23 research outputs found

    Signal Transduction and Pathogenic Modifications at the Melanocortin-4 Receptor: A Structural Perspective

    Get PDF
    The melanocortin-4 receptor (MC4R) can be endogenously activated by binding of melanocyte-stimulating hormones (MSH), which mediates anorexigenic effects. In contrast, the agouti-related peptide (AgRP) acts as an endogenous inverse agonist and suppresses ligand-independent basal signaling activity (orexigenic effects). Binding of ligands to MC4R leads to the activation of different G-protein subtypes or arrestin and concomitant signaling pathways. This receptor is a key protein in the hypothalamic regulation of food intake and energy expenditure and naturally-occurring inactivating MC4R variants are the most frequent cause of monogenic obesity. In general, obesity is a growing problem on a global scale and is of social, medical, and economic relevance. A significant goal is to develop optimized pharmacological tools targeting MC4R without adverse effects. To date, this has not been achieved because of inter alia non-selective ligands across the five functionally different MCR subtypes (MC1-5R). This motivates further investigation of (i) the three-dimensional MC4R structure, (ii) binding mechanisms of various ligands, and (iii) the molecular transfer process of signal transduction, with the aim of understanding how structural features are linked with functional-physiological aspects. Unfortunately, experimentally elucidated structural information is not yet available for theMC receptors, a group of class A G-protein coupled receptors (GPCRs). We, therefore, generated MC4R homology models and complexes with interacting partners to describe approximate structural properties associated with signaling mechanisms. In addition, molecular insights from pathogenic mutations were incorporated to discriminate more precisely their individual malfunction of the signal transfer mechanism

    Expression and Characterization of Relaxin Family Peptide Receptor 1 Variants

    Get PDF
    G-protein coupled receptors (GPCR) transduce extracellular stimuli into the cell interior and are thus centrally involved in almost all physiological-neuronal processes. This essential function and association with many diseases or pathological conditions explain why GPCRs are one of the priority targets in medical and pharmacological research, including structure determination. Despite enormous experimental efforts over the last decade, both the expression and purification of these membrane proteins remain elusive. This is attributable to specificities of each GPCR subtype and the finding of necessary experimental in vitro conditions, such as expression in heterologous cell systems or with accessory proteins. One of these specific GPCRs is the leucine-rich repeat domain (LRRD) containing GPCR 7 (LGR7), also termed relaxin family peptide receptor 1 (RXFP1). This receptor is characterized by a large extracellular region of around 400 amino acids constituted by several domains, a rare feature among rhodopsin-like (class A) GPCRs. In the present study, we describe the expression and purification of RXFP1, including the design of various constructs suitable for functional/biophysical studies and structure determination. Based on available sequence information, homology models, and modern biochemical and genetic tools, several receptor variations with different purification tags and fusion proteins were prepared and expressed in Sf9 cells (small-scale), followed by an analytic fluorescence-detection size-exclusion chromatography (F-SEC) to evaluate the constructs. The most promising candidates were expressed and purified on a large-scale, accompanied by ligand binding studies using surface plasmon resonance spectroscopy (SPR) and by determination of signaling capacities. The results may support extended studies on RXFP1 receptor constructs serving as targets for small molecule ligand screening or structural elucidation by protein X-ray crystallography or cryo-electron microscopy

    Molecular Effects of Auto-Antibodies on Angiotensin II Type 1 Receptor Signaling and Cell Proliferation

    Get PDF
    The angiotensin II (Ang II) type 1 receptor (AT1R) is involved in the regulation of blood pressure (through vasoconstriction) and water and ion homeostasis (mediated by interaction with the endogenous agonist). AT1R can also be activated by auto-antibodies (AT1R-Abs), which are associated with manifold diseases, such as obliterative vasculopathy, preeclampsia and systemic sclerosis. Knowledge of the molecular mechanisms related to AT1R-Abs binding and associated signaling cascade (dys-)regulation remains fragmentary. The goal of this study was, therefore, to investigate details of the effects of AT1R-Abs on G-protein signaling and subsequent cell proliferation, as well as the putative contribution of the three extracellular receptor loops (ELs) to Abs-AT1R signaling. AT1R-Abs induced nuclear factor of activated T-cells (NFAT) signaling, which reflects Gq/11 and Gi activation. The impact on cell proliferation was tested in different cell systems, as well as activation-triggered receptor internalization. Blockwise alanine substitutions were designed to potentially investigate the role of ELs in AT1R-Abs-mediated effects. First, we demonstrate that Ang II-mediated internalization of AT1R is impeded by binding of AT1R-Abs. Secondly, exclusive AT1RAbs- induced Gq/11 activation is most significant for NFAT stimulation and mediates cell proliferation. Interestingly, our studies also reveal that ligand-independent, baseline AT1R activation of Gi signaling has, in turn, a negative effect on cell proliferation. Indeed, inhibition of Gi basal activity potentiates proliferation triggered by AT1R-Abs. Finally, although AT1R containing EL1 and EL3 blockwise alanine mutations were not expressed on the human embryonic kidney293T (HEK293T) cell surface, we at least confirmed that parts of EL2 are involved in interactions between AT1R and Abs. This current study thus provides extended insights into the molecular action of AT1R-Abs and associated mechanisms of interrelated pathogenesis

    Structural snapshot of a bacterial phytochrome in its functional intermediate state

    Get PDF
    Phytochromes are modular photoreceptors of plants, bacteria and fungi that use light as a source of information to regulate fundamental physiological processes. Interconversion between the active and inactive states is accomplished by a photoinduced reaction sequence which couples the sensor with the output module. However, the underlying molecular mechanism is yet not fully understood due to the lack of structural data of functionally relevant intermediate states. Here we report the crystal structure of a Meta-F intermediate state of an Agp2 variant from Agrobacterium fabrum. This intermediate, the identity of which was verified by resonance Raman spectroscopy, was formed by irradiation of the parent Pfr state and displays significant reorientations of almost all amino acids surrounding the chromophore. Structural comparisons allow identifying structural motifs that might serve as conformational switch for initiating the functional secondary structure change that is linked to the (de-)activation of these photoreceptors

    MicroRNA-100-5p and microRNA-298-5p released from apoptotic cortical neurons are endogenous Toll-like receptor 7/8 ligands that contribute to neurodegeneration

    Get PDF
    Background: MicroRNA (miRNA) expression in the brain is altered in neurodegenerative diseases. Recent studies demonstrated that selected miRNAs conventionally regulating gene expression at the post-transcriptional level can act extracellularly as signaling molecules. The identity of miRNA species serving as membrane receptor ligands involved in neuronal apoptosis in the central nervous system (CNS), as well as the miRNAs' sequence and structure required for this mode of action remained largely unresolved. Methods. Using a microarray-based screening approach we analyzed apoptotic cortical neurons of C56BL/6 mice and their supernatant with respect to alterations in miRNA expression/presence. HEK-Blue Toll-like receptor (TLR) 7/8 reporter cells, primary microglia and macrophages derived from human and mouse were employed to test the potential of the identified miRNAs released from apoptotic neurons to serve as signaling molecules for the RNA-sensing receptors. Biophysical and bioinformatical approaches, as well as immunoassays and sequential microscopy were used to analyze the interaction between candidate miRNA and TLR. Immunocytochemical and -histochemical analyses of murine CNS cultures and adult mice intrathecally injected with miRNAs, respectively, were performed to evaluate the impact of miRNA-induced TLR activation on neuronal survival and microglial activation. Results: We identified a specific pattern of miRNAs released from apoptotic cortical neurons that activate TLR7 and/or TLR8, depending on sequence and species. Exposure of microglia and macrophages to certain miRNA classes released from apoptotic neurons resulted in the sequence-specific production of distinct cytokines/chemokines and increased phagocytic activity. Out of those miRNAs miR-100-5p and miR-298-5p, which have consistently been linked to neurodegenerative diseases, entered microglia, located to their endosomes, and directly bound to human TLR8. The miRNA-TLR interaction required novel sequence features, but no specific structure formation of mature miRNA. As a consequence of miR-100-5p- and miR-298-5p-induced TLR activation, cortical neurons underwent cell-autonomous apoptosis. Presence of miR-100-5p and miR-298-5p in cerebrospinal fluid led to neurodegeneration and microglial accumulation in the murine cerebral cortex through TLR7 signaling. Conclusion: Our data demonstrate that specific miRNAs are released from apoptotic cortical neurons, serve as endogenous TLR7/8 ligands, and thereby trigger further neuronal apoptosis in the CNS. Our findings underline the recently discovered role of miRNAs as extracellular signaling molecules, particularly in the context of neurodegeneration

    Structure-based redesign of the dimerization interface reduces the toxicity of zinc-finger nucleases

    Full text link
    Artificial endonucleases consisting of a Fokl cleavage domain tethered to engineered zinc-finger DNA-binding proteins have proven useful for stimulating homologous recombination in a variety of cell types. Because the catalytic domain of zinc-finger nucleases (ZFNs) must dimerize to become active, two subunits are typically assembled as heterodimers at the cleavage site. The use of ZFNs is often associated with significant cytotoxicity, presumably due to cleavage at off- target sites. Here we describe a structure- based approach to reducing off- target cleavage. Using in silico protein modeling and energy calculations, we increased the specificity of target site cleavage by preventing homodimerization and lowering the dimerization energy. Cell-based recombination assays confirmed that the modified ZFNs were as active as the original ZFNs but elicit significantly less genotoxicity. The improved safety profile may facilitate therapeutic application of the ZFN technology

    Regulation der Enzymaktivität der Restriktionsendonuklease EcoRII durch Autoinhibition

    Get PDF
    DNA-Restriktions und -Modifikationssysteme sind in Prokaryoten weit verbreitet und stellen einen wirksamen Schutz gegen das Eindringen mobiler genetischer Elemente dar. Sie kodieren für eine Restriktionsendonuklease (REase) und eine DNA-Methyltransferase (MTase) gleicher Nukleotidsequenz Spezifität. Die MTase methyliert die zelluläre DNA und schützt sie durch diesen epigenetischen Marker vor der Wirkung der REase. Die REase verhindert die Aufnahme fremder, unmethylierter DNA durch sequenzspezifische Spaltung. EcoRII ist eine REase, die für die effiziente DNA-Spaltung mindestens zwei Kopien ihrer Erkennungssequenz benötigt. Untersuchungen der EcoRII-Struktur und -Funktion offenbarten, dass das Protein aus zwei stabilen Domänen aufgebaut ist, wobei die N-terminale Domäne wie ein Repressor die C-terminale Domäne sterisch blockiert und deren katalytische Aktivität verhindert. Dieser als Autoinhibition bezeichnete und von eukaryotischen Proteinen gut bekannter Regulationsmechanismus wurde erstmals für eine REase vorgeschlagen. In dieser Arbeit konnten wir die Regulation der EcoRII-Enzymaktivität durch Autoinhibition auf molekularer Ebene beweisen. Wir identifizierten ß-Strang 1 (B1: 18YFVYIKR24) und a-Helix 2 (H2: 26SANDT30) als essenzielle inhibitorische Elemente der N-terminalen Domäne des EcoRII-Moleküls. Die Deletion von B1 oder H2 führte zu einer vollständigen Aufhebung der Autoinhibition. Darüber hinaus ist es uns gelungen, die 3D-Röntgenkristallstruktur von EcoRII mit 1,9 Å zu lösen und mit Hilfe von Computermodellen neue Interaktionen des Enzyms mit der DNA „minor groove“ zu beschreiben sowie eine Mg2+-Bindungstasche zu charakterisieren. Die Untersuchung der EcoRII-MTase durch limitierte Proteolyse zeigte, dass das Enzym in Abhängigkeit von der DNA-Sequenz und von seinen Kofaktoren, DNA auf unterschiedliche Weise binden kann. Kristallisierungsversuche der EcoRII-MTase in Anwesenheit der hemi-methylierten DNA-Erkennungssequenz ergaben erste diffraktierende Kristalle, deren Qualität optimiert werden muss und zur Strukturlösung führen soll.Restriction and modification systems are wide spread among prokaryotes and pre-sent an efficient protection against invasion of mobile genetic elements. In general, they code for a restriction endonuclease (REase) and a DNA-methyltransferase (MTase) of the same DNA specificity. The MTase methylates the cellular DNA and by this epigenetic marker protects it against the action of the REase. The REase pre-vents the entry of foreign unmethylated DNA by site-specific cleavage. EcoRII is an REase which needs at least two copies of the recognition sequence for efficient cleavage. Investigations of the EcoRII structure and function revealed that the pro-tein is composed of two stable domains: the N-terminal domain acts as a repressor by sterically blocking the C-terminal domain and thereby inhibiting its catalytic activity. This regulatory mechanism is known as autoinhibition and has been often described for eukaryotic proteins, but for the first time was proposed for a REase. In this work, we verified the regulation of the EcoRII enzyme activity by autoinhibition at the molecular level. We identified ß-strand 1 (B1: 18YFVYIKR24) and a-helix 2 (H2: 26SANDT30) as essential inhibitory elements of the N-terminal domain. Deletion of B1 or H2 caused a complete abolishment of the autoinhibition. Fur-thermore, we were able to solve the 3D-X-ray crystal structure of EcoRII at 1.9 Å. Based on computer modelling we discovered new interactions between EcoRII and the DNA minor groove and defined the position of the Mg2+ binding pocket. Investigations of the EcoRII MTase by limited proteolysis showed that the enzyme binds DNA depending on DNA sequence and cofactors in different manners. Crystallography experiments with EcoRII MTase in the presence of hemimethylated recognition site DNA showed for the first time diffracting crystals which need further optimisation to create high quality crystals which allow structure solution

    Angiotensin and Endothelin Receptor Structures With Implications for Signaling Regulation and Pharmacological Targeting

    No full text
    In conjunction with the endothelin (ET) type A (ETAR) and type B (ETBR) receptors, angiotensin (AT) type 1 (AT1R) and type 2 (AT2R) receptors, are peptide-binding class A G-protein-coupled receptors (GPCRs) acting in a physiologically overlapping context. Angiotensin receptors (ATRs) are involved in regulating cell proliferation, as well as cardiovascular, renal, neurological, and endothelial functions. They are important therapeutic targets for several diseases or pathological conditions, such as hypertrophy, vascular inflammation, atherosclerosis, angiogenesis, and cancer. Endothelin receptors (ETRs) are expressed primarily in blood vessels, but also in the central nervous system or epithelial cells. They regulate blood pressure and cardiovascular homeostasis. Pathogenic conditions associated with ETR dysfunctions include cancer and pulmonary hypertension. While both receptor groups are activated by their respective peptide agonists, pathogenic autoantibodies (auto-Abs) can also activate the AT1R and ETAR accompanied by respective clinical conditions. To date, the exact mechanisms and differences in binding and receptor-activation mediated by auto-Abs as opposed to endogenous ligands are not well understood. Further, several questions regarding signaling regulation in these receptors remain open. In the last decade, several receptor structures in the apo- and ligand-bound states were determined with protein X-ray crystallography using conventional synchrotrons or X-ray Free-Electron Lasers (XFEL). These inactive and active complexes provide detailed information on ligand binding, signal induction or inhibition, as well as signal transduction, which is fundamental for understanding properties of different activity states. They are also supportive in the development of pharmacological strategies against dysfunctions at the receptors or in the associated signaling axis. Here, we summarize current structural information for the AT1R, AT2R, and ETBR to provide an improved molecular understanding
    corecore