Network


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

Hotspot


Dive into the research topics where Simone Prömel is active.

Publication


Featured researches published by Simone Prömel.


Nature | 2012

TNF receptor 1 genetic risk mirrors outcome of anti-TNF therapy in multiple sclerosis.

Adam Patrick Gregory; Calliope A. Dendrou; Kathrine E. Attfield; Aiden Haghikia; Dionysia K. Xifara; Falk Butter; Gereon Poschmann; Gurman Kaur; Lydia Lambert; Oliver A. Leach; Simone Prömel; Divya Punwani; James H. Felce; Simon J. Davis; Ralf Gold; Finn C. Nielsen; Richard M. Siegel; Matthias Mann; John I. Bell; Gil McVean; Lars Fugger

Although there has been much success in identifying genetic variants associated with common diseases using genome-wide association studies (GWAS), it has been difficult to demonstrate which variants are causal and what role they have in disease. Moreover, the modest contribution that these variants make to disease risk has raised questions regarding their medical relevance. Here we have investigated a single nucleotide polymorphism (SNP) in the TNFRSF1A gene, that encodes tumour necrosis factor receptor 1 (TNFR1), which was discovered through GWAS to be associated with multiple sclerosis (MS), but not with other autoimmune conditions such as rheumatoid arthritis, psoriasis and Crohn’s disease. By analysing MS GWAS data in conjunction with the 1000 Genomes Project data we provide genetic evidence that strongly implicates this SNP, rs1800693, as the causal variant in the TNFRSF1A region. We further substantiate this through functional studies showing that the MS risk allele directs expression of a novel, soluble form of TNFR1 that can block TNF. Importantly, TNF-blocking drugs can promote onset or exacerbation of MS, but they have proven highly efficacious in the treatment of autoimmune diseases for which there is no association with rs1800693. This indicates that the clinical experience with these drugs parallels the disease association of rs1800693, and that the MS-associated TNFR1 variant mimics the effect of TNF-blocking drugs. Hence, our study demonstrates that clinical practice can be informed by comparing GWAS across common autoimmune diseases and by investigating the functional consequences of the disease-associated genetic variation.


Pharmacological Reviews | 2015

International Union of Basic and Clinical Pharmacology. XCIV. Adhesion G Protein–Coupled Receptors

Jörg Hamann; Gabriela Aust; Demet Araç; Felix B. Engel; Caroline J. Formstone; Robert Fredriksson; Randy A. Hall; Breanne L. Harty; Christiane Kirchhoff; Barbara Knapp; Arunkumar Krishnan; Ines Liebscher; Hsi-Hsien Lin; David C. Martinelli; Kelly R. Monk; Miriam C. Peeters; Xianhua Piao; Simone Prömel; Torsten Schöneberg; Thue W. Schwartz; Kathleen Singer; Martin Stacey; Yuri A. Ushkaryov; Mario Vallon; Uwe Wolfrum; Mathew W. Wright; Lei Xu; Tobias Langenhan; Helgi B. Schiöth

The Adhesion family forms a large branch of the pharmacologically important superfamily of G protein–coupled receptors (GPCRs). As Adhesion GPCRs increasingly receive attention from a wide spectrum of biomedical fields, the Adhesion GPCR Consortium, together with the International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification, proposes a unified nomenclature for Adhesion GPCRs. The new names have ADGR as common dominator followed by a letter and a number to denote each subfamily and subtype, respectively. The new names, with old and alternative names within parentheses, are: ADGRA1 (GPR123), ADGRA2 (GPR124), ADGRA3 (GPR125), ADGRB1 (BAI1), ADGRB2 (BAI2), ADGRB3 (BAI3), ADGRC1 (CELSR1), ADGRC2 (CELSR2), ADGRC3 (CELSR3), ADGRD1 (GPR133), ADGRD2 (GPR144), ADGRE1 (EMR1, F4/80), ADGRE2 (EMR2), ADGRE3 (EMR3), ADGRE4 (EMR4), ADGRE5 (CD97), ADGRF1 (GPR110), ADGRF2 (GPR111), ADGRF3 (GPR113), ADGRF4 (GPR115), ADGRF5 (GPR116, Ig-Hepta), ADGRG1 (GPR56), ADGRG2 (GPR64, HE6), ADGRG3 (GPR97), ADGRG4 (GPR112), ADGRG5 (GPR114), ADGRG6 (GPR126), ADGRG7 (GPR128), ADGRL1 (latrophilin-1, CIRL-1, CL1), ADGRL2 (latrophilin-2, CIRL-2, CL2), ADGRL3 (latrophilin-3, CIRL-3, CL3), ADGRL4 (ELTD1, ETL), and ADGRV1 (VLGR1, GPR98). This review covers all major biologic aspects of Adhesion GPCRs, including evolutionary origins, interaction partners, signaling, expression, physiologic functions, and therapeutic potential.


Developmental Cell | 2009

Latrophilin Signaling Links Anterior-Posterior Tissue Polarity and Oriented Cell Divisions in the C. elegans Embryo

Tobias Langenhan; Simone Prömel; Behrooz Esmaeili; Helen Waller-Evans; Christian Hennig; Yuji Kohara; Leon Avery; Ioannis Vakonakis; Ralf Schnabel; Andreas P. Russ

Understanding the mechanisms that coordinate the orientation of cell division planes during embryogenesis and morphogenesis is a fundamental problem in developmental biology. Here we show that the orphan receptor lat-1, a homolog of vertebrate latrophilins, plays an essential role in the establishment of tissue polarity in the C. elegans embryo. We provide evidence that lat-1 is required for the alignment of cell division planes to the anterior-posterior axis and acts in parallel to known polarity and morphogenesis signals. lat-1 is a member of the Adhesion-GPCR protein family and is structurally related to flamingo/CELSR, an essential component of the planar cell polarity pathway. We dissect the molecular requirements of lat-1 signaling and implicate lat-1 in an anterior-posterior tissue polarity pathway in the premorphogenesis stage of C. elegans development.


Cell Reports | 2012

The GPS Motif Is a Molecular Switch for Bimodal Activities of Adhesion Class G Protein-Coupled Receptors

Simone Prömel; Marie Frickenhaus; Samantha Hughes; David Staunton; Alison Woollard; Ioannis Vakonakis; Torsten Schöneberg; Ralf Schnabel; Andreas P. Russ; Tobias Langenhan

Summary Adhesion class G protein-coupled receptors (aGPCR) form the second largest group of seven-transmembrane-spanning (7TM) receptors whose molecular layout and function differ from canonical 7TM receptors. Despite their essential roles in immunity, tumorigenesis, and development, the mechanisms of aGPCR activation and signal transduction have remained obscure to date. Here, we use a transgenic assay to define the protein domains required in vivo for the activity of the prototypical aGPCR LAT-1/Latrophilin in Caenorhabditis elegans. We show that the GPCR proteolytic site (GPS) motif, the molecular hallmark feature of the entire aGPCR class, is essential for LAT-1 signaling serving in two different activity modes of the receptor. Surprisingly, neither mode requires cleavage but presence of the GPS, which relays interactions with at least two different partners. Our work thus uncovers the versatile nature of aGPCR activity in molecular detail and places the GPS motif in a central position for diverse protein-protein interactions.


Structure | 2008

Solution structure and sugar-binding mechanism of mouse latrophilin-1 RBL: a 7TM receptor-attached lectin-like domain.

Ioannis Vakonakis; Tobias Langenhan; Simone Prömel; Andreas P. Russ; Iain D. Campbell

Summary Latrophilin-1 (Lat-1), a target receptor for α-Latrotoxin, is a putative G protein-coupled receptor implicated in synaptic function. The extracellular portion of Lat-1 contains a rhamnose binding lectin (RBL)-like domain of unknown structure. RBL domains, first isolated from the eggs of marine species, are also found in the ectodomains of other metazoan transmembrane proteins, including a recently discovered coreceptor of the neuronal axon guidance molecule SLT-1/Slit. Here, we describe a structure of this domain from the mouse Lat-1. RBL adopts a unique α/β fold with long structured loops important for monosaccharide recognition, as shown in the structure of a complex with L-rhamnose. Sequence alignments and mutagenesis show that residues important for carbohydrate binding are often absent in other receptor-attached examples of RBL, including the SLT-1/Slit coreceptor. We postulate that this domain class facilitates direct protein-protein interactions in many transmembrane receptors.


PLOS ONE | 2010

The Orphan Adhesion-GPCR GPR126 Is Required for Embryonic Development in the Mouse

Helen Waller-Evans; Simone Prömel; Tobias Langenhan; John Dixon; Dirk Zahn; William H. Colledge; Joanne Doran; Mark B. L. Carlton; Ben Davies; Samuel Aparicio; Johannes Grosse; Andreas P. Russ

Adhesion-GPCRs provide essential cell-cell and cell-matrix interactions in development, and have been implicated in inherited human diseases like Usher Syndrome and bilateral frontoparietal polymicrogyria. They are the second largest subfamily of seven-transmembrane spanning proteins in vertebrates, but the function of most of these receptors is still not understood. The orphan Adhesion-GPCR GPR126 has recently been shown to play an essential role in the myelination of peripheral nerves in zebrafish. In parallel, whole-genome association studies have implicated variation at the GPR126 locus as a determinant of body height in the human population. The physiological function of GPR126 in mammals is still unknown. We describe a targeted mutation of GPR126 in the mouse, and show that GPR126 is required for embryonic viability and cardiovascular development.


Trends in Pharmacological Sciences | 2013

Matching structure with function: the GAIN domain of Adhesion-GPCR and PKD1-like proteins

Simone Prömel; Tobias Langenhan; Demet Araç

Elucidation of structural information can greatly facilitate the understanding of molecular function. A recent example is the description of the G-protein-coupled receptor (GPCR) autoproteolysis-inducing (GAIN) domain, an evolutionarily ancient fold present in Adhesion-GPCRs (aGPCRs) and polycystic kidney disease 1 (PKD1)-like proteins. In the past, the peculiar autoproteolytic capacity of both membrane protein families at the conserved GPCR proteolysis site (GPS) had not been described in detail. The physiological performance of aGPCRs and PKD1-like proteins is thought to be regulated through the GPS, but it is debated how. A recent report provides pivotal details by discovery and analysis of the GAIN domain structure that incorporates the GPS motif. Complementary studies have commenced to analyze physiological requirements of the GAIN domain for aGPCR function, indicating that it serves as the linchpin for multiple receptor signals. Structural analysis and functional assays now allow for the dissection of the biological duties conferred through the GAIN domain.


Biological Chemistry | 2013

Progress in demystification of adhesion G protein-coupled receptors.

Ines Liebscher; Torsten Schöneberg; Simone Prömel

Abstract Adhesion G protein-coupled receptors (aGPCR) form the second largest class of GPCR. They are phylogenetically old and have been highly conserved during evolution. Mutations in representatives of this class are associated with severe diseases such as Usher Syndrome, a combined congenital deaf-blindness, or bifrontal parietal polymicrogyria. The main characteristics of aGPCR are their enormous size and the complexity of their N termini. They contain a highly conserved GPCR proteolytic site (GPS) and several functional domains that have been implicated in cell-cell and cell-matrix interactions. Adhesion GPCR have been proposed to serve a dual function as adhesion molecules and as classical receptors. However, until recently there was no proof that aGPCR indeed couple to G proteins or even function as classical receptors. In this review, we have summarized and discussed recent evidence that aGPCR present many functional features of classical GPCR, including multiple G protein-coupling abilities, G protein-independent signaling and oligomerization, but also specific signaling properties only found in aGPCR.


Developmental Dynamics | 2012

Characterization and Functional Study of a Cluster of Four Highly Conserved Orphan Adhesion-GPCR in Mouse

Simone Prömel; Helen Waller-Evans; John Dixon; Dirk Zahn; William H. Colledge; Joanne Doran; Mark B. L. Carlton; Johannes Grosse; Torsten Schöneberg; Andreas P. Russ; Tobias Langenhan

Background: Adhesion G protein‐coupled receptors (aGPCR) constitute a structurally and functionally diverse class of seven‐transmembrane receptor proteins. Although for some of the members important roles in immunology, neurology, as well as developmental biology have been suggested, most receptors have been poorly characterized. Results: We have studied evolution, expression, and function of an entire receptor group containing four uncharacterized aGPCR: Gpr110, Gpr111, Gpr115, and Gpr116. We show that the genomic loci of these four receptors are clustered tightly together in mouse and human genomes and that this cluster likely derives from a single common ancestor gene. Using transcriptional profiling on wild‐type and knockout/LacZ reporter knockin mice strains, we have obtained detailed expression maps that show ubiquitous expression of Gpr116, co‐expression of Gpr111 and Gpr115 in developing skin, and expression of Gpr110 in adult kidney. Loss of Gpr110, Gpr111, or Gpr115 function did not result in detectable defects, indicating that genes of this aGPCR group might function redundantly. Conclusions: The aGPCR cluster Gpr110, Gpr111, Gpr115, and Gpr116 developed from one common ancestor in vertebrates. Expression suggests a role in epithelia, and one can speculate about a possible redundant function of GPR111 and GPR115. Developmental Dynamics 241:1591–1602, 2012.


Annals of the New York Academy of Sciences | 2012

Dissecting signaling and functions of adhesion G protein-coupled receptors.

Demet Araç; Gabriela Aust; Davide Calebiro; Felix B. Engel; Caroline J. Formstone; André M. Goffinet; Jörg Hamann; Robert J. Kittel; Ines Liebscher; Hsi-Hsien Lin; Kelly R. Monk; Alexander G. Petrenko; Xianhua Piao; Simone Prömel; Helgi B. Schiöth; Thue W. Schwartz; Martin Stacey; Yuri A. Ushkaryov; Manja Wobus; Uwe Wolfrum; Lei Xu; Tobias Langenhan

G protein–coupled receptors (GPCRs) comprise an expanded superfamily of receptors in the human genome. Adhesion class G protein–coupled receptors (adhesion‐GPCRs) form the second largest class of GPCRs. Despite the abundance, size, molecular structure, and functions in facilitating cell and matrix contacts in a variety of organ systems, adhesion‐GPCRs are by far the most poorly understood GPCR class. Adhesion‐GPCRs possess a unique molecular structure, with extended N‐termini containing various adhesion domains. In addition, many adhesion‐GPCRs are autoproteolytically cleaved into an N‐terminal fragment (NTF, NT, α‐subunit) and C‐terminal fragment (CTF, CT, β‐subunit) at a conserved GPCR autoproteolysis–inducing (GAIN) domain that contains a GPCR proteolysis site (GPS). These two features distinguish adhesion‐GPCRs from other GPCR classes. Though active research on adhesion‐GPCRs in diverse areas, such as immunity, neuroscience, and development and tumor biology has been intensified in the recent years, the general biological and pharmacological properties of adhesion‐GPCRs are not well known, and they have not yet been used for biomedical purposes. The “6th International Adhesion‐GPCR Workshop,” held at the Institute of Physiology of the University of Würzburg on September 6–8, 2012, assembled a majority of the investigators currently actively pursuing research on adhesion‐GPCRs, including scientists from laboratories in Europe, the United States, and Asia. The meeting featured the nascent mechanistic understanding of the molecular events driving the signal transduction of adhesion‐GPCRs, novel models to evaluate their functions, and evidence for their involvement in human disease.

Collaboration


Dive into the Simone Prömel'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

Ralf Schnabel

Braunschweig University of Technology

View shared research outputs
Top Co-Authors

Avatar

Felix B. Engel

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge