CCC). α‐catenin is thought to link both CCCs to actin filaments. The anchorage of cadherins to the cytoskeleton appears to be regulated by tyrosine phosphorylation. Phosphorylation‐induced junctional disassembly targets the catenins, indicating that catenins are components of signal transduction pathways. The unexpected association of catenins with the product of the tumor suppressor gene APC has led to the discovery of a second, cadherin‐independent catenin complex. Two separate catenin complexes are therefore involved in the cross‐talk between cell adhesion and signal transduction. In this review we focus on protein interactions regulating the molecular architecture and function of the CCC. In the light of a fundamental role of the CCC during mammalian development and tissue morphogenesis, we also discuss the phenotypes of embryos lacking E‐cadherin or β‐catenin.


The EMBO Journal | 2000

The p300/CBP acetyltransferases function as transcriptional coactivators of β‐catenin in vertebrates

Andreas Hecht; Kris Vleminckx; Marc P. Stemmler; Frans van Roy; Rolf Kemler

Wnt growth factors regulate a variety of developmental processes by altering specific gene expression patterns. In vertebrates β‐catenin acts as transcriptional activator, which is needed to overcome target gene repression by Groucho/TLE proteins, and to permit promoter activation as the final consequence of Wnt signaling. However, the molecular mechanisms of transcriptional activation by β‐catenin are only poorly understood. Here we demonstrate that the closely related acetyltransferases p300 and CBP potentiate β‐catenin‐mediated activation of the siamois promoter, a known Wnt target. β‐catenin and p300 also synergize to stimulate a synthetic reporter gene construct, whereas activation of the cyclin D1 promoter by β‐catenin is refractory to p300 stimulation. Axis formation and activation of the β‐catenin target genes siamois and Xnr‐3 in Xenopus embryos are sensitive to the E1A oncoprotein, a known inhibitor of p300/CBP. The C‐terminus of β‐catenin interacts directly with a region overlapping the CH‐3 domain of p300. p300 could participate in alleviating promoter repression imposed by chromatin structure and in recruiting the basal transcription machinery to promoters of particular Wnt target genes.


Cell | 2002

Identification of a Wnt/Dvl/β-Catenin → Pitx2 Pathway Mediating Cell-Type-Specific Proliferation during Development

Chrissa Kioussi; Paola Briata; Sung Hee Baek; David W. Rose; Natasha S. Hamblet; Thomas Herman; Kenneth A. Ohgi; Chijen Lin; Anatoli S. Gleiberman; Jianbo Wang; Véronique Brault; Pilar Ruiz-Lozano; H.D. Nguyen; Rolf Kemler; Christopher K. Glass; Anthony Wynshaw-Boris; Michael G. Rosenfeld

Understanding the cell type-specific molecular mechanisms by which distinct signaling pathways combinatorially control proliferation during organogenesis is a central issue in development and disease. Here, we report that the bicoid-related transcription factor Pitx2 is rapidly induced by the Wnt/Dvl/beta-catenin pathway and is required for effective cell-type-specific proliferation by directly activating specific growth-regulating genes. Regulated exchange of HDAC1/beta-catenin converts Pitx2 from repressor to activator, analogous to control of TCF/LEF1. Pitx2 then serves as a competence factor required for the temporally ordered and growth factor-dependent recruitment of a series of specific coactivator complexes that prove necessary for Cyclin D2 gene induction. The molecular strategy underlying interactions between the Wnt and growth factor-dependent signaling pathways in cardiac outflow tract and pituitary proliferation is likely to be prototypic of cell-specific proliferation strategies in other tissues.


Developmental Cell | 2003

Wnt3a plays a major role in the segmentation clock controlling somitogenesis.

Alexander Aulehla; Christian Wehrle; Beate Brand-Saberi; Rolf Kemler; Achim Gossler; Benoît Kanzler; Bernhard G. Herrmann

The vertebral column derives from somites generated by segmentation of presomitic mesoderm (PSM). Somitogenesis involves a molecular oscillator, the segmentation clock, controlling periodic Notch signaling in the PSM. Here, we establish a novel link between Wnt/beta-catenin signaling and the segmentation clock. Axin2, a negative regulator of the Wnt pathway, is directly controlled by Wnt/beta-catenin and shows oscillating expression in the PSM, even when Notch signaling is impaired, alternating with Lfng expression. Moreover, Wnt3a is required for oscillating Notch signaling activity in the PSM. We propose that the segmentation clock is established by Wnt/beta-catenin signaling via a negative-feedback mechanism and that Wnt3a controls the segmentation process in vertebrates.


Cell | 1990

Novel function of the cell adhesion molecule uvomorulin as an inducer of cell surface polarity

Helen McNeill; Masayuki Ozawa; Rolf Kemler; W. James Nelson

Na+,K(+)-ATPase has distinctly different distributions in mesenchymal cells, where it has an unrestricted distribution over the entire cell surface, compared with polarized epithelial cells, where it is restricted to the basal-lateral membrane domain. The generation of this restricted distribution is important in mesenchyme to epithelia conversion in development and the function of transporting epithelia, but the mechanisms involved are unknown. Here we show that expression of the epithelial CAM uvomorulin in transfected fibroblasts is sufficient to induce a redistribution of Na+,K(+)-ATPase to sites of uvomorulin-mediated cell-cell contacts, similar to that in polarized epithelial cells. This restricted distribution of Na+,K(+)-ATPase occurs in the absence of tight junctions but coincides with the reorganization of the membrane cytoskeleton. The results indicate a direct role for CAMs as inducers of cell surface polarity of selective cytoplasmic and membrane proteins.


Journal of Cell Biology | 2003

Contact inhibition of VEGF-induced proliferation requires vascular endothelial cadherin, β-catenin, and the phosphatase DEP-1/CD148

Maria Grazia Lampugnani; Adriana Zanetti; Monica Corada; Takamune Takahashi; Giovanna Balconi; Ferruccio Breviario; Fabrizio Orsenigo; Anna Cattelino; Rolf Kemler; Thomas O. Daniel; Elisabetta Dejana

Confluent endothelial cells respond poorly to the proliferative signals of VEGF. Comparing isogenic endothelial cells differing for vascular endothelial cadherin (VE-cadherin) expression only, we found that the presence of this protein attenuates VEGF-induced VEGF receptor (VEGFR) 2 phosphorylation in tyrosine, p44/p42 MAP kinase phosphorylation, and cell proliferation. VE-cadherin truncated in β-catenin but not p120 binding domain is unable to associate with VEGFR-2 and to induce its inactivation. β-Catenin–null endothelial cells are not contact inhibited by VE-cadherin and are still responsive to VEGF, indicating that this protein is required to restrain growth factor signaling. A dominant-negative mutant of high cell density–enhanced PTP 1 (DEP-1)//CD148 as well as reduction of its expression by RNA interference partially restore VEGFR-2 phosphorylation and MAP kinase activation. Overall the data indicate that VE-cadherin–β-catenin complex participates in contact inhibition of VEGF signaling. Upon stimulation with VEGF, VEGFR-2 associates with the complex and concentrates at cell–cell contacts, where it may be inactivated by junctional phosphatases such as DEP-1. In sparse cells or in VE-cadherin–null cells, this phenomenon cannot occur and the receptor is fully activated by the growth factor.

Collaboration


Dive into the Rolf Kemler'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

Marc P. Stemmler

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge