153 results on '"Heinrich Sticht"'
Search Results
2. Assessing clinical utility of preconception expanded carrier screening regarding residual risk for neurodevelopmental disorders
- Author
-
Paranchai Boonsawat, Anselm H. C. Horn, Katharina Steindl, Alessandra Baumer, Pascal Joset, Dennis Kraemer, Angela Bahr, Ivan Ivanovski, Elena M. Cabello, Michael Papik, Markus Zweier, Beatrice Oneda, Pietro Sirleto, Tilo Burkhardt, Heinrich Sticht, and Anita Rauch
- Subjects
Medicine ,Genetics ,QH426-470 - Abstract
Abstract The magnitude of clinical utility of preconception expanded carrier screening (ECS) concerning its potential to reduce the risk of affected offspring is unknown. Since neurodevelopmental disorders (NDDs) in their offspring is a major concern of parents-to-be, we addressed the question of residual risk by assessing the risk-reduction potential for NDDs in a retrospective study investigating ECS with different criteria for gene selection and definition of pathogenicity. We used exome sequencing data from 700 parents of children with NDDs and blindly screened for carrier-alleles in up to 3046 recessive/X-linked genes. Depending on variant pathogenicity thresholds and gene content, NDD-risk-reduction potential was up to 43.5% in consanguineous, and 5.1% in nonconsanguineous couples. The risk-reduction-potential was compromised by underestimation of pathogenicity of missense variants (false-negative-rate 4.6%), inherited copy-number variants and compound heterozygosity of one inherited and one de novo variant (0.9% each). Adherence to the ACMG recommendations of restricting ECS to high-frequency genes in nonconsanguineous couples would more than halve the detectable inherited NDD-risk. Thus, for optimized clinical utility of ECS, screening in recessive/X-linked genes regardless of their frequency (ACMG Tier-4) and sensible pathogenicity thresholds should be considered for all couples seeking ECS.
- Published
- 2022
- Full Text
- View/download PDF
3. Probing the role of intercalating protein sidechains for kink formation in DNA.
- Author
-
Achim Sandmann and Heinrich Sticht
- Subjects
Medicine ,Science - Abstract
Protein binding can induce DNA kinks, which are for example important to enhance the specificity of the interaction and to facilitate the assembly of multi protein complexes. The respective proteins frequently exhibit amino acid sidechains that intercalate between the DNA base steps at the site of the kink. However, on a molecular level there is only little information available about the role of individual sidechains for kink formation. To unravel structural principles of protein-induced DNA kinking we have performed molecular dynamics (MD) simulations of five complexes that varied in their architecture, function, and identity of intercalated residues. Simulations were performed for the DNA complexes of wildtype proteins (Sac7d, Sox-4, CcpA, TFAM, TBP) and for mutants, in which the intercalating residues were individually or combined replaced by alanine. The work revealed that for systems with multiple intercalated residues, not all of them are necessarily required for kink formation. In some complexes (Sox-4, TBP), one of the residues proved to be essential for kink formation, whereas the second residue has only a very small effect on the magnitude of the kink. In other systems (e.g. Sac7d) each of the intercalated residues proved to be individually capable of conferring a strong kink suggesting a partially redundant role of the intercalating residues. Mutation of the key residues responsible for kinking either resulted in stable complexes with reduced kink angles or caused conformational instability as evidenced by a shift of the kink to an adjacent base step. Thus, MD simulations can help to identify the role of individual inserted residues for kinking, which is not readily apparent from an inspection of the static structures. This information might be helpful for understanding protein-DNA interactions in more detail and for designing proteins with altered DNA binding properties in the future.
- Published
- 2018
- Full Text
- View/download PDF
4. Probing the potential of CnaB-type domains for the design of tag/catcher systems.
- Author
-
Marlene Pröschel, Max E Kraner, Anselm H C Horn, Lena Schäfer, Uwe Sonnewald, and Heinrich Sticht
- Subjects
Medicine ,Science - Abstract
Building proteins into larger, post-translational assemblies in a defined and stable way is still a challenging task. A promising approach relies on so-called tag/catcher systems that are fused to the proteins of interest and allow a durable linkage via covalent intermolecular bonds. Tags and catchers are generated by splitting protein domains that contain intramolecular isopeptide or ester bonds that form autocatalytically under physiological conditions. There are already numerous biotechnological and medical applications that demonstrate the usefulness of covalent linkages mediated by these systems. Additional covalent tag/catcher systems would allow creating more complex and ultra-stable protein architectures and networks. Two of the presently available tag/catcher systems were derived from closely related CnaB-domains of Streptococcus pyogenes and Streptococcus dysgalactiae proteins. However, it is unclear whether domain splitting is generally tolerated within the CnaB-family or only by a small subset of these domains. To address this point, we have selected a set of four CnaB domains of low sequence similarity and characterized the resulting tag/catcher systems by computational and experimental methods. Experimental testing for intermolecular isopeptide bond formation demonstrated two of the four systems to be functional. For these two systems length and sequence variations of the peptide tags were investigated revealing only a relatively small effect on the efficiency of the reaction. Our study suggests that splitting into tag and catcher moieties is tolerated by a significant portion of the naturally occurring CnaB-domains, thus providing a large reservoir for the design of novel tag/catcher systems.
- Published
- 2017
- Full Text
- View/download PDF
5. Role of the N-terminus for the stability of an amyloid-β fibril with three-fold symmetry.
- Author
-
Christian A Söldner, Heinrich Sticht, and Anselm H C Horn
- Subjects
Medicine ,Science - Abstract
A key player in Alzheimer's disease is the peptide amyloid-beta (Aβ), whose aggregation into small soluble oligomers, protofilaments, and fibrils finally leads to plaque deposits in human brains. The aggregation behavior of Aβ is strongly modulated by the nature and composition of the peptide's environment and by its primary sequence properties. The N-terminal residues of Aβ play an important role, because they are known to change the peptide's aggregation propensity. Since these residues are for the first time completely resolved at the molecular level in a three-fold symmetric fibril structure derived from a patient, we chose that system as template for a systematic investigation of the influence of the N-terminus upon structural stability. Using atomistic molecular dynamics simulations, we examined several fibrillar systems comprising three, six, twelve and an infinite number of layers, both with and without the first eight residues. First, we found that three layers are not sufficient to stabilize the respective Aβ topology. Second, we observed a clear stabilizing effect of the N-terminal residues upon the overall fibril fold: truncated Aβ systems were less stable than their full-length counterparts. The N-terminal residues Arg5, Asp7, and Ser8 were found to form important interfilament contacts stabilizing the overall fibril structure of three-fold symmetry. Finally, similar structural rearrangements of the truncated Aβ species in different simulations prompted us to suggest a potential mechanism involved in the formation of amyloid fibrils with three-fold symmetry.
- Published
- 2017
- Full Text
- View/download PDF
6. Selection and Characterization of Tau Binding ᴅ-Enantiomeric Peptides with Potential for Therapy of Alzheimer Disease.
- Author
-
Christina Dammers, Deniz Yolcu, Laura Kukuk, Dieter Willbold, Marcus Pickhardt, Eckhard Mandelkow, Anselm H C Horn, Heinrich Sticht, Marwa Nidal Malhis, Nadja Will, Judith Schuster, and Susanne Aileen Funke
- Subjects
Medicine ,Science - Abstract
A variety of neurodegenerative disorders, including Alzheimer disease (AD), are associated with neurofibrillary tangles composed of the tau protein, as well as toxic tau oligomers. Inhibitors of pathological tau aggregation, interrupting tau self-assembly, might be useful for the development of therapeutics. Employing mirror image phage display with a large peptide library (over 109 different peptides), we have identified tau fibril binding peptides consisting of d-enantiomeric amino acids. d-enantiomeric peptides are extremely protease stable and not or less immunogenic than l-peptides, and the suitability of d-peptides for in vivo applications have already been demonstrated. Phage display selections were performed using fibrils of the d-enantiomeric hexapeptide VQIVYK, representing residues 306 to 311 of the tau protein, as a target. VQIVYK has been demonstrated to be important for fibril formation of the full lengths protein and forms fibrils by itself. Here, we report on d-enantiomeric peptides, which bind to VQIVYK, tau isoforms like tau3RD (K19) as well as to full lengths tau fibrils, and modulate the aggregation of the respective tau form. The peptides are able to penetrate cells and might be interesting for therapeutic and diagnostic applications in AD research.
- Published
- 2016
- Full Text
- View/download PDF
7. Alanine-glyoxylate aminotransferase 2 (AGXT2) polymorphisms have considerable impact on methylarginine and β-aminoisobutyrate metabolism in healthy volunteers.
- Author
-
Anja Kittel, Fabian Müller, Jörg König, Maren Mieth, Heinrich Sticht, Oliver Zolk, Ana Kralj, Markus R Heinrich, Martin F Fromm, and Renke Maas
- Subjects
Medicine ,Science - Abstract
Elevated plasma concentrations of asymmetric (ADMA) and symmetric (SDMA) dimethylarginine have repeatedly been linked to adverse clinical outcomes. Both methylarginines are substrates of alanine-glyoxylate aminotransferase 2 (AGXT2). It was the aim of the present study to simultaneously investigate the functional relevance and relative contributions of common AGXT2 single nucleotide polymorphisms (SNPs) to plasma and urinary concentrations of methylarginines as well as β-aminoisobutyrate (BAIB), a prototypic substrate of AGXT2. In a cohort of 400 healthy volunteers ADMA, SDMA and BAIB concentrations were determined in plasma and urine using HPLC-MS/MS and were related to the coding AGXT2 SNPs rs37369 (p.Val140Ile) and rs16899974 (p.Val498Leu). Volunteers heterozygous or homozygous for the AGXT2 SNP rs37369 had higher SDMA plasma concentrations by 5% and 20% (p = 0.002) as well as higher BAIB concentrations by 54% and 146%, respectively, in plasma and 237% and 1661%, respectively, in urine (both p
- Published
- 2014
- Full Text
- View/download PDF
8. Correction: HIV-1 Fusion Is Blocked through Binding of GB Virus C E2D Peptides to the HIV-1 gp41 Disulfide Loop.
- Author
-
Kristin Eissmann, Sebastian Mueller, Heinrich Sticht, Susan Jung, Peng Zou, Shibo Jiang, Andrea Gross, Jutta Eichler, Bernhard Fleckenstein, and Heide Reil
- Subjects
Medicine ,Science - Published
- 2014
- Full Text
- View/download PDF
9. Systematic analysis of phosphotyrosine antibodies recognizing single phosphorylated EPIYA-motifs in CagA of Western-type Helicobacter pylori strains.
- Author
-
Judith Lind, Steffen Backert, Klaus Pfleiderer, Douglas E Berg, Yoshio Yamaoka, Heinrich Sticht, and Nicole Tegtmeyer
- Subjects
Medicine ,Science - Abstract
The clinical outcome of Helicobacter pylori infections is determined by multiple host-pathogen interactions that may develop to chronic gastritis, and sometimes peptic ulcers or gastric cancer. Highly virulent strains encode a type IV secretion system (T4SS) that delivers the effector protein CagA into gastric epithelial cells. Translocated CagA undergoes tyrosine phosphorylation at EPIYA-sequence motifs, called A, B and C in Western-type strains, by members of the oncogenic Src and Abl host kinases. Phosphorylated EPIYA-motifs mediate interactions of CagA with host signaling factors--in particular various SH2-domain containing human proteins--thereby hijacking multiple downstream signaling cascades. Observations of tyrosine-phosphorylated CagA are mainly based on the use of commercial phosphotyrosine antibodies, which originally were selected to detect phosphotyrosines in mammalian proteins. Systematic studies of phosphorylated EPIYA-motif detection by the different antibodies would be very useful, but are not yet available. To address this issue, we synthesized phospho- and non-phosphopeptides representing each predominant Western CagA EPIYA-motif, and determined the recognition patterns of seven different phosphotyrosine antibodies in Western blots, and also performed infection studies with diverse representative Western H. pylori strains. Our results show that a total of 9-11 amino acids containing the phosphorylated EPIYA-motifs are necessary and sufficient for specific detection by these antibodies, but revealed great variability in sequence recognition. Three of the antibodies recognized phosphorylated EPIYA-motifs A, B and C similarly well; whereas preferential binding to phosphorylated motif A and motifs A and C was found with two and one antibodies, respectively, and the seventh anti-phosphotyrosine antibody did not recognize any phosphorylated EPIYA-motif. Controls showed that none of the antibodies recognized the corresponding non-phospho CagA peptides, and that all of them recognized phosphotyrosines in mammalian proteins. These data are valuable in judicious application of commercial anti-phosphotyrosine antibodies and in characterization of CagA phosphorylation during infection and disease development.
- Published
- 2014
- Full Text
- View/download PDF
10. Characterization of a single-chain variable fragment recognizing a linear epitope of aβ: a biotechnical tool for studies on Alzheimer's disease?
- Author
-
Silke Dornieden, Andreas Müller-Schiffmann, Heinrich Sticht, Nan Jiang, Yeliz Cinar, Michael Wördehoff, Carsten Korth, Susanne Aileen Funke, and Dieter Willbold
- Subjects
Medicine ,Science - Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with devastating effects. Currently, therapeutic options are limited to symptomatic treatment. For more than a decade, research focused on immunotherapy for the causal treatment of AD. However, clinical trials with active immunization using Aβ encountered severe complications, for example meningoencephalitis. Consequently, attention focused on passive immunization using antibodies. As an alternative to large immunoglobulins (IgGs), Aβ binding single-chain variable fragments (scFvs) were used for diagnostic and therapeutic research approaches. scFvs can be expressed in E. coli and may provide improved pharmacokinetic properties like increased blood-brain barrier permeability or reduced side-effects in vivo. In this study, we constructed an scFv from an Aβ binding IgG, designated IC16, which binds the N-terminal region of Aβ (Aβ(1-8)). scFv-IC16 was expressed in E. coli, purified and characterized with respect to its interaction with different Aβ species and its influence on Aβ fibril formation. We were able to show that scFv-IC16 strongly influenced the aggregation behavior of Aβ and could be applied as an Aβ detection probe for plaque staining in the brains of transgenic AD model mice. The results indicate potential for therapy and diagnosis of AD.
- Published
- 2013
- Full Text
- View/download PDF
11. HIV-1 fusion is blocked through binding of GB Virus C E2-derived peptides to the HIV-1 gp41 disulfide loop [corrected].
- Author
-
Kristin Eissmann, Sebastian Mueller, Heinrich Sticht, Susan Jung, Peng Zou, Shibo Jiang, Andrea Gross, Jutta Eichler, Bernhard Fleckenstein, and Heide Reil
- Subjects
Medicine ,Science - Abstract
A strategy for antiviral drug discovery is the elucidation and imitation of viral interference mechanisms. HIV-1 patients benefit from a coinfection with GB Virus C (GBV-C), since HIV-positive individuals with long-term GBV-C viraemia show better survival rates than HIV-1 patients without persisting GBV-C. A direct influence of GBV-C on HIV-1 replication has been shown in coinfection experiments. GBV-C is a human non-pathogenic member of the flaviviridae family that can replicate in T and B cells. Therefore, GBV-C shares partly the same ecological niche with HIV-1. In earlier work we have demonstrated that recombinant glycoprotein E2 of GBV-C and peptides derived from the E2 N-terminus interfere with HIV entry. In this study we investigated the underlying mechanism. Performing a virus-cell fusion assay and temperature-arrested HIV-infection kinetics, we provide evidence that the HIV-inhibitory E2 peptides interfere with late HIV-1 entry steps after the engagement of gp120 with CD4 receptor and coreceptor. Binding and competition experiments revealed that the N-terminal E2 peptides bind to the disulfide loop region of HIV-1 transmembrane protein gp41. In conjunction with computational analyses, we identified sequence similarities between the N-termini of GBV-C E2 and the HIV-1 glycoprotein gp120. This similarity appears to enable the GBV-C E2 N-terminus to interact with the HIV-1 gp41 disulfide loop, a crucial domain involved in the gp120-gp41 interface. Furthermore, the results of the present study provide initial proof of concept that peptides targeted to the gp41 disulfide loop are able to inhibit HIV fusion and should inspire the development of this new class of HIV-1 entry inhibitors.
- Published
- 2013
- Full Text
- View/download PDF
12. Conformational stability of fibrillar amyloid-beta oligomers via protofilament pair formation - a systematic computational study.
- Author
-
Anna Kahler, Heinrich Sticht, and Anselm H C Horn
- Subjects
Medicine ,Science - Abstract
Amyloid-[Formula: see text] (A[Formula: see text]) oligomers play a crucial role in Alzheimer's disease due to their neurotoxic aggregation properties. Fibrillar A[Formula: see text] oligomerization can lead to protofilaments and protofilament pairs via oligomer elongation and oligomer association, respectively. Small fibrillar oligomers adopt the protofilament topology, whereas fibrils contain at least protofilament pairs. To date, the underlying growth mechanism from oligomers to the mature fibril still remains to be elucidated. Here, we performed all-atom molecular dynamics simulations in explicit solvent on single layer-like protofilaments and fibril-like protofilament pairs of different size ranging from the tetramer to the 48-mer. We found that the initial U-shaped topology per monomer is maintained over time in all oligomers. The observed deviations of protofilaments from the starting structure increase significantly with size due to the twisting of the in-register parallel [Formula: see text]-sheets. This twist causes long protofilaments to be unstable and leads to a breakage. Protofilament pairs, which are stabilized by a hydrophobic interface, exhibit more fibril-like properties such as the overall structure and the twist angle. Thus, they can act as stable conformational templates for further fibril growth. Key properties like the twist angle, shape complementarity, and energetics show a size-dependent behavior so that small oligomers favor the protofilament topology, whereas large oligomers favor the protofilament pair topology. The region for this conformational transition is at the size of approximately twelve A[Formula: see text] monomers. From that, we propose the following growth mechanism from A[Formula: see text] oligomers to fibrils: (1) elongation of short protofilaments; (2) breakage of large protofilaments; (3) formation of short protofilament pairs; and (4) elongation of protofilament pairs.
- Published
- 2013
- Full Text
- View/download PDF
13. A molecular model for the differential activation of STAT3 and STAT6 by the herpesviral oncoprotein tip.
- Author
-
Eman Dey Mazumder, Christophe Jardin, Benjamin Vogel, Elke Heck, Brigitte Scholz, Doris Lengenfelder, Heinrich Sticht, and Armin Ensser
- Subjects
Medicine ,Science - Abstract
Constitutive STAT signaling provides growth promoting signals in many forms of malignancy. We performed molecular modeling and molecular dynamics studies of the interaction between the regulatory Src homology 2 (SH2) domains of STAT3 and 6 with phosphorylated peptides of the herpesviral oncoprotein Tip, which facilitates Src kinase mediated STAT-activation and T cell proliferation. The studies give insight into the ligand binding specificity of the STAT SH2 domains and provide the first model for the differential activation of STAT3 or STAT6 by two distinct regions of the viral Tip protein. The biological relevance of the modeled interactions was then confirmed by activation studies using corresponding recombinant oncoproteins, and finally by respective recombinant viruses. The functional data give experimental validation of the molecular dynamics study, and provide evidence for the involvement of STAT6 in the herpesvirus induced T cell proliferation.
- Published
- 2012
- Full Text
- View/download PDF
14. Episodic psychosis, ataxia, motor neuropathy with pyramidal signs (PAMP syndrome) caused by a novel mutation in ADPRHL2 (AHR3)
- Author
-
Hacer Durmus, Said Hashemolhosseini, Isin Baral Kulaksizoglu, Yesim Parman, Evren Önay Uçar, Serdar Ceylaner, Heinrich Sticht, and Elif Mertoğlu
- Subjects
Adult ,Pathology ,medicine.medical_specialty ,Psychosis ,Pes cavus ,Weakness ,Ataxia ,Neurology ,Cerebellar Ataxia ,Glycoside Hydrolases ,Dermatology ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,030212 general & internal medicine ,Child ,business.industry ,Neurodegeneration ,General Medicine ,Postural tremor ,medicine.disease ,Psychiatry and Mental health ,medicine.anatomical_structure ,Psychotic Disorders ,Mutation ,Female ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Sensory nerve - Abstract
The protein “ADP-Ribosylarginine Hydrolase-Like Protein 2” is encoded by ADPRHL2 and reverses ADP-ribosylation. Recently, mutations in ADPRHL2 were found to be associated with a very rare childhood onset severe neurodegeneration syndrome with episodic, stress-induced seizures, ataxia, and axonal neuropathy. In this study, we evaluate a novel mutation in ADPRHL2 leading to an unknown adult onset syndrome “episodic psychosis, ataxia, motor neuropathy with pyramidal signs (PAMP syndrome).” Four patients with episodic psychosis, ataxia, and motor neuropathy with pyramidal signs were included in this study. An index patient presented ataxia, postural tremor in the hands, and hallucinations at age 20 years, which had started after a viral infection. She improved within 3 months without any treatment. Her neurological exam revealed mild distal weakness, brisk DTRs, bilateral Babinski sign, impaired vibration sensation, position, and ataxia. Pes cavus and hammer toes were also noted. EMG revealed neurogenic changes in distal muscles and normal sensory nerve conduction studies. Cranial MRI was normal. She had three more severe episodes in recent years, and her neurologic findings got progressively worse. Two of her older sisters had much milder phenotypes. The phenotype of the fourth patient from an unrelated family was identical with the index patient. All affected patients had homozygous novel NM_017825.3:c.838G>A (p.Ala280Thr) mutations in a highly conserved region of ADPRHL2. Western blot analyses demonstrated that ADPRHL2 was not expressed in these patients. Here, we describe a novel mutation in ADPRHL2, which further expands the phenotypic and genetic spectrum of the patients harboring these mutations.
- Published
- 2021
15. Matricellular Protein SPARCL1 Regulates Blood Vessel Integrity and Antagonizes Inflammatory Bowel Disease
- Author
-
Carol Geppert, Rocío López-Posadas, Andreas Ramming, Clara Tenkerian, Heinrich Sticht, Elisabeth Naschberger, Victoria Pürzer, Claudia Günther, Katja Petter, Tim Thoenissen, Christoph Becker, Benjamin Schmid, Thomas Wohlfahrt, Tobias Gass, Valerie Meniel, Christian Flierl, Maximilian J. Waldner, Nathalie Britzen-Laurent, Iris Stolzer, Michael Stürzl, and Daniela Regensburger
- Subjects
0301 basic medicine ,DSS colitis ,Angiogenesis ,SPARCL1 ,Inflammation ,Inflammatory bowel disease ,03 medical and health sciences ,angiogenesis ,Mice ,0302 clinical medicine ,blood vessel ,inflammatory bowel disease ,Immunology and Allergy ,Medicine ,Animals ,Colitis ,Ibdjnl/4 ,AcademicSubjects/MED00260 ,Sprouting angiogenesis ,Mice, Knockout ,Extracellular Matrix Proteins ,Neovascularization, Pathologic ,business.industry ,Matricellular protein ,Calcium-Binding Proteins ,Dextran Sulfate ,Gastroenterology ,medicine.disease ,Cell biology ,Endothelial stem cell ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,cardiovascular system ,endothelial cell ,medicine.symptom ,permeability ,business ,Basic Science Research ,Blood vessel - Abstract
Background The understanding of vascular plasticity is key to defining the role of blood vessels in physiologic and pathogenic processes. In the present study, the impact of the vascular quiescence marker SPARCL1 on angiogenesis, capillary morphogenesis, and vessel integrity was evaluated. Methods Angiogenesis was studied using the metatarsal test, an ex vivo model of sprouting angiogenesis. In addition, acute and chronic dextran sodium sulfate colitis models with SPARCL1 knockout mice were applied. Results This approach indicated that SPARCL1 inhibits angiogenesis and supports vessel morphogenesis and integrity. Evidence was provided that SPARCL1-mediated stabilization of vessel integrity counteracts vessel permeability and inflammation in acute and chronic dextran sodium sulfate colitis models. Structure-function analyses of purified SPARCL1 identified the acidic domain of the protein necessary for its anti-angiogenic activity. Conclusions Our findings inaugurate SPARCL1 as a blood vessel–derived anti-angiogenic molecule required for vessel morphogenesis and integrity. SPARCL1 opens new perspectives as a vascular marker of susceptibility to colitis and as a therapeutic molecule to support blood vessel stability in this disease.
- Published
- 2021
16. A pair of noncompeting neutralizing human monoclonal antibodies protecting from disease in a SARS-CoV-2 infection model
- Author
-
Markus Hoffmann, Sandra Ciesek, Dirk Mielenz, Leila Issmail, Sebastian R. Schulz, Eileen Socher, Eva Grüner, Katharina Habenicht, Roman Wölfel, Paul F. McKay, Hans-Martin Jäck, Tobit Steinmetz, Robin J. Shattock, Ralf Wagner, Valentina Eberlein, Edith Roth, Heinrich Sticht, Jutta Eichler, Stefan Pöhlmann, Matthias Tenbusch, Frank Neipel, Wolfgang Schuh, Elie Richel, Kirsten Fraedrich, Thomas Winkler, Thomas Grunwald, Sandra Mueller-Schmucker, Antonia Sophia Peter, Simon Dolles, Klaus Überla, Nadja Uhlig, Armin Ensser, David Peterhoff, Dominik Damm, Manuela Hauke, and Publica
- Subjects
medicine.drug_class ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Transgene ,Immunology ,Mutant ,Immunity to infection ,Disease ,Antibodies, Viral ,spike protein ,variants of concern ,Monoclonal antibody ,SARS‐CoV‐2 ,DNA sequencing ,Mice ,03 medical and health sciences ,0302 clinical medicine ,COVID‐19 ,ddc:570 ,medicine ,Animals ,Humans ,Immunology and Allergy ,neutralizing antibodies ,ddc:610 ,Basic ,Gene ,Research Articles ,030304 developmental biology ,0303 health sciences ,biology ,SARS-CoV-2 ,Antibodies, Monoclonal ,COVID-19 ,Antibodies, Neutralizing ,Virology ,3. Good health ,030220 oncology & carcinogenesis ,Spike Glycoprotein, Coronavirus ,biology.protein ,Research Article|Basic ,Antibody - Abstract
TRIANNI mice carry an entire set of human immunoglobulin V region gene segments and are a powerful tool to rapidly isolate human monoclonal antibodies. After immunizing these mice with DNA encoding the spike protein of SARS‐CoV‐2 and boosting with spike protein, we identified 29 hybridoma antibodies that reacted with the SARS‐CoV‐2 spike protein. Nine antibodies neutralize SARS‐CoV‐2 infection at IC50 values in the subnanomolar range. ELISA‐binding studies and DNA sequence analyses revealed one cluster of three clonally related neutralizing antibodies that target the receptor‐binding domain and compete with the cellular receptor hACE2. A second cluster of six clonally related neutralizing antibodies bind to the N‐terminal domain of the spike protein without competing with the binding of hACE2 or cluster 1 antibodies. SARS‐CoV‐2 mutants selected for resistance to an antibody from one cluster are still neutralized by an antibody from the other cluster. Antibodies from both clusters markedly reduced viral spread in mice transgenic for human ACE2 and protected the animals from SARS‐CoV‐2‐induced weight loss. The two clusters of potent non‐competing SARS‐CoV‐2 neutralizing antibodies provide potential candidates for therapy and prophylaxis of COVID‐19. The study further supports transgenic animals with a human immunoglobulin gene repertoire as a powerful platform in pandemic preparedness initiatives. This article is protected by copyright. All rights reserved
- Published
- 2022
17. T4SS-dependent TLR5 activation by Helicobacter pylori infection
- Author
-
Christina Falkeis-Veits, Michael Vieth, Steffen Backert, Anne Müller, Matthias Neddermann, Sujay Chattopadhyay, Nicole Tegtmeyer, Mark Brönstrup, Isabelle C. Arnold, Suneesh Kumar Pachathundikandi, Minsun Hong, Judith Lind, Heinrich Sticht, Werner Tegge, University of Zurich, Backert, Steffen, and HZI,Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstr. 7,38124 Braunschweig, Germany.
- Subjects
0301 basic medicine ,Biopsy ,General Physics and Astronomy ,medicine.disease_cause ,Mice ,0302 clinical medicine ,lcsh:Science ,Receptor ,Mice, Knockout ,Multidisciplinary ,10061 Institute of Molecular Cancer Research ,NF-kappa B ,3100 General Physics and Astronomy ,3. Good health ,030220 oncology & carcinogenesis ,Host-Pathogen Interactions ,Female ,Pathogens ,Signal Transduction ,Science ,Virulence ,610 Medicine & health ,1600 General Chemistry ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Microbiology ,Helicobacter Infections ,Type IV Secretion Systems ,03 medical and health sciences ,Immune system ,Bacterial Proteins ,1300 General Biochemistry, Genetics and Molecular Biology ,medicine ,Animals ,Humans ,Secretion ,Helicobacter pylori ,Activator (genetics) ,Pathogenic bacteria ,General Chemistry ,biology.organism_classification ,Toll-like receptors ,Disease Models, Animal ,Toll-Like Receptor 5 ,030104 developmental biology ,TLR5 ,Gastric Mucosa ,570 Life sciences ,biology ,lcsh:Q - Abstract
Toll-like receptor TLR5 recognizes a conserved domain, termed D1, that is present in flagellins of several pathogenic bacteria but not in Helicobacter pylori. Highly virulent H. pylori strains possess a type IV secretion system (T4SS) for delivery of virulence factors into gastric epithelial cells. Here, we show that one of the H. pylori T4SS components, protein CagL, can act as a flagellin-independent TLR5 activator. CagL contains a D1-like motif that mediates adherence to TLR5+ epithelial cells, TLR5 activation, and downstream signaling in vitro. TLR5 expression is associated with H. pylori infection and gastric lesions in human biopsies. Using Tlr5-knockout and wild-type mice, we show that TLR5 is important for efficient control of H. pylori infection. Our results indicate that CagL, by activating TLR5, may modulate immune responses to H. pylori., Toll-like receptor TLR5 recognizes a domain, D1, that is present in flagellins of several pathogenic bacteria but not in Helicobacter pylori. Here, the authors show that TLR5 can be activated independently of flagellin by a component of the H. pylori type IV secretion system that contains a D1-like motif.
- Published
- 2019
18. Computational decomposition reveals reshaping of the SARS‐CoV‐2–ACE2 interface among viral variants expressing the N501Y mutation
- Author
-
Marcus Conrad, Friedrich Paulsen, Lukas Heger, Eileen Socher, Philipp Arnold, Friederike Zunke, and Heinrich Sticht
- Subjects
Infectivity ,chemistry.chemical_classification ,Mutation ,Binding Sites ,Chemistry ,SARS-CoV-2 ,In silico ,Mutation, Missense ,Cell Biology ,Computational biology ,Molecular Dynamics Simulation ,medicine.disease_cause ,Biochemistry ,Virus ,Amino acid ,Amino Acid Substitution ,Spike Glycoprotein, Coronavirus ,medicine ,Humans ,Angiotensin-Converting Enzyme 2 ,ddc:610 ,Threonine ,Tyrosine ,Molecular Biology ,Coronavirus - Abstract
Variants of concern of the SARS‐CoV‐2 virus with an asparagine‐to‐tyrosine substitution at position 501 (N501Y) in the receptor‐binding domain (RBD) show enhanced infectivity compared to wild‐type, resulting in an altered pandemic situation in affected areas. These SARS‐Cov‐2 variants comprise the two Alpha variants (B.1.1.7, United Kingdom and B.1.1.7 with the additional E484K mutation), the Beta variant (B.1.351, South Africa), and the Gamma variant (P.1, Brazil). Understanding the binding modalities between these viral variants and the host cell receptor ACE2 allows to depict changes, but also common motifs of virus–host cell interaction. The trimeric spike protein expressed at the viral surface contains the RBD that forms the molecular interface with ACE2. All the above‐mentioned variants carry between one and three amino acid exchanges within the interface‐forming region of the RBD, thereby altering the binding interface with ACE2. Using molecular dynamics (MD) simulations and decomposition of intermolecular contacts between the RBD and ACE2, we identified phenylalanine 486, glutamine 498, threonine 500, and tyrosine 505 as important interface‐forming residues across viral variants. However, especially the N501Y exchange increased contact formation for this residue and also induced some local conformational changes. Comparing here, the in silico generated B.1.1.7 RBD–ACE2 complex with the now available experimentally solved structure reveals very similar behavior during MD simulation. We demonstrate, how computational methods can help to identify differences in conformation as well as contact formation for newly emerging viral variants. Altogether, we provide extensive data on all N501Y expressing SARS‐CoV‐2 variants of concern with respect to their interaction with ACE2 and how this induces reshaping of the RBD–ACE2 interface.
- Published
- 2021
19. Bi-allelic variants in CHKA cause a neurodevelopmental disorder with epilepsy and microcephaly
- Author
-
Reza Maroofian, Henry Houlden, Jess Williams, Maha S. Zaki, Andreas Merkenschlager, M. M. Noureldeen, Konrad Platzer, H. Darvish, Christopher R. McMaster, C. Kloeckner, Saghar Ghasemi Firouzabadi, Michael C. Kruer, Somayeh Bakhtiari, S. H. Banu, M. Tavasoli, J. P. Fernandez Murray, Heinrich Sticht, Anju Shukla, Parneet Kaur, Dhanya Lakshmi Narayanan, A. Pagnozzi, E. Cali, R. Abou Jamra, L. Scholle, Janina Gburek-Augustat, K. Nahar, Katta M. Girisha, Stephanie Efthymiou, and G. Stoltenburg-Didinger
- Subjects
Genetics ,Epilepsy ,Microcephaly ,Neurodevelopmental disorder ,Movement disorders ,medicine ,Dystrophy ,Global developmental delay ,Biology ,Muscular dystrophy ,medicine.symptom ,Allele ,medicine.disease - Abstract
The Kennedy pathways catalyze the de novo synthesis of phosphatidylcholine and phosphatidylethanolamine, the most abundant components of eukaryotic cell membranes. In recent years, these pathways have moved into clinical focus since four out of ten genes involved have been associated with a range of autosomal recessive rare diseases such as a neurodevelopmental disorder with muscular dystrophy (CHKB), bone abnormalities and cone-rod dystrophy (PCYT1A), and spastic paraplegia (PCYT2, SELENOI).We identified six individuals from five families with bi-allelic variants in CHKA presenting with severe global developmental delay, epilepsy, movement disorders, and microcephaly. Using structural molecular modeling and functional testing of the variants in a in a cell-based S. cerevisiae model, we determined that these variants reduce the enzymatic activity of CHKA and confer a significant impairment of the first enzymatic step of the Kennedy pathway.In summary, we present CHKA as a novel autosomal recessive gene for a neurodevelopmental disorder with epilepsy and microcephaly.
- Published
- 2021
20. A Novel Strain-Specific Neutralizing Epitope on Glycoprotein H of Human Cytomegalovirus
- Author
-
Nina Reuter, Barbara Kropff, Andrea Schneider, Heinrich Sticht, Thomas Winkler, Irene Görzer, Michael Mach, Marco Thomas, and William J. Britt
- Subjects
Human cytomegalovirus ,medicine.drug_class ,Immunology ,Cytomegalovirus ,Monoclonal antibody ,Antibodies, Viral ,Microbiology ,Epitope ,Epitopes ,Mice ,Viral Envelope Proteins ,Virology ,Vaccines and Antiviral Agents ,medicine ,Animals ,Humans ,Neutralizing antibody ,Mice, Inbred BALB C ,biology ,Antibodies, Monoclonal ,Antiviral antibody ,medicine.disease ,Antibodies, Neutralizing ,Transplantation ,HEK293 Cells ,Insect Science ,Cytomegalovirus Infections ,biology.protein ,Antibody ,Conformational epitope - Abstract
Human cytomegalovirus (HCMV) is a ubiquitous pathogen that causes severe clinical disease in immunosuppressed patients and congenitally infected newborn infants. Viral envelope glycoproteins represent attractive targets for vaccination or passive immunotherapy. To extend the knowledge of mechanisms of virus neutralization, monoclonal antibodies (MAbs) were generated following immunization of mice with HCMV virions. Hybridoma supernatants were screened for in vitro neutralization activity, yielding three potent MAbs, 6E3, 3C11, and 2B10. MAbs 6E3 and 3C11 blocked infection of all viral strains that were tested, while MAb 2B10 neutralized only 50% of the HCMV strains analyzed. Characterization of the MAbs using indirect immunofluorescence analyses demonstrated their reactivity with recombinantly derived gH. While MAbs 6E3 and 3C11 reacted with gH when expressed alone, 2B10 detected gH only when it was coexpressed with gB and gL. Recognition of gH by 3C11 was dependent on the expression of the entire ectodomain of gH, whereas 6E3 required residues 1 to 629 of gH. The strain-specific determinant for neutralization by Mab 2B10 was identified as a single Met→Ile amino acid polymorphism within gH, located within the central part of the protein. The polymorphism is evenly distributed among described HCMV strains. The 2B10 epitope thus represents a novel strain-specific antibody target site on gH of HCMV. The dependence of the reactivity of 2B10 on the simultaneous presence of gB/gH/gL will be of value in the structural definition of this tripartite complex. The 2B10 epitope may also represent a valuable tool for diagnostics to monitor infections/reinfections with different HCMV strains during pregnancy or after transplantation. IMPORTANCE HCMV infections are life threatening to people with compromised or immature immune systems. Understanding the antiviral antibody repertoire induced during HCMV infection is a necessary prerequisite to define protective antibody responses. Here, we report three novel anti-gH MAbs that potently neutralized HCMV infectivity. One of these MAbs (2B10) targets a novel strain-specific conformational epitope on gH that only becomes accessible upon coexpression of the minimal fusion machinery gB/gH/gL. Strain specificity is dependent on a single amino acid polymorphism within gH. Our data highlight the importance of strain-specific neutralizing antibody responses against HCMV. The 2B10 epitope may also represent a valuable tool for diagnostics to monitor infections/reinfections with different HCMV strains during pregnancy or after transplantation. In addition, the dependence of the reactivity of 2B10 on the simultaneous presence of gB/gH/gL will be of value in the structural definition of this tripartite complex.
- Published
- 2021
21. The crystal structure of the varicella zoster Orf24-Orf27 nuclear egress complex spotlights multiple determinants of herpesvirus subfamily specificity
- Author
-
Claudia Egerer-Sieber, Julia Tillmanns, T. Lenac Rovis, Sewar Alkhashrom, Heinrich Sticht, Marcus Conrad, Manfred Marschall, Yves A. Muller, Sigrid Weiler, Johannes Schweininger, A. Decker, Mark Kriegel, Sigrun Häge, Josephine Lösing, and Jutta Eichler
- Subjects
Human cytomegalovirus ,Subfamily ,Chemistry ,viruses ,Varicella zoster virus ,virus diseases ,Computational biology ,Alanine scanning ,medicine.disease ,medicine.disease_cause ,Virus ,medicine.anatomical_structure ,Capsid ,medicine ,Nucleus ,Binding affinities - Abstract
Varicella zoster virus (VZV) is a human pathogen from the α-subfamily of herpesviruses. Here, the crystal structure of the VZV Orf24-Orf27 complex is described, representing the essential viral core nuclear egress complex (NEC) that orchestrates the egress of the preassembled capsids from the nucleus. While previous studies have primarily emphasized the finding that the architecture of core NEC complexes is highly conserved among herpesviruses, the present report focusses on subfamily-specific structural and functional features that help explain the differences in the autologous versus nonautologous interaction patterns observed for NEC formation across herpesviruses. CoIP and confocal imaging data show that Orf24-Orf27 complex formation displays some promiscuity in a herpesvirus subfamily-restricted manner. At the same time, analysis of the NEC formation thermodynamic parameters of three prototypical α-, β- and γ herpesviruses, i.e. VZV, human cytomegalovirus (HCMV) and Epstein-Barr virus (EBV) reveals highly similar binding affinities for the autologous interaction with some specific differences in the enthalpy and entropy terms. Computational alanine scanning and structural comparisons highlight intermolecular interactions shared among α-herpesviruses that are clearly distinct from those seen in β- and γ-herpesviruses. Combined, these data allow to explain the distinct properties of specificity and permissivity so far observed in herpesviral NEC interactions. These findings might prove highly valuable when attempting to target multiple herpesvirus core NECs with selective or broad-acting drug candidates.
- Published
- 2021
22. Inhibition of SARS CoV Envelope Protein by Flavonoids and Classical Viroporin Inhibitors
- Author
-
Hans-Georg Breitinger, Heinrich Sticht, Nourhan K M Ali, and Ulrike Breitinger
- Subjects
Microbiology (medical) ,Signal peptide ,cell viability assay ,Rimantadine ,viroporins ,ion channel inhibitors ,SARS CoV-1 ,Microbiology ,Virus ,03 medical and health sciences ,medicine ,ddc:610 ,Viability assay ,Original Research ,030304 developmental biology ,E protein ,0303 health sciences ,patch-clamp electrophysiology ,Chemistry ,030302 biochemistry & molecular biology ,Amantadine ,QR1-502 ,Transmembrane protein ,Virus Release ,Cell biology ,Intracellular ,medicine.drug - Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV), an enveloped single-stranded positive-sense RNA virus, is a member of the genus Betacoronavirus, family Coronaviridae. The SARS-CoV envelope protein E is a small (∼8.4 kDa) channel-forming membrane protein whose sequence is highly conserved between SARS-CoV and SARS-CoV-2. As a viroporin, it is involved in various aspects of the virus life cycle including assembly, budding, envelope formation, virus release, and inflammasome activation. Here, SARS-CoV E protein was recombinantly expressed in HEK293 cells and channel activity and the effects of viroporin inhibitors studied using patch-clamp electrophysiology and a cell viability assay. We introduced a membrane-directing signal peptide to ensure transfer of recombinant E protein to the plasma membrane. E protein expression induced transmembrane currents that were blocked by various inhibitors. In an ion-reduced buffer system, currents were proton-dependent and blocked by viroporin inhibitors rimantadine and amantadine. I-V relationships of recombinant E protein were not pH-dependent in a classical buffer system with high extracellular Na+ and high intracellular K+. E-protein mediated currents were inhibited by amantadine and rimantadine, as well as 5-(N,N-hexamethylene)amiloride (HMA). We tested a total of 10 flavonoids, finding inhibitory activity of varying potency. Epigallocatechin and quercetin were most effective, with IC50 values of 1.5 ± 0.1 and 3.7 ± 0.2 nM, respectively, similar to the potency of rimantadine (IC50 = 1.7 ± 0.6 nM). Patch-clamp results were independently verified using a modified cell viability assay for viroporin inhibitors. These results contribute to the development of novel antiviral drugs that suppress virus activity and proliferation.
- Published
- 2021
23. Author response for 'A pair of non‐competing neutralizing human monoclonal antibodies protecting from disease in a SARS‐CoV‐2 infection model'
- Author
-
Roman Wölfel, Paul F. McKay, Jutta Eichler, Stefan Pöhlmann, Manuela Hauke, Dirk Mielenz, Edith Roth, Eva Grüner, Matthias Tenbusch, Thomas Grunwald, Thomas Winkler, Tobit Steinmetz, Sebastian R. Schulz, Ralf Wagner, Heinrich Sticht, Markus Hoffmann, Hans-Martin Jäck, Sandra Ciesek, Dominik Damm, Valentina Eberlein, Eileen Socher, Leila Issmail, Katharina Habenicht, Armin Ensser, David Peterhoff, Elie Richel, Wolfgang Schuh, Klaus Überla, Nadja Uhlig, Antonia Sophia Peter, Simon Dolles, Frank Neipel, Sandra Mueller-Schmucker, Robin J. Shattock, and Kirsten Fraedrich
- Subjects
medicine.drug_class ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,medicine ,Disease ,Biology ,Monoclonal antibody ,Virology - Published
- 2021
24. Loss of function of SVBP leads to autosomal recessive intellectual disability, microcephaly, ataxia, and hypotonia
- Author
-
Steffen Uebe, Nael Nadif Kasri, Heinrich Sticht, André Reis, Hans van Bokhoven, Wei Ba, Bassam Al Halak, Zafar Iqbal, Hasan Tawamie, Rami Abou Jamra, and Sheikh Riazuddin
- Subjects
Genetics ,Microcephaly ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Ataxia ,Muscular hypotonia ,Biology ,medicine.disease ,Hypotonia ,All institutes and research themes of the Radboud University Medical Center ,Real-time polymerase chain reaction ,Intellectual disability ,medicine ,medicine.symptom ,Gene ,Genetics (clinical) ,Loss function - Abstract
Identifying and characterizing novel causes of autosomal recessive intellectual disability based on systematic clinical and genetic evaluation, followed by functional experiments. Clinical examinations, genome-wide positional mapping, and sequencing were followed by quantitative polymerase chain reaction and western blot of the protein SVBP and its interaction partners. We then knocked down the gene in rat primary hippocampal neurons and evaluated the consequences on synapses. We identified a founder, homozygous stop-gain variant in SVBP (c.82C>T; p.[Gln28*]) in four affected individuals from two independent families with intellectual disability, microcephaly, ataxia, and muscular hypotonia. SVBP encodes a small chaperone protein that transports and stabilizes two angiogenesis regulators, VASH1 and VASH2. The altered protein is unstable and nonfunctional since transfected HeLa cells with mutant SVBP did not reveal evidence for immunoreactive SVBP protein fragments and cotransfection with VASH1 showed a severe reduction of VASH1 in medium and cell lysate. Knocking down Svbp in rat primary hippocampal neurons led to a significant decrease in the number of excitatory synapses. SVBP is not only involved in angiogenesis, but also has vital functions in the central nervous system. Biallelic loss-of-function variants in SVBP lead to intellectual disability.
- Published
- 2019
25. Decomposition of the SARS-CoV-2-ACE2 interface reveals a common trend among emerging viral variants
- Author
-
Eileen Socher, Heinrich Sticht, Friederike Zunke, Friedrich Paulsen, Philipp Arnold, Marcus Conrad, and Lukas Heger
- Subjects
chemistry.chemical_classification ,Infectivity ,Genetics ,Mutation ,biology ,Wild type ,medicine.disease_cause ,Virus ,Amino acid ,chemistry ,biology.protein ,medicine ,Tyrosine ,Antibody ,Threonine - Abstract
New viral variants of the SARS-CoV-2 virus show enhanced infectivity compared to wild type, resulting in an altered pandemic situation in affected areas. These variants are the B.1.1.7 (United Kingdom), B.1.1.7 with the additional E484K mutation, the B.1.351 variant (South Africa) and the P.1 variant (Brazil). Understanding the binding modalities between these viral variants and the host cell receptor ACE2 allows depicting changes, but also common motifs of virus-host cell interaction. The trimeric spike protein expressed at the viral surface contains the receptor-binding domain (RBD) that forms the molecular interface with ACE2. All the above-mentioned variants carry between one and three amino acid exchanges within the interface-forming region of the RBD, thereby altering the binding interface with ACE2. Using molecular dynamics simulations and decomposition of the interaction energies between the RBD and ACE2, we identified phenylalanine 486, glutamine 498, threonine 500 and tyrosine 505 as important interface-forming residues across viral variants. We also suggest a reduced binding energy between RBD and ACE2 in viral variants with higher infectivity, attributed to residue-specific differences in electrostatic interaction energy. Importantly, individual amino acid exchanges not only influence the affected position, but also alter the conformation of surrounding residues and affect their interaction potential as well. We demonstrate how computational methods can help to identify changed as well as common motifs across viral variants. These identified motifs might play a crucial role, in the strategical development of therapeutic interventions against the fast mutating SARS-CoV-2 virus.Significance StatementThe COVID-19 pandemic caused by the SARS-CoV-2 virus has significantly changed our lives. To date, there is a lack of neutralizing drugs that specifically target SARS-CoV-2. Hope lies in newly developed vaccines that effectively prevent severe cases of acute respiratory syndrome. However, emerging viral variants escape vaccine-induced immune-protection. Therefore, identification of appropriate molecular targets across viral variants is important for the development of second- and third-generation vaccines and inhibitory antibodies. In this study, we identify residues across viral variants that are important for viral binding to the host cell. As such residues cannot be replaced without diminishing infectivity of the virus, these residues represent primary targets for intervention, for example by neutralizing antibodies.
- Published
- 2021
26. Pontocerebellar hypoplasia due to bi-allelic variants in MINPP1
- Author
-
Henry Houlden, Mohammad Yahya Vahidi Mehrjardi, Timo Roser, Julia Hoefele, Mohammadreza Dehghani, Rami Abou Jamra, Reza Maroofian, Anja Heinze, Stefan D. Roosendaal, Dagmar Wieczorek, Frank Baas, Margarete Koch-Hogrebe, Farrah Rajabi, Heinrich Sticht, Erin Torti, Matias Wagner, Bart Appelhof, Graduate School, ANS - Amsterdam Neuroscience, Radiology and Nuclear Medicine, Human Genetics, and ARD - Amsterdam Reproduction and Development
- Subjects
Male ,Protein Folding ,Nonsense-mediated decay ,Pontocerebellar hypoplasia ,Mutation, Missense ,Article ,Serine ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cerebellar Diseases ,Aspartic acid ,INPP5E ,Genetics ,medicine ,Missense mutation ,Humans ,Inositol ,Allele ,Child ,Genetics (clinical) ,Alleles ,030304 developmental biology ,0303 health sciences ,Chemistry ,Homozygote ,Neurodevelopmental disorders ,Infant ,medicine.disease ,Phosphoric Monoester Hydrolases ,ddc ,Codon, Nonsense ,Child, Preschool ,Mutation ,Female ,030217 neurology & neurosurgery - Abstract
Pontocerebellar hypoplasia (PCH) describes a group of rare heterogeneous neurodegenerative diseases with prenatal onset. Here we describe eight children with PCH from four unrelated families harboring the homozygous MINPP1 (NM_004897.4) variants; c.75_94del, p.(Leu27Argfs*39), c.851 C > A, p.(Ala284Asp), c.1210 C > T, p.(Arg404*), and c.992 T > G, p.(Ile331Ser). The homozygous p.(Leu27Argfs*39) change is predicted to result in a complete absence of MINPP1. The p.(Arg404*) would likely lead to a nonsense mediated decay, or alternatively, a loss of several secondary structure elements impairing protein folding. The missense p.(Ala284Asp) affects a buried, hydrophobic residue within the globular domain. The introduction of aspartic acid is energetically highly unfavorable and therefore predicted to cause a significant reduction in protein stability. The missense p.(Ile331Ser) affects the tight hydrophobic interactions of the isoleucine by the disruption of the polar side chain of serine, destabilizing the structure of MINPP1. The overlap of the above-mentioned genotypes and phenotypes is highly improbable by chance. MINPP1 is the only enzyme that hydrolyses inositol phosphates in the endoplasmic reticulum lumen and several studies support its role in stress induced apoptosis. The pathomechanism explaining the disease mechanism remains unknown, however several others genes of the inositol phosphatase metabolism (e.g., INPP5K, FIG4, INPP5E, ITPR1) are correlated with phenotypes of neurodevelopmental disorders. Taken together, we present MINPP1 as a novel autosomal recessive pontocerebellar hypoplasia gene.
- Published
- 2021
27. Bi-allelic variants in SPATA5L1 lead to intellectual disability, spastic-dystonic cerebral palsy, epilepsy, and hearing loss
- Author
-
Serdal Güngör, Benita Grossmann, Bethany Y. Norton, Zubair M. Ahmed, Wendy K. Chung, John Neidhardt, Julie S. Cohen, Elodie Richard, Yoel Hirsch, Jiankang Li, Jozef Gecz, Ralf A. Husain, Saima Riazuddin, Maria J. Guillen Sacoto, Claudia Steen, Andreas Ziegler, G. Christoph Korenke, Dominic Lenz, Mahim Jain, Urania Kotzaeridou, Henry Houlden, Theresa Brunet, Yavuz Oktay, Semra Hiz, Patricia Cornejo, Sheetal Shetty, Alastair H. MacLennan, Nazira Zharkinbekova, Bader Alhaddad, Dani L. Webber, Mary Alice Abbott, Hanns Lochmüller, Rauan Kaiyrzhanov, Melissa Yelton, Cecilia Mancini, Hakon Hakonarson, Amy Crunk, Simona Amenta, Yiran Guo, Jan Kaslin, Clare L. van Eyk, Richard Webster, Arianna Tucci, Alex M. Pagnozzi, Robert B. Hufnagel, Kirsty McWalter, Sandra M. Nordlie, Kaya Bilguvar, Pasquale Striano, Matias Wagner, Florian Kreuder, Lisa Worgan, Ashley P.L. Marsh, Anna Chassevent, Warren A. Marks, James Liu, Brandon S. Guida, Maria Margherita Mancardi, Kelly Harper, Lance H. Rodan, Rhonda E. Schnur, Dianela Judith Claps Sepulveda, Tzvi Weiden, Michele Pinelli, Marion Rapp, Helen Magee, Jesia G. Berry, Aboulfazl Rad, Michael C. Kruer, Mark A. Corbett, Rita Horvath, Constance Smith-Hicks, Joseph Ekstein, Marta Owczarek-Lipska, Somayeh Bakhtiari, Heinrich Sticht, Thomas Meitinger, Anne M. Comi, Alyssa Blesson, Iris Marquardt, Francesca Clementina Radio, Sergio Padilla-Lopez, Giuseppe Marangi, Christine Makowski, Mona Grimmel, Marco Tartaglia, Sheng Chih Jin, Federico Zara, Andreas Hahn, Shrikant Mane, Michael C Fahey, Marcella Zollino, Barbara Vona, Peter D. Turnpenny, Manuela Morleo, Ute Grasshoff, Amber Begtrup, Richard E. Person, Annalaura Torella, Alexander Münchau, Vincenzo Nigro, Reza Maroofian, John Christodoulou, Tobias B. Haack, Vincenzo Salpietro, Richard, E. M., Bakhtiari, S., Marsh, A. P. L., Kaiyrzhanov, R., Wagner, M., Shetty, S., Pagnozzi, A., Nordlie, S. M., Guida, B. S., Cornejo, P., Magee, H., Liu, J., Norton, B. Y., Webster, R. I., Worgan, L., Hakonarson, H., Li, J., Guo, Y., Jain, M., Blesson, A., Rodan, L. H., Abbott, M. -A., Comi, A., Cohen, J. S., Alhaddad, B., Meitinger, T., Lenz, D., Ziegler, A., Kotzaeridou, U., Brunet, T., Chassevent, A., Smith-Hicks, C., Ekstein, J., Weiden, T., Hahn, A., Zharkinbekova, N., Turnpenny, P., Tucci, A., Yelton, M., Horvath, R., Gungor, S., Hiz, S., Oktay, Y., Lochmuller, H., Zollino, M., Morleo, M., Marangi, G., Nigro, V., Torella, A., Pinelli, M., Amenta, S., Husain, R. A., Grossmann, B., Rapp, M., Steen, C., Marquardt, I., Grimmel, M., Grasshoff, U., Korenke, G. C., Owczarek-Lipska, M., Neidhardt, J., Radio, F. C., Mancini, C., Claps Sepulveda, D. J., Mcwalter, K., Begtrup, A., Crunk, A., Guillen Sacoto, M. J., Person, R., Schnur, R. E., Mancardi, M. M., Kreuder, F., Striano, P., Zara, F., Chung, W. K., Marks, W. A., van Eyk, C. L., Webber, D. L., Corbett, M. A., Harper, K., Berry, J. G., Maclennan, A. H., Gecz, J., Tartaglia, M., Salpietro, V., Christodoulou, J., Kaslin, J., Padilla-Lopez, S., Bilguvar, K., Munchau, A., Ahmed, Z. M., Hufnagel, R. B., Fahey, M. C., Maroofian, R., Houlden, H., Sticht, H., Mane, S. M., Rad, A., Vona, B., Jin, S. C., Haack, T. B., Makowski, C., Hirsch, Y., Riazuddin, S., and Kruer, M. C.
- Subjects
Male ,Microcephaly ,Pathology ,Settore MED/03 - GENETICA MEDICA ,sensorineural hearing loss ,Epilepsy ,Neurodevelopmental disorder ,sensorineural hearing lo ,Genetics (clinical) ,Allele ,ATPases Associated with Diverse Cellular Activitie ,medicine.anatomical_structure ,Muscle Spasticity ,Child, Preschool ,Sensorineural hearing loss ,Female ,movement disorder ,medicine.symptom ,AAA+ superfamily ,Human ,Adult ,medicine.medical_specialty ,Adolescent ,Hearing loss ,Aaa+ Superfamily ,Atpase ,Spata5l1 ,Cerebral Palsy ,Intellectual Disability ,Movement Disorder ,Neurodevelopmental Disorder ,Sensorineural Hearing Loss ,Biology ,Cerebral palsy ,White matter ,Young Adult ,Report ,Genetics ,medicine ,Animals ,Humans ,ATPase ,Genetic Predisposition to Disease ,Hearing Loss ,SPATA5L1 ,Hearing Lo ,Alleles ,cerebral palsy ,Periventricular leukomalacia ,Animal ,Infant, Newborn ,Infant ,Genetic Variation ,medicine.disease ,neurodevelopmental disorder ,Rats ,ATPases Associated with Diverse Cellular Activities ,Rat - Abstract
Spermatogenesis-associated 5 like 1 (SPATA5L1) represents an orphan gene encoding a protein of unknown function. We report 28 bi-allelic variants in SPATA5L1 associated with sensorineural hearing loss in 47 individuals from 28 (26 unrelated) families. In addition, 25/47 affected individuals (53%) presented with microcephaly, developmental delay/intellectual disability, cerebral palsy, and/or epilepsy. Modeling indicated damaging effect of variants on the protein, largely via destabilizing effects on protein domains. Brain imaging revealed diminished cerebral volume, thin corpus callosum, and periventricular leukomalacia, and quantitative volumetry demonstrated significantly diminished white matter volumes in several individuals. Immunofluorescent imaging in rat hippocampal neurons revealed localization of Spata511 in neuronal and glial cell nuclei and more prominent expression in neurons. In the rodent inner ear, Spata511 is expressed in the neurosensory hair cells and inner ear supporting cells. Transcriptomic analysis performed with fibroblasts from affected individuals was able to distinguish affected from controls by principal components. Analysis of differentially expressed genes and networks suggested a role for SPATA5L1 in cell surface adhesion receptor function, intracellular focal adhesions, and DNA replication and mitosis. Collectively, our results indicate that bi-allelic SPATA5L1 variants lead to a human disease characterized by sensorineural hearing loss (SNHL) with or without a nonprogressive mixed neurodevelopmental phenotype.
- Published
- 2021
28. Clinical and molecular delineation of spondylocostal dysostosis type 3
- Author
-
Cornelia Kraus, Mandy Krumbiegel, Arif B. Ekici, Heiko Koller, Sarah Schuhmann, Christian Thiel, Steffen Uebe, André Reis, and Heinrich Sticht
- Subjects
business.industry ,Genetics ,Medicine ,Computational biology ,business ,medicine.disease ,Genetics (clinical) ,Spondylocostal dysostosis ,ddc - Published
- 2020
29. New insights into the clinical and molecular spectrum of the novel CYFIP2-related neurodevelopmental disorder and impairment of the WRC-mediated actin dynamics
- Author
-
Katharina Steindl, Alain Verloes, Cornelia Kraus, Rachel Fisher, Katrin Õunap, Amber Begtrup, Steffen Syrbe, Theresa Brunet, Antonio Vitobello, Laurence Faivre, Reza Asadollahi, Jessica Becker, Maja Hempel, Dave A Dyment, Christiane Zweier, John H McDermott, Bernt Popp, Elaine Suk-Ying Goh, Lynette G. Sadleir, Anaïs Begemann, Siddharth Banka, Gwenaël Le Guyader, Elisabeth Schuler, Anne-Sophie Denommé-Pichon, Kathleen Brown, Gaetan Lesca, Frédéric Tran Mau-Them, Lucia Ribeiro Machado Haertel, Maryline Carneiro, Amelie Theresa Van der Ven, Markus Zweier, Hartmut Engels, Heinrich Sticht, Theresia Herget, Jessika Johannsen, Bader Alhaddad, Nadine N. Hauer, Robert C. Day, Tiia Reimand, M. J. Hajianpour, Manuel Schiff, Kirsty McWalter, Margarita Saenz, Tatjana Bierhals, Pierre Meyer, Ange-Line Bruel, Martina Russo, Korbinian M. Riedhammer, Kirsten Cremer, Anita Rauch, Marjolaine Willems, Equipe GAD (LNC - U1231), Lipides - Nutrition - Cancer [Dijon - U1231] (LNC), Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM), FHU TRANSLAD (CHU de Dijon), Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand (CHU Dijon), Centre hospitalier universitaire de Poitiers (CHU Poitiers), Centre de recherche en neurosciences de Lyon (CRNL), Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Université Jean Monnet [Saint-Étienne] (UJM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Physiologie & médecine expérimentale du Cœur et des Muscles [U 1046] (PhyMedExp), Université de Montpellier (UM)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Maladies neurodéveloppementales et neurovasculaires (NeuroDiderot (UMR_S_1141 / U1141)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Paris (UP), and Hôpital Robert Debré
- Subjects
0301 basic medicine ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,WAVEregulatory complex (WRC) ,030105 genetics & heredity ,Biology ,Article ,Intellectual disability ,Epilepsy ,CYFIP2 ,WAVE-regulatory complex (WRC) ,WASF ,03 medical and health sciences ,Neurodevelopmental disorder ,Seizures ,medicine ,Missense mutation ,Humans ,Genetics(clinical) ,Gene ,Genetics (clinical) ,Actin ,Adaptor Proteins, Signal Transducing ,Genetics ,medicine.disease ,Actin cytoskeleton ,Phenotype ,Hypotonia ,Actins ,3. Good health ,ddc ,030104 developmental biology ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,Neurodevelopmental Disorders ,intellectual disability ,epilepsy ,medicine.symptom - Abstract
International audience; Purpose: A few de novo missense variants in the cytoplasmic FMRP-interacting protein 2 (CYFIP2) gene have recently been described as a novel cause of severe intellectual disability, seizures, and hypotonia in 18 individuals, with p.Arg87 substitutions in the majority.Methods: We assembled data from 19 newly identified and all 18 previously published individuals with CYFIP2 variants. By structural modeling and investigation of WAVE-regulatory complex (WRC)-mediated actin polymerization in six patient fibroblast lines we assessed the impact of CYFIP2 variants on the WRC.Results: Sixteen of 19 individuals harbor two previously described and 11 novel (likely) disease-associated missense variants. We report p.Asp724 as second mutational hotspot (4/19 cases). Genotype–phenotype correlation confirms a consistently severe phenotype in p.Arg87 patients but a more variable phenotype in p.Asp724 and other substitutions. Three individuals with milder phenotypes carry putative loss-of-function variants, which remain of unclear pathogenicity. Structural modeling predicted missense variants to disturb interactions within the WRC or impair CYFIP2 stability. Consistent with its role in WRC-mediated actin polymerization we substantiate aberrant regulation of the actin cytoskeleton in patient fibroblasts.Conclusion: Our study expands the clinical and molecular spectrum of CYFIP2-related neurodevelopmental disorder and provides evidence for aberrant WRC-mediated actin dynamics as contributing cellular pathomechanism.
- Published
- 2020
30. Fine-Tuning of Neuronal Ion Channels-Mapping of Residues Involved in Glucose Sensitivity of Recombinant Human Glycine Receptors
- Author
-
Ulrike Breitinger, Rama Ashraf Hussein, Marwa Ahmed, Hans-Georg Breitinger, and Heinrich Sticht
- Subjects
Agonist ,Physiology ,medicine.drug_class ,Cognitive Neuroscience ,Ethanol binding ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Receptors, Glycine ,medicine ,Humans ,Binding site ,Receptor ,Glycine receptor ,030304 developmental biology ,Alanine ,chemistry.chemical_classification ,Neurons ,0303 health sciences ,Ethanol ,Chemistry ,Cell Biology ,General Medicine ,Recombinant Proteins ,Amino acid ,Glucose binding ,Glucose ,Biophysics ,Mutagenesis, Site-Directed ,030217 neurology & neurosurgery - Abstract
The inhibitory glycine receptor (GlyR) mediates synaptic inhibition in the spinal cord, brain stem, and other regions of the mammalian central nervous system. Glucose was shown to potentiate α1 GlyRs by interacting with K143. Here, additional amino acids involved in glucose modulation were identified using a structure-based approach of site-directed mutagenesis followed by whole-cell patch-clamp analysis. We identified two additional lysine residues in the α1 GlyR extracellular domain, K16 and K281, that were involved in glucose modulation. Mutation of either residue to alanine abolished glucose potentiation. Residue K281 is located in the same pocket as K143 and could thus contribute to glucose binding. The double mutant K143A-K281A showed a 6-fold increase of EC50, while EC50 of both single mutants K143A and K281A was only slightly increased (1.7- and 1.3-fold, respectively). K16 is located at an analgesic binding site that is distant from the agonist or glucose sites, and the K16A mutation may generate a receptor species that is not potentiated. GlyR position α1-S267 is close to the postulated glucose binding site and known for interactions with ethanol and anesthetics. In the presence of glucose, GlyR α1 mutants S267A, S267I, and S267R showed potentiation, no effect, and reduction of current responses, respectively. This pattern follows that of ethanol modulation and suggests that the interaction sites of glucose and ethanol are identical or located close to each other. Our results support the presence of a distinct binding site for glucose on the glycine receptor, overlapping with the ivermectin/ethanol binding pocket near the transmembrane region and the TM2-3 loop.
- Published
- 2020
31. Rare slow channel congenital myasthenic syndromes without repetitive compound muscle action potential and dramatic response to low dose fluoxetine
- Author
-
Hacer Durmus, Feza Deymeer, Said Hashemolhosseini, Heinrich Sticht, and Serdar Ceylaner
- Subjects
Adult ,Male ,medicine.medical_specialty ,Neurology ,Mutation, Missense ,Action Potentials ,Bioinformatics ,medicine.disease_cause ,Neuromuscular junction ,Protein Structure, Secondary ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Fluoxetine ,medicine ,Missense mutation ,Humans ,Receptors, Cholinergic ,030212 general & internal medicine ,Amino Acid Sequence ,Muscle, Skeletal ,Exome sequencing ,Sanger sequencing ,Myasthenic Syndromes, Congenital ,Mutation ,Dose-Response Relationship, Drug ,business.industry ,General Medicine ,Compound muscle action potential ,Pedigree ,Protein Structure, Tertiary ,Nicotinic acetylcholine receptor ,medicine.anatomical_structure ,Treatment Outcome ,symbols ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Congenital myasthenic syndromes are rare hereditary disorders caused by mutations associated with proteins of the neuromuscular junction. Abnormal ‘‘gain of function’’ mutations result in prolonged nicotinic acetylcholine receptor channel open state causing a rare subtype of CMS, slow-channel CMS (SCCMS). Mutations in the delta subunit encoding the gene, CHRND, resulting in SCCMS are extremely rare. An important clue to the diagnosis of SCCMS is repetitive CMAP’s. Fluoxetine, usually at high doses, is used to treat SCCMS. The mutation, recently described in one patient, was identified by whole exome sequencing and validated, and its segregation with the disease was ascertained by Sanger sequencing. Here, we describe clinical and genetic findings of an early onset SCCMS patient carrying a very rare missense mutation c.880C > T in CHRND causing a highly conserved leucine to phenylalanine substitution in the M2 domain of CHRND. The patient had no repetitive CMAP. He had a dramatic response to fluoxetine at low–moderate doses (40 mg/day), increasing over months: Being wheelchair bound, he could walk independently after treatment. Rare cases may offer insight into the pathological gating mechanism leading to CMS. SCCMS should be suspected even without a repetitive CMAP. Fluoxetine at relatively low doses can be a very effective treatment.
- Published
- 2020
32. De novo variants in SNAP25 cause an early-onset developmental and epileptic encephalopathy
- Author
-
Katy Barwick, F. Lucy Raymond, Pia Zacher, Saskia B. Wortmann, Koen L.I. van Gassen, Michaela Bonfert, Bernt Popp, Eva H. Brilstra, Marie-José van den Bogaard, Alyssa Gates, Johan Lundgren, Dewi P. Bakker, Ashley C. Taylor, Alba Sanchis-Juan, Rikke S. Møller, Jessica Van Ziffle, Matias Wagner, Anne Slavotinek, Heather C Mefford, Heinrich Sticht, Joseph T. Shieh, Manju A. Kurian, Marwan Shinawi, Patrick Devine, Boris Keren, Konrad Platzer, Gabriella Horvath, Jennifer Keller-Ramey, Kathleen A. Leppig, Nicholas Stong, Alexandrea Wadley, Tomi L. Toler, Julian R. Sampson, Richard H. van Jaarsveld, Caroline Nava, Saskia N. van der Crabben, Jennifer Friedman, Mohamad A. Mikati, Marie T. McDonald, Vandana Shashi, Angus John Clarke, Wendy K. Chung, Amy McTague, Johannes R. Lemke, Tommy Stödberg, Jennifer A. Sullivan, Chiara Klöckner, Virgina Lee, and Cyril Mignot
- Subjects
0303 health sciences ,Movement disorders ,business.industry ,Cortical visual impairment ,medicine.disease ,Bioinformatics ,Phenotype ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Atrophy ,Intellectual disability ,Medicine ,STXBP1 ,Missense mutation ,medicine.symptom ,business ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
PurposeThis study aims to provide the first comprehensive description of the phenotypic and genotypic spectrum of SNAP25 developmental and epileptic encephalopathy (SNAP25- DEE) by reviewing newly identified and previously reported individuals.MethodsIndividuals harboring heterozygous missense or truncating variants in SNAP25 were assembled through collaboration with international colleagues, matchmaking platforms and literature review. For each individual, detailed phenotyping, classification and structural modeling of the identified variant was performed.ResultsThe cohort comprises 20 individuals with (likely) pathogenic de novo variants in SNAP25. Intellectual disability and early-onset epilepsy were identified as the core symptoms of SNAP25-DEE, with recurrent findings of movement disorders, cortical visual impairment and brain atrophy. Structural modeling for all variants predicted possible functional defects concerning SNAP25 or impaired interaction with other components of the SNARE complex.ConclusionWe provide a first comprehensive description of SNAP25-DEE with intellectual disability and early onset epilepsy mostly occurring before the age of two years. These core symptoms and additional recurrent phenotypes show an overlap to genes encoding other components or associated proteins of the SNARE complex such as STX1B, STXBP1 or VAMP2. Thus, these findings advance the concept of a group of neurodevelopmental disorders that may be termed “SNAREopathies”.
- Published
- 2020
- Full Text
- View/download PDF
33. Expanding the genotypic and phenotypic spectrum of severe serine biosynthesis disorders
- Author
-
Perrine Pennamen, Caroline Rooryck, Yves Sznajer, Anne-Karin Kahlert, Isolina Riaño-Galán, Denny Schanze, David J. Amor, Eva Bermejo-Sánchez, Maie Walsh, Ariana Kariminejad, Siavash Ghaderi-Sohi, Mohamad Hasan Kariminejad, Ian P Hayes, Sönke Weinert, Patrick J. Morrison, Patrick Shannon, Martin Zenker, Gemma Poke, Annick Toutain, Suonavy Khung‐Savatovsky, Heinrich Sticht, David Chitayat, Fatima Abdelfattah, Evren Gumus, Marie-Laure Vuillaume, Katherine D. Mathews, Sabine Weidensee, Luisa Weiß, Benjamin W. Darbro, German Federal Ministry of Education and Research, Instituto de Salud Carlos III, and Fundación 1000 sobre Defectos Congénitos
- Subjects
Male ,Neu–Laxova syndrome ,Mutant ,Autosomal recessive ,Limb Deformities, Congenital ,Biology ,medicine.disease_cause ,Serine ,03 medical and health sciences ,Fetus ,Genotype ,Genetics ,medicine ,Neu-Laxova syndrome ,Humans ,Abnormalities, Multiple ,Phosphoglycerate dehydrogenase ,Gene ,PHGDH ,Genetics (clinical) ,Genetic Association Studies ,Phosphoglycerate Dehydrogenase ,Transaminases ,030304 developmental biology ,0303 health sciences ,Mutation ,Brain Diseases ,Fetal Growth Retardation ,Genotype–phenotype correlation ,030305 genetics & heredity ,Infant, Newborn ,L-Serine biosynthesis ,Ichthyosis ,medicine.disease ,Phenotype ,PSAT1 ,Microcephaly ,Female - Abstract
Financial assistance was received in support of the study by grants from the German Federal Ministry of Education and Research (BMBF) (GeNeRARe, FKZ: 01GM1519D) to M. Z. and from the Institute of Health Carlos III: Convenio ISCIII‐ASEREMAC, and Fundación 1000 sobre Defectos Congénitos, of Spain to E. B.‐S. and I. R. G., Abdelfattah, F., Kariminejad, A., Kahlert, A.-K., Morrison, P.J., Gumus, E., Mathews, K.D., Darbro, B.W., Amor, D.J., Walsh, M., Sznajer, Y., Weiß, L., Weidensee, S., Chitayat, D., Shannon, P., Bermejo-Sánchez, E., Riaño-Galán, I., Hayes, I., Poke, G., Rooryck, C., Pennamen, P., Khung-Savatovsky, S., Toutain, A., Vuillaume, M.-L., Ghaderi-Sohi, S., Kariminejad, M.H., Weinert, S., Sticht, H., Zenker, M., Schanze, D.
- Published
- 2020
34. The CD83 Molecule – An Important Immune Checkpoint
- Author
-
Linda Grosche, Matthias Lechmann, Elisabeth Zinser, Alexander Steinkasserer, Heinrich Sticht, Ilka Knippertz, Christina König, Andreas B. Wild, Dmytro Royzman, and Yves A. Muller
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,immune tolerance ,Immunology ,Immunoglobulins ,Inflammation ,chemical and pharmacologic phenomena ,Review ,Adaptive Immunity ,Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Autoimmunity ,Immune tolerance ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigens, CD ,CD83 ,Medizinische Fakultät ,viral escape mechanism ,medicine ,Animals ,Humans ,Immunology and Allergy ,ddc:610 ,B-Lymphocytes ,Membrane Glycoproteins ,Host Microbial Interactions ,Microglia ,autoimmunity ,Cell Differentiation ,hemic and immune systems ,Dendritic Cells ,Immune Checkpoint Proteins ,Immune checkpoint ,Cell biology ,Transplantation ,030104 developmental biology ,medicine.anatomical_structure ,medicine.symptom ,Signal transduction ,lcsh:RC581-607 ,Treg cells ,030215 immunology - Abstract
The CD83 molecule has been identified to be expressed on numerous activated immune cells, including B and T lymphocytes, monocytes, dendritic cells, microglia, and neutrophils. Both isoforms of CD83, the membrane-bound as well as its soluble form are topic of intensive research investigations. Several studies revealed that CD83 is not a typical co-stimulatory molecule, but rather plays a critical role in controlling and resolving immune responses. Moreover, CD83 is an essential factor during the differentiation of T and B lymphocytes, and the development and maintenance of tolerance. The identification of its interaction partners as well as signaling pathways have been an enigma for the last decades. Here, we report the latest data on the expression, structure, and the signaling partners of CD83. In addition, we review the regulatory functions of CD83, including its striking modulatory potential to maintain the balance between tolerance versus inflammation during homeostasis or pathologies. These immunomodulatory properties of CD83 emphasize its exceptional therapeutic potential, which has been documented in specific preclinical disease models.
- Published
- 2020
35. Myeloperoxidase Modulates Inflammation in Generalized Pustular Psoriasis and Additional Rare Pustular Skin Diseases
- Author
-
Jörg C. Prinz, Florina Kersting, Markus H. Hoffmann, Peter Schulz, Sabine Löhr, Jonas Hahn, Ulrike Hüffmeier, Christine Schauer, Gunter Aßmann, Anne Gregor, André Reis, Abdelaziz Sefiani, Arif B. Ekici, Sandra Philipp, Claudia Riepe, Wiebke Sondermann, Georg Schett, Madelaine Hahn, Knut Schäkel, Mark Ringer, Michael Sticherling, Silke Frey, Maximilien Euler, Christian Thiel, Jaber Lyahyai, Steffen Uebe, Heinrich Sticht, Stefan Haskamp, Dagmar Wilsmann-Theis, Cindy Flamann, Adam Lesner, Rotraut Mößner, Heiko Bruns, Vinzenz Oji, Stephan von Hörsten, and Benjamin Frey
- Subjects
0301 basic medicine ,Male ,Neutrophils ,Medizin ,Extracellular Traps ,Pathogenesis ,acute generalized 4 exanthematous pustulosis ,Mice ,0302 clinical medicine ,Medicine ,oligogenic inheritance ,impaired NETosis ,Genetics (clinical) ,Skin ,efferocytosis ,biology ,AGEP ,3. Good health ,myeloperoxidase ,030220 oncology & carcinogenesis ,Myeloperoxidase ,Cytokines ,Female ,MPO deficiency ,generalized pustular psoriasis ,medicine.symptom ,acrodermatitis continua suppurativa Hallopeau ,Adult ,Proteases ,Phagocytosis ,Inflammation ,Skin Diseases ,Article ,03 medical and health sciences ,Rare Diseases ,Genetics ,Animals ,Humans ,Psoriasis ,ACH ,Efferocytosis ,Peroxidase ,business.industry ,Interleukins ,Neutrophil extracellular traps ,activation of IL-36 precursors ,medicine.disease ,030104 developmental biology ,Immunology ,Mutation ,Generalized pustular psoriasis ,biology.protein ,GPP ,business ,Interleukin-1 - Abstract
Generalized pustular psoriasis (GPP) is a severe multi-systemic inflammatory disease characterized by neutrophilic pustulosis and triggered by pro-inflammatory IL-36 cytokines in skin. While 19%–41% of affected individuals harbor bi-allelic mutations in IL36RN, the genetic cause is not known in most cases. To identify and characterize new pathways involved in the pathogenesis of GPP, we performed whole-exome sequencing in 31 individuals with GPP and demonstrated effects of mutations in MPO encoding the neutrophilic enzyme myeloperoxidase (MPO). We discovered eight MPO mutations resulting in MPO -deficiency in neutrophils and monocytes. MPO mutations, primarily those resulting in complete MPO deficiency, cumulatively associated with GPP (p = 1.85E−08; OR = 6.47). The number of mutant MPO alleles significantly differed between 82 affected individuals and >4,900 control subjects (p = 1.04E−09); this effect was stronger when including IL36RN mutations (1.48E−13) and correlated with a younger age of onset (p = 0.0018). The activity of four proteases, previously implicated as activating enzymes of IL-36 precursors, correlated with MPO deficiency. Phorbol-myristate-acetate-induced formation of neutrophil extracellular traps (NETs) was reduced in affected cells (p = 0.015), and phagocytosis assays in MPO-deficient mice and human cells revealed altered neutrophil function and impaired clearance of neutrophils by monocytes (efferocytosis) allowing prolonged neutrophil persistence in inflammatory skin. MPO mutations contribute significantly to GPP’s pathogenesis. We implicate MPO as an inflammatory modulator in humans that regulates protease activity and NET formation and modifies efferocytosis. Our findings indicate possible implications for the application of MPO inhibitors in cardiovascular diseases. MPO and affected pathways represent attractive targets for inducing resolution of inflammation in neutrophil-mediated skin diseases.
- Published
- 2020
36. High-resolution crystal structures of two prototypical β- and γ-herpesviral nuclear egress complexes unravel the determinants of subfamily specificity
- Author
-
Josephine Lösing, Kerstin Hof, Sascha A. Walzer, Stephanie Holst, Sebastian Weigert, Claudia Egerer-Sieber, Yves A. Muller, Simon Dolles, Tobias Höllriegl, Sigrun Häge, Jutta Eichler, Marcus Conrad, Sewar Alkhashrom, Heinrich Sticht, Manfred Marschall, and Eric Sonntag
- Subjects
0301 basic medicine ,Human cytomegalovirus ,Herpesvirus 4, Human ,Subfamily ,Viral protein ,Recombinant Fusion Proteins ,Cytomegalovirus ,Computational biology ,medicine.disease_cause ,Crystallography, X-Ray ,Biochemistry ,Virus ,Protein–protein interaction ,03 medical and health sciences ,Viral Proteins ,Gammaherpesvirinae ,medicine ,Betaherpesvirinae ,Humans ,Amino Acid Sequence ,Molecular Biology ,030102 biochemistry & molecular biology ,Chemistry ,Molecular Bases of Disease ,Cell Biology ,Surface Plasmon Resonance ,medicine.disease ,digestive system diseases ,Protein Structure, Tertiary ,030104 developmental biology ,Capsid ,Docking (molecular) ,Nuclear lamina ,Peptides ,HeLa Cells - Abstract
Herpesviruses uniquely express two essential nuclear egress-regulating proteins forming a heterodimeric basic structure of the nuclear egress complex (core NEC). These core NECs serve as a hexameric lattice-structured platform for capsid docking and recruit viral and cellular NEC-associated factors that jointly exert nuclear lamina- and membrane-rearranging functions (multicomponent NEC). Here, we report the X-ray structures of β- and γ-herpesvirus core NECs obtained through an innovative recombinant expression strategy based on NEC-hook::NEC-groove protein fusion constructs. This approach yielded the first structure of γ-herpesviral core NEC, namely the 1.56 A structure of Epstein-Barr virus (EBV) BFRF1–BFLF2, as well as an increased resolution 1.48 A structure of human cytomegalovirus (HCMV) pUL50-pUL53. Detailed analysis of these structures revealed that the prominent hook segment is absolutely required for core NEC formation and contributes approximately 80% of the interaction surface of the globular domains of NEC proteins. Moreover, using HCMV::EBV hook domain swap constructs, computational prediction of the roles of individual hook residues for binding, and quantitative binding assays with synthetic peptides presenting the HCMV- and EBV-specific NEC hook sequences, we characterized the unique hook-into-groove NEC interaction at various levels. Although the overall physicochemical characteristics of the protein interfaces differ considerably in these β- and γ-herpesvirus NECs, the binding free energy contributions of residues displayed from identical positions are similar. In summary, the results of our study reveal critical details of the molecular mechanism of herpesviral NEC interactions and highlight their potential as an antiviral drug target.
- Published
- 2020
37. De novo variants in SNAP25 cause an early-onset developmental and epileptic encephalopathy
- Author
-
Marwan Shinawi, Jessica Van Ziffle, Carsten G. Bönnemann, Mohamad A. Mikati, Vandana Shashi, Konrad Platzer, Manju A. Kurian, Katy Barwick, Kathleen A. Leppig, Patrick Devine, F. Lucy Raymond, Tomi L. Toler, Johan Lundgren, Koen L.I. van Gassen, Anne Slavotinek, Saskia N. van der Crabben, Wendy K. Chung, Richard H. van Jaarsveld, Matias Wagner, Rikke S. Møller, Marie T. McDonald, Pia Zacher, Kristen Wigby, Heather C Mefford, Dewi P. Bakker, Jennifer Friedman, Angus John Clarke, Joseph T. Shieh, Holly E. Babcock, Julian R. Sampson, Amy McTague, Jamal Ghoumid, Bernt Popp, Saskia B. Wortmann, Emma Hobson, Michaela Bonfert, Gabriella Horvath, Chiara Klöckner, Virgina Lee, Cyril Mignot, Yuri A. Zarate, Jennifer A. Sullivan, Marie-José H. van den Boogaard, Johannes R. Lemke, Alba Sanchis-Juan, Tommy Stödberg, Heinrich Sticht, Eva H. Brilstra, Alyssa Gates, Caroline Nava, Nicholas Stong, Sandra Donkervoort, Alexandrea Wadley, Boris Keren, Jamie L. Fraser, Ashley C. Taylor, Jennifer Keller-Ramey, Human Genetics, Pediatric surgery, Amsterdam Neuroscience - Neuroinfection & -inflammation, and Human genetics
- Subjects
medicine.medical_specialty ,Movement disorders ,Synaptosomal-Associated Protein 25 ,Bioinformatics ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Atrophy ,Intellectual Disability ,Intellectual disability ,Medicine ,STXBP1 ,Missense mutation ,Humans ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Brain Diseases ,business.industry ,STX1B ,medicine.disease ,Phenotype ,Neurodevelopmental Disorders ,Child, Preschool ,Medical genetics ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Purpose\ud This study aims to provide a comprehensive description of the phenotypic and genotypic spectrum of SNAP25 developmental and epileptic encephalopathy (SNAP25-DEE) by reviewing newly identified and previously reported individuals.\ud \ud Methods\ud Individuals harboring heterozygous missense or loss-of-function variants in SNAP25 were assembled through collaboration with international colleagues, matchmaking platforms, and literature review. For each individual, detailed phenotyping, classification, and structural modeling of the identified variant were performed.\ud \ud Results\ud The cohort comprises 23 individuals with pathogenic or likely pathogenic de novo variants in SNAP25. Intellectual disability and early-onset epilepsy were identified as the core symptoms of SNAP25-DEE, with recurrent findings of movement disorders, cerebral visual impairment, and brain atrophy. Structural modeling for all variants predicted possible functional defects concerning SNAP25 or impaired interaction with other components of the SNARE complex.\ud \ud Conclusion\ud We provide a comprehensive description of SNAP25-DEE with intellectual disability and early-onset epilepsy mostly occurring before the age of two years. These core symptoms and additional recurrent phenotypes show an overlap to genes encoding other components or associated proteins of the SNARE complex such as STX1B, STXBP1, or VAMP2. Thus, these findings advance the concept of a group of neurodevelopmental disorders that may be termed “SNAREopathies.”
- Published
- 2020
38. Haploinsufficiency of vascular endothelial growth factor related signaling genes is associated with tetralogy of Fallot
- Author
-
Anne S. Bassett, Meredith Curtis, Gregory Costain, Miriam S. Reuter, Stephen W. Scherer, Chelsea Lowther, Rebekah Jobling, Christian R. Marshall, Candice K. Silversides, Erwin Oechslin, Raymond H. Kim, Heinrich Sticht, Bhooma Thiruvahindrapuram, Rajiv Chaturvedi, Susan Walker, S. Mohsen Hosseini, Roozbeh Manshaei, Spencer van Mil, Tracy Heung, Rachel M. Wald, and Eriskay Liston
- Subjects
0301 basic medicine ,Proband ,Adult ,Male ,Vascular Endothelial Growth Factor A ,Haploinsufficiency ,030105 genetics & heredity ,Biology ,Brief Communication ,Pathogenesis ,03 medical and health sciences ,chemistry.chemical_compound ,Loss of Function Mutation ,Conotruncal defect ,medicine ,Humans ,Genetic Predisposition to Disease ,FLT4 ,Genetics (clinical) ,Genetic Association Studies ,Tetralogy of Fallot ,Aged ,Genetics ,Whole Genome Sequencing ,conotruncal defects ,Middle Aged ,medicine.disease ,Vascular Endothelial Growth Factor Receptor-3 ,VEGF ,congenital heart disease ,Vascular Endothelial Growth Factor Receptor-2 ,3. Good health ,Vascular endothelial growth factor ,genome sequencing ,Vascular endothelial growth factor A ,030104 developmental biology ,chemistry ,Female ,Signal Transduction - Abstract
Purpose To determine disease-associated single-gene variants in conotruncal defects, particularly tetralogy of Fallot (TOF). Methods We analyzed for rare loss-of-function and deleterious variants in FLT4 (VEGFR3) and other genes in the vascular endothelial growth factor (VEGF) pathway, as part of a genome sequencing study involving 175 adults with TOF from a single site. Results We identified nine (5.1%) probands with novel FLT4 variants: seven loss-of-function, including an 8-kb deletion, and two predicted damaging. In ten other probands we found likely disruptive variants in VEGF-related genes: KDR (VEGFR2; two stopgain and two nonsynonymous variants), VEGFA, FGD5, BCAR1, IQGAP1, FOXO1, and PRDM1. Detection of VEGF-related variants (19/175, 10.9%) was associated with an increased prevalence of absent pulmonary valve (26.3% vs. 3.4%, p
- Published
- 2018
39. IFN-γ-response mediator GBP-1 represses human cell proliferation by inhibiting the Hippo signaling transcription factor TEAD
- Author
-
Jürgen Behrens, Bea Unterer, Marina Leone, Clara Tenkerian, Felix B. Engel, Heinrich Sticht, Nathalie Britzen-Laurent, Mekala Gunasekaran, Thomas Wittenberg, Elisabeth Naschberger, Veit Wiesmann, Michael Stürzl, and Publica
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Protein domain ,Mutation, Missense ,colorectal cancer ,GTPase ,medicine.disease_cause ,Biochemistry ,Protein Structure, Secondary ,03 medical and health sciences ,Interferon-gamma ,0302 clinical medicine ,Mediator ,Protein Domains ,GTP-Binding Proteins ,medicine ,Humans ,Molecular Biology ,Transcription factor ,Research Articles ,Mutation ,Cell growth ,Chemistry ,Cell Biology ,Cell biology ,interferons ,030104 developmental biology ,Cytokine ,cell proliferation ,guanylate-binding proteins ,Hippo signaling ,030220 oncology & carcinogenesis ,Research Article ,HeLa Cells ,Transcription Factors - Abstract
Interferon-gamma (IFN-γ) is a pleiotropic cytokine that exerts important functions in inflammation, infectious diseases, and cancer. The large GTPase human guanylate-binding protein 1 (GBP-1) is among the most strongly IFN-γ-induced cellular proteins. Previously, it has been shown that GBP-1 mediates manifold cellular responses to IFN-γ including the inhibition of proliferation, spreading, migration, and invasion and through this exerts anti-tumorigenic activity. However, the mechanisms of GBP-1 anti-tumorigenic activities remain poorly understood. Here, we elucidated the molecular mechanism of the human GBP-1-mediated suppression of proliferation by demonstrating for the first time a cross-talk between the anti-tumorigenic IFN-γ and Hippo pathways. The α9-helix of GBP-1 was found to be sufficient to inhibit proliferation. Protein-binding and molecular modeling studies revealed that the α9-helix binds to the DNA-binding domain of the Hippo signaling transcription factor TEA domain protein (TEAD) mediated by the 376VDHLFQK382 sequence at the N-terminus of the GBP-1-α9-helix. Mutation of this sequence resulted in abrogation of both TEAD interaction and suppression of proliferation. Further on, the interaction caused inhibition of TEAD transcriptional activity associated with the down-regulation of TEAD-target genes. In agreement with these results, IFN-γ treatment of the cells also impaired TEAD activity, and this effect was abrogated by siRNA-mediated inhibition of GBP-1 expression. Altogether, this demonstrated that the α9-helix is the proliferation inhibitory domain of GBP-1, which acts independent of the GTPase activity through the inhibition of the Hippo transcription factor TEAD in mediating the anti-proliferative cell response to IFN-γ.
- Published
- 2018
40. The peptidyl-prolyl cis/trans isomerase Pin1 interacts with three early regulatory proteins of human cytomegalovirus
- Author
-
Jens Milbradt, Heinrich Sticht, Theresa Errerd, Christina Wangen, Marco Thomas, Manfred Marschall, Martin Schütz, Melissa Kießling, Luisa Rauschert, and Sabrina Wagner
- Subjects
Human cytomegalovirus ,Cancer Research ,Viral protein ,Immunoprecipitation ,viruses ,Cytomegalovirus ,medicine.disease_cause ,Virus Replication ,03 medical and health sciences ,Viral Proteins ,Virology ,medicine ,Humans ,Binding site ,Polymerase ,030304 developmental biology ,0303 health sciences ,biology ,030306 microbiology ,medicine.disease ,Cell biology ,NIMA-Interacting Peptidylprolyl Isomerase ,Infectious Diseases ,HEK293 Cells ,Cytomegalovirus Infections ,PIN1 ,biology.protein ,Nuclear lamina ,Lamin - Abstract
Human cytomegalovirus (HCMV) is a ubiquitous human pathogen of high clinical relevance. Despite intensive research of virus-host interaction, crucial details still remain unknown. In this study, the role of the cellular peptidyl-prolyl cis/trans isomerase Pin1 during HCMV infection was investigated. Pin1 is able to recognize phosphorylated serine/threonine-proline motifs and regulates the structural conformation, stability and function of its substrates. Concerning HCMV replication, our recent studies revealed that Pin1 plays an important role in viral nuclear egress by contributing to the depletion of the nuclear lamina at distinct sites through the cis/trans conversion of lamin proteins. Here, novel data illustrate the HCMV-induced upregulation of Pin1 including various cell types being permissive, semi-permissive or non-permissive for productive HCMV replication. Addressing the question of functional impact, Pin1 knock-out (KO) did not show a measurable effect on viral protein expression, at least when assessed by Western blot analysis. Applying highly sensitive methods of qPCR and plaque titration, a pharmacological inhibition of Pin1 activity, however, led to a significant decrease of viral genome equivalents and production of infectious virus, respectively. When focusing on the identification of viral proteins interacting with Pin1 by various coimmunoprecipitation (CoIP) settings, we obtained positive signals for (i) the core nuclear egress complex protein pUL50, (ii) the viral mRNA export factor pUL69 and (iii) the viral DNA polymerase processivity factor pUL44. Confocal immunofluorescence analysis focusing on partial colocalization between Pin1 and the coexpressed viral proteins pUL50, pUL69 or pUL44, respectively, was consistent with the CoIP experiments. Mapping experiments, using transient expression constructs for a series of truncated protein versions and specific replacement mutants, revealed a complex pattern of Pin1 interaction with these three early regulatory HCMV proteins. Data suggest a combination of different modes of Pin1 interactions, involving both classical phosphorylation-dependent Pin1 binding motifs and additional phosphorylation-independent binding sites. Combined, these results support the concept that Pin1 may play an important role in several stages of HCMV infection, thus determining viral replicative efficiency.
- Published
- 2019
41. CTCF variants in 39 individuals with a variable neurodevelopmental disorder broaden the mutational and clinical spectrum
- Author
-
Enrico D.H. Konrad, Niels Nardini, Almuth Caliebe, Inga Nagel, Dana Young, Gabriella Horvath, Stephanie L. Santoro, Christine Shuss, Alban Ziegler, Dominique Bonneau, Marlies Kempers, Rolph Pfundt, Eric Legius, Arjan Bouman, Kyra E. Stuurman, Katrin Õunap, Sander Pajusalu, Monica H. Wojcik, Georgia Vasileiou, Gwenaël Le Guyader, Hege M. Schnelle, Siren Berland, Evelien Zonneveld-Huijssoon, Simone Kersten, Aditi Gupta, Patrick R. Blackburn, Marissa S. Ellingson, Matthew J. Ferber, Radhika Dhamija, Eric W. Klee, Meriel McEntagart, Klaske D. Lichtenbelt, Amy Kenney, Samantha A. Vergano, Rami Abou Jamra, Konrad Platzer, Mary Ella Pierpont, Divya Khattar, Robert J. Hopkin, Richard J. Martin, Marjolijn C.J. Jongmans, Vivian Y. Chang, Julian A. Martinez-Agosto, Outi Kuismin, Mitja I. Kurki, Olli Pietiläinen, Aarno Palotie, Timothy J. Maarup, Diana S. Johnson, Katja Venborg Pedersen, Lone W. Laulund, Sally A. Lynch, Moira Blyth, Katrina Prescott, Natalie Canham, Rita Ibitoye, Eva H. Brilstra, Marwan Shinawi, Emily Fassi, Heinrich Sticht, Anne Gregor, Hilde Van Esch, Christiane Zweier, Graduate School, Clinical Genetics, Institute for Molecular Medicine Finland, Genomics of Neurological and Neuropsychiatric Disorders, University of Helsinki, Center for Population, Health and Society, Centre of Excellence in Complex Disease Genetics, and Aarno Palotie / Principal Investigator
- Subjects
Male ,CCCTC-Binding Factor ,Developmental Disabilities ,lnfectious Diseases and Global Health Radboud Institute for Molecular Life Sciences [Radboudumc 4] ,Transcriptome ,0302 clinical medicine ,Neurodevelopmental disorder ,Intellectual disability ,Drosophila Proteins ,Missense mutation ,TOOL ,Genetics(clinical) ,Child ,Genetics (clinical) ,Genetics & Heredity ,Genetics ,0303 health sciences ,biology ,GENE ONTOLOGY ,neurodevelopmental disorders ,1184 Genetics, developmental biology, physiology ,HUMANS ,Chromatin ,3. Good health ,DROSOPHILA ,Drosophila melanogaster ,intellectual disability ,LIBRARY ,Female ,INACTIVATION ,Life Sciences & Biomedicine ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,EXPRESSION ,DATABASE ,Mutation, Missense ,Article ,Young Adult ,03 medical and health sciences ,Exome Sequencing ,medicine ,Animals ,Gene ,030304 developmental biology ,Science & Technology ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Gene Expression Profiling ,biology.organism_classification ,medicine.disease ,CTCF ,Gene Expression Regulation ,DE-NOVO MUTATIONS ,Mutation ,030217 neurology & neurosurgery ,Transcription Factors ,chromatin organization - Abstract
PURPOSE: Pathogenic variants in the chromatin organizer CTCF were previously reported in seven individuals with a neurodevelopmental disorder (NDD). METHODS: Through international collaboration we collected data from 39 subjects with variants in CTCF. We performed transcriptome analysis on RNA from blood samples and utilized Drosophila melanogaster to investigate the impact of Ctcf dosage alteration on nervous system development and function. RESULTS: The individuals in our cohort carried 2 deletions, 8 likely gene-disruptive, 2 splice-site, and 20 different missense variants, most of them de novo. Two cases were familial. The associated phenotype was of variable severity extending from mild developmental delay or normal IQ to severe intellectual disability. Feeding difficulties and behavioral abnormalities were common, and variable other findings including growth restriction and cardiac defects were observed. RNA-sequencing in five individuals identified 3828 deregulated genes enriched for known NDD genes and biological processes such as transcriptional regulation. Ctcf dosage alteration in Drosophila resulted in impaired gross neurological functioning and learning and memory deficits. CONCLUSION: We significantly broaden the mutational and clinical spectrum ofCTCF-associated NDDs. Our data shed light onto the functional role of CTCF by identifying deregulated genes and show that Ctcf alterations result in nervous system defects in Drosophila. ispartof: GENETICS IN MEDICINE vol:21 issue:12 pages:2723-2733 ispartof: location:United States status: published
- Published
- 2019
42. The polynucleotide kinase 3′-phosphatase gene (PNKP) is involved in Charcot-Marie-Tooth disease (CMT2B2) previously related to MED25
- Author
-
André Reis, Martin Berghoff, Bernal Morera, Sixto Bogantes-Ledezma, Arif B. Ekici, Alejandro Leal, Corinna Berghoff, Michael Meisterernst, Steffen Uebe, Heinrich Sticht, and Christian Thiel
- Subjects
Adult ,Costa Rica ,Male ,Models, Molecular ,0301 basic medicine ,Microcephaly ,Ataxia ,DNA Mutational Analysis ,Mutation, Missense ,Locus (genetics) ,Biology ,Compound heterozygosity ,MED25 ,Polymorphism, Single Nucleotide ,CMT2B2 ,Consanguinity ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Charcot-Marie-Tooth Disease ,Genetics ,medicine ,Humans ,Missense mutation ,Family ,Genetic Predisposition to Disease ,PNKP ,Oculomotor apraxia ,Allele ,Genetics (clinical) ,Exome sequencing ,Mediator Complex ,CMT ,Middle Aged ,medicine.disease ,Pedigree ,Phosphotransferases (Alcohol Group Acceptor) ,DNA Repair Enzymes ,030104 developmental biology ,Amino Acid Substitution ,Case-Control Studies ,Original Article ,Female ,AOA4 ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
Charcot-Marie-Tooth disease (CMT) represents a heterogeneous group of hereditary peripheral neuropathies. We previously reported a CMT locus on chromosome 19q13.3 segregating with the disease in a large Costa Rican family with axonal neuropathy and autosomal recessive pattern of inheritance (CMT2B2). We proposed a homozygous missense variant in the Mediator complex 25 (MED25) gene as causative of the disease. Nevertheless, the fact that no other CMT individuals with MED25 variants were reported to date led us to reevaluate the original family. Using exome sequencing, we now identified a homozygous nonsense variant (p.Gln517ter) in the last exon of an adjacent gene, the polynucleotide kinase 3'-phosphatase (PNKP) gene. It encodes a DNA repair protein recently associated with recessive ataxia with oculomotor apraxia type 4 (AOA4) and microcephaly, seizures, and developmental delay (MCSZ). Subsequently, five unrelated Costa Rican CMT2 subjects initially identified as being heterozygous for the same MED25 variant were found to be also compound heterozygote for PNKP. All were heterozygous for the same variant found homozygous in the large family and a second one previously associated with ataxia (p.Thr408del). Detailed clinical reassessment of the initial family and the new individuals revealed in all an adult-onset slowly progressive CMT2 associated with signs of cerebellar dysfunction such as slurred speech and oculomotor involvement, but neither microcephaly, seizures, nor developmental delay. We propose that PKNP variants are the major causative variant for the CMT2 phenotype in these individuals and that the milder clinical manifestation is due to an allelic effect.
- Published
- 2018
43. Missense Variants in RHOBTB2 Cause a Developmental and Epileptic Encephalopathy in Humans, and Altered Levels Cause Neurological Defects in Drosophila
- Author
-
Holly Dubbs, Diana Johnson, Anna Fliedner, Jonas Straub, Anne Gregor, Enrico D.H. Konrad, Meredith Phillips, Alexander P.A. Stegmann, Christiane Zweier, Maaike Vreeburg, Avni Santani, Lacey Smith, Bryan L. Krock, Levinus A. Bok, Constance T. R. M. Stumpel, Annapurna Poduri, Ganka Douglas, Addie I. Nesbitt, Heinrich Sticht, Heather P. Crawford, Johanna Grüner, Xilma R. Ortiz-Gonzalez, Rebekah Jobling, Mohamad A. Mikati, Zöe Powis, Joost Nicolai, Megan T. Cho, Annick Toutain, RS: MHeNs - R1 - Cognitive Neuropsychiatry and Clinical Neuroscience, Klinische Neurowetenschappen, MUMC+: MA Med Staf Spec Neurologie (9), MUMC+: DA KG Lab Centraal Lab (9), RS: GROW - R4 - Reproductive and Perinatal Medicine, Klinische Genetica, and MUMC+: DA KG Polikliniek (9)
- Subjects
0301 basic medicine ,Male ,INTELLECTUAL DISABILITY ,Adolescent ,Gene Dosage ,Mutation, Missense ,AUTISM SPECTRUM DISORDERS ,GTPase ,Biology ,UBIQUITIN LIGASE COMPLEX ,RHO-GTPASES ,Article ,03 medical and health sciences ,Epilepsy ,GTP-Binding Proteins ,SENSITIVE PARALYTIC MUTANTS ,Genetics ,medicine ,Missense mutation ,Animals ,Drosophila Proteins ,Humans ,SPINE MORPHOGENESIS ,BREAST-CANCER ,TUMOR-SUPPRESSOR ,Amino Acid Sequence ,RhoBTB ,Child ,Genetics (clinical) ,Behavior, Animal ,Tumor Suppressor Proteins ,HEK 293 cells ,Dendrites ,medicine.disease ,Phenotype ,Cell biology ,030104 developmental biology ,Drosophila melanogaster ,HEK293 Cells ,DE-NOVO MUTATIONS ,Ubiquitin ligase complex ,Child, Preschool ,Synaptic plasticity ,Synapses ,Female ,MENTAL-RETARDATION - Abstract
Although the role of typical Rho GTPases and other Rho-linked proteins in synaptic plasticity and cognitive function and dysfunction is widely acknowledged, the role of atypical Rho GTPases (such as RHOBTB2) in neurodevelopment has barely been characterized. We have now identified de novo missense variants clustering in the BTB-domain-encoding region of RHOBTB2 in ten individuals with a similar phenotype, including early-onset epilepsy, severe intellectual disability, postnatal microcephaly, and movement disorders. Three of the variants were recurrent. Upon transfection of HEK293 cells, we found that mutant RHOBTB2 was more abundant than the wild-type, most likely because of impaired degradation in the proteasome. Similarly, elevated amounts of the Drosophila ortholog RhoBTB in vivo were associated with seizure susceptibility and severe locomotor defects. Knockdown of RhoBTB in the Drosophila dendritic arborization neurons resulted in a decreased number of dendrites, thus suggesting a role of RhoBTB in dendritic development. We have established missense variants in the BTB-domain-encoding region of RHOBTB2 as causative for a developmental and epileptic encephalopathy and have elucidated the role of atypical Rho GTPase RhoBTB in Drosophila neurological function and possibly dendrite development.
- Published
- 2018
44. Evidence for genetic overlap between adult onset Still’s disease and hereditary periodic fever syndromes
- Author
-
Norbert Blank, Sabine Löhr, R. Sighart, Ulrike Hüffmeier, André Reis, Heinrich Sticht, Jürgen Rech, and A. Hueber
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Pathology ,Pediatrics ,Adolescent ,Immunology ,Familial Mediterranean fever ,Disease ,Pyrin domain ,Pathogenesis ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Rheumatology ,Internal medicine ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Humans ,Immunology and Allergy ,Genetic Predisposition to Disease ,Young adult ,Child ,Aged ,030203 arthritis & rheumatology ,Autoimmune disease ,business.industry ,Hereditary Autoinflammatory Diseases ,Middle Aged ,Pyrin ,medicine.disease ,Phosphotransferases (Alcohol Group Acceptor) ,030104 developmental biology ,Receptors, Tumor Necrosis Factor, Type I ,TNF receptor associated periodic syndrome ,Mutation ,Female ,business ,Still's Disease, Adult-Onset - Abstract
Adult onset Still's disease (AOSD) is a severe, autoimmune disease that can be challenging to treat with conventional therapeutics and biologicals in a considerable number of cases. Therefore, there is a high need to understand its pathogenesis better. As major clinical symptoms overlap between AOSD and hereditary periodic fever syndromes (HPFS), we analysed four known HPFS genes in AOSD.We performed Sanger sequencing and quantitative analysis of all coding regions of MEFV, TNFRSF1A, MVK and NLRP3 in 40 AOSD patients. All rare coding variants (n = 6) were evaluated for several aspects to classify them as benign to pathogenic variants. Statistical analysis was performed to analyse whether variants classified as (likely) pathogenic were associated with AOSD.We identified three rare variants in MEFV, one previously not described. Association to the three likely pathogenic MEFV variants was significant (pOur results indicate TNFRSF1A as a relevant gene in AOSD, especially in patients with a more challenging course of disease, while causal variants remain to be identified in the majority of patients.
- Published
- 2017
45. Dynamic regulatory interaction between cytomegalovirus major tegument protein pp65 and protein kinase pUL97 in intracellular compartments, dense bodies and virions
- Author
-
Eileen Socher, Bodo Plachter, Heinrich Sticht, Patrick König, Stefan Tenzer, Manfred Marschall, Nicole Büscher, and Mirjam Steingruber
- Subjects
0301 basic medicine ,Human cytomegalovirus ,viruses ,DNA Mutational Analysis ,Mutant ,Cytomegalovirus ,Biology ,Virus Replication ,Viral Matrix Proteins ,Viral Proteins ,03 medical and health sciences ,Viral entry ,Virology ,Protein Interaction Mapping ,Viral structural protein ,medicine ,Humans ,Protein kinase A ,virus diseases ,Viral tegument ,biochemical phenomena, metabolism, and nutrition ,Phosphoproteins ,medicine.disease ,Cell biology ,030104 developmental biology ,Viral replication ,Phosphoprotein - Abstract
Human cytomegalovirus (HCMV) is a ubiquitous pathogen of considerable clinical importance. Understanding the processes that are important for viral replication is essential for the development of therapeutic strategies against HCMV infection. The HCMV-encoded protein kinase pUL97 is an important multifunctional regulator of viral replication. Several viral and cellular proteins are phosphorylated by pUL97. The phosphoprotein pp65 is one important substrate of pUL97. It is the most abundant tegument protein of HCMV virions, mediating the upload of other virion constituents and contributing to particle integrity. Further to that, it interferes with host innate immune defences, thereby enabling efficient viral replication. By applying different approaches, we characterized the pp65-pUL97 interaction in various compartments. Specifically, the pUL97 interaction domain of pp65 was defined (282-415). A putative cyclin bridge that enhances pUL97-pp65 interaction was identified. The impact of pUL97 mutation on virion and dense body morphogenesis was addressed using pUL97 mutant viruses. Alterations in the proteome of viral particles were seen, especially with mutant viruses expressing cytoplasmic variants of pUL97. On the basis of these data we postulate a so far poorly recognized functional relationship between pp65 and pUL97, and present a refined model of pp65-pUL97 interaction.
- Published
- 2017
46. Clinical relevance of systematic phenotyping and exome sequencing in patients with short stature
- Author
-
Cornelia Kraus, Christiane Zweier, Rami Abou Jamra, Patricia Klinger, Sarah Schuhmann, Nadine N. Hauer, Steffen Uebe, Heinrich Sticht, Christian Thiel, Christian Büttner, Helmuth-Günther Dörr, Arif B. Ekici, Karen E. Heath, Fulvia Ferrazzi, Bernt Popp, Antje Wiesener, Alfonso Hisado-Oliva, Erdmute Kunstmann, Anita Rauch, Martin Zenker, Eva Schoeller, Dagmar Wieczorek, André Reis, and Udo Trautmann
- Subjects
0301 basic medicine ,Male ,Heterozygote ,Genetic counseling ,Medizin ,030209 endocrinology & metabolism ,Bioinformatics ,medicine.disease_cause ,Short stature ,growth ,phenotypic spectrum ,Whole Exome Sequencing ,03 medical and health sciences ,0302 clinical medicine ,Exome Sequencing ,medicine ,Humans ,Clinical significance ,Original Research Article ,whole-exome sequencing ,Genetic Testing ,Allele ,Genetics (clinical) ,Exome sequencing ,Genetic testing ,Mutation ,medicine.diagnostic_test ,business.industry ,High-Throughput Nucleotide Sequencing ,Sequence Analysis, DNA ,medicine.disease ,Body Height ,Pedigree ,030104 developmental biology ,Phenotype ,short stature ,skeletal dysplasia ,Dysplasia ,Female ,medicine.symptom ,business - Abstract
Purpose Short stature is a common condition of great concern to patients and their families. Mostly genetic in origin, the underlying cause often remains elusive due to clinical and genetic heterogeneity. Methods We systematically phenotyped 565 patients where common nongenetic causes of short stature were excluded, selected 200 representative patients for whole-exome sequencing, and analyzed the identified variants for pathogenicity and the affected genes regarding their functional relevance for growth. Results By standard targeted diagnostic and phenotype assessment, we identified a known disease cause in only 13.6% of the 565 patients. Whole-exome sequencing in 200 patients identified additional mutations in known short-stature genes in 16.5% of these patients who manifested only part of the symptomatology. In 15.5% of the 200 patients our findings were of significant clinical relevance. Heterozygous carriers of recessive skeletal dysplasia alleles represented 3.5% of the cases. Conclusion A combined approach of systematic phenotyping, targeted genetic testing, and whole-exome sequencing allows the identification of the underlying cause of short stature in at least 33% of cases, enabling physicians to improve diagnosis, treatment, and genetic counseling. Exome sequencing significantly increases the diagnostic yield and consequently care in patients with short stature.
- Published
- 2017
47. Hypomorphic Pathogenic Variants in TAF13 Are Associated with Autosomal-Recessive Intellectual Disability and Microcephaly
- Author
-
Roberto Colombo, Franz Wolfgang Hirsch, Natalie Wohlfahrt, Irwin Davidson, André Reis, Rami Abou Jamra, Johannes Schumacher, Rebecca Buchert, Igor Martianov, Hasan Tawamie, Luigi Janiri, Steffen Uebe, Heinrich Sticht, Gabrielle Mengus, Fulvia Ferrazzi, MENGUS, Gabrielle, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), University of Tübingen, Università cattolica del Sacro Cuore = Catholic University of the Sacred Heart [Roma] (Unicatt), University Hospital Leipzig, and University of Bonn
- Subjects
Male ,0301 basic medicine ,Microcephaly ,Transcription, Genetic ,Protein Conformation ,[SDV]Life Sciences [q-bio] ,RNA polymerase II ,Biology ,03 medical and health sciences ,pathogenic variant ,0302 clinical medicine ,Report ,Gene expression ,Genetics ,medicine ,Humans ,Immunoprecipitation ,Promoter Regions, Genetic ,transcription factor IID ,Gene ,Alleles ,TAF13 ,Genetics (clinical) ,Exome sequencing ,TATA-Binding Protein Associated Factors ,Gene knockdown ,Genetic Variation ,Infant ,Promoter ,medicine.disease ,Pedigree ,[SDV] Life Sciences [q-bio] ,030104 developmental biology ,intellectual disability ,biology.protein ,Female ,Transcription Factor TFIID ,RNA-seq ,Transcription factor II D ,exome sequencing ,030217 neurology & neurosurgery - Abstract
International audience; In two independent consanguineous families each with two children affected by mild intellectual disability and microcephaly, we identified two homozygous missense variants (c.119T>A [p.Met40Lys] and c.92T>A [p.Leu31His]) in TATA-box-binding-protein-associated factor 13 (TAF13). Molecular modeling suggested a pathogenic effect of both variants through disruption of the interaction between TAF13 and TAF11. These two proteins form a histone-like heterodimer that is essential for their recruitment into the general RNA polymerase II transcription factor IID (TFIID) complex. Co-immunoprecipitation in HeLa cells transfected with plasmids encoding TAF11 and TAF13 revealed that both variants indeed impaired formation of the TAF13-TAF11 heterodimer, thus confirming the protein modeling analysis. To further understand the functional role of TAF13, we performed RNA sequencing of neuroblastoma cell lines upon TAF13 knockdown. The transcriptional profile showed significant deregulation of gene expression patterns with an emphasis on genes related to neuronal and skeletal functions and those containing E-box motives in their promoters. Here, we expand the spectrum of TAF-associated phenotypes and highlight the importance of TAF13 in neuronal functions.
- Published
- 2017
48. Pathogenic WDFY3 variants cause neurodevelopmental disorders and opposing effects on brain size
- Author
-
Meron Azage, David B. Everman, Brooke T. Smith, Steffen Syrbe, Stephen W. Scherer, Jennifer Keller-Ramey, Gregory M. Cooper, Verónica Martínez-Cerdeño, Susan M. Hiatt, Mathew J Wallis, Dmitriy Niyazov, Benjamin Büttner, Rami Abou Jamra, Ryan K. C. Yuen, Johannes R. Lemke, Natasha J Brown, Amber Begtrup, Richard E. Person, Barbara Kellam, Chloé Quélin, Heinrich Sticht, Laurence J. Walsh, Angelo Harlan De Crescenzo, Konstantinos Zarbalis, Jonathan B. Strober, Susan Walker, Alexios A Panoutsopoulos, Shuxi Liu, Diana Le Duc, Urania Kotzaeridou, Michael S. Hildebrand, Michael C. Pride, Eleonora Napoli, Jacqueline N. Crawley, Francis Jeshira Reynoso Santos, Katelyn Payne, Renee Bend, Sandra Yang, Megan T. Cho, Evdokia Anagnostou, Cecilia R Giulivi, Rhonda E. Schnur, Lori Orosco, Andreas Ziegler, Jan H Doering, Christèle Dubourg, Jill L. Silverman, Universität Leipzig [Leipzig], University of California [Davis] (UC Davis), University of California, GeneDx [Gaithersburg, MD, USA], Institut de Génétique et Développement de Rennes (IGDR), Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique )-Centre National de la Recherche Scientifique (CNRS)-Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES), Service de génétique clinique [Rennes], Université de Rennes 1 (UR1), Université de Rennes (UNIV-RENNES)-Université de Rennes (UNIV-RENNES)-CHU Pontchaillou [Rennes]-hôpital Sud, Shriners Hospitals for Children, U54 HD079125, National Institute of Child Health and Human Development, R21MH115347, National Institute of Mental Health, 286567, Simons Foundation, Nancy Lurie Marks Family Foundation, UM1HG007301, National Human Genome Research Institute, Clinician Scientist Programm, Medizinische Fakultät der Universität Leipzig, Shriners Hospitals for Children Postdoctoral Fellowship, The MIND Institute IDDRC, Universität Leipzig, University of California (UC), Université de Rennes (UR)-Centre National de la Recherche Scientifique (CNRS)-Structure Fédérative de Recherche en Biologie et Santé de Rennes ( Biosit : Biologie - Santé - Innovation Technologique ), and Université de Rennes (UR)-CHU Pontchaillou [Rennes]-hôpital Sud
- Subjects
0301 basic medicine ,Proband ,Male ,Microcephaly ,Secondary ,[SDV]Life Sciences [q-bio] ,Autophagy-Related Proteins ,Medical and Health Sciences ,Transgenic ,Mice ,0302 clinical medicine ,Intellectual disability ,2.1 Biological and endogenous factors ,Aetiology ,Child ,Exome ,ComputingMilieux_MISCELLANEOUS ,Pediatric ,Brain ,Adaptor Proteins ,Organ Size ,Mental Health ,WDFY3 ,Autism spectrum disorder ,intellectual disability ,brain size ,Neurological ,Female ,medicine.symptom ,Haploinsufficiency ,Biotechnology ,Protein Structure ,Adolescent ,Intellectual and Developmental Disabilities (IDD) ,Biology ,neurodevelopmental delay ,03 medical and health sciences ,Clinical Research ,medicine ,Genetics ,Animals ,Humans ,Preschool ,Neurology & Neurosurgery ,Psychology and Cognitive Sciences ,Macrocephaly ,Signal Transducing ,Neurosciences ,Genetic Variation ,medicine.disease ,Stem Cell Research ,Associative learning ,Brain Disorders ,030104 developmental biology ,Neurodevelopmental Disorders ,Neurology (clinical) ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The underpinnings of mild to moderate neurodevelopmental delay remain elusive, often leading to late diagnosis and interventions. Here, we present data on exome and genome sequencing as well as array analysis of 13 individuals that point to pathogenic, heterozygous, mostly de novo variants in WDFY3 (significant de novo enrichment P = 0.003) as a monogenic cause of mild and non-specific neurodevelopmental delay. Nine variants were protein-truncating and four missense. Overlapping symptoms included neurodevelopmental delay, intellectual disability, macrocephaly, and psychiatric disorders (autism spectrum disorders/attention deficit hyperactivity disorder). One proband presented with an opposing phenotype of microcephaly and the only missense-variant located in the PH-domain of WDFY3. Findings of this case are supported by previously published data, demonstrating that pathogenic PH-domain variants can lead to microcephaly via canonical Wnt-pathway upregulation. In a separate study, we reported that the autophagy scaffolding protein WDFY3 is required for cerebral cortical size regulation in mice, by controlling proper division of neural progenitors. Here, we show that proliferating cortical neural progenitors of human embryonic brains highly express WDFY3, further supporting a role for this molecule in the regulation of prenatal neurogenesis. We present data on Wnt-pathway dysregulation in Wdfy3-haploinsufficient mice, which display macrocephaly and deficits in motor coordination and associative learning, recapitulating the human phenotype. Consequently, we propose that in humans WDFY3 loss-of-function variants lead to macrocephaly via downregulation of the Wnt pathway. In summary, we present WDFY3 as a novel gene linked to mild to moderate neurodevelopmental delay and intellectual disability and conclude that variants putatively causing haploinsufficiency lead to macrocephaly, while an opposing pathomechanism due to variants in the PH-domain of WDFY3 leads to microcephaly.
- Published
- 2019
49. A Metadynamics-Based Protocol for the Determination of GPCR-Ligand Binding Modes
- Author
-
Christian A. Söldner, Heinrich Sticht, and Anselm H. C. Horn
- Subjects
0301 basic medicine ,Agonist ,medicine.drug_class ,Molecular Conformation ,Computational biology ,Molecular Dynamics Simulation ,Ligands ,01 natural sciences ,Catalysis ,Article ,Receptors, G-Protein-Coupled ,Inorganic Chemistry ,lcsh:Chemistry ,03 medical and health sciences ,Molecular dynamics ,chemistry.chemical_compound ,Structure-Activity Relationship ,medicine ,receptor-ligand interactions ,ddc:610 ,Physical and Theoretical Chemistry ,Binding site ,Molecular Biology ,Conformational ensembles ,lcsh:QH301-705.5 ,Spectroscopy ,G protein-coupled receptor ,Binding Sites ,metadynamics simulations ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,ligand binding modes ,Metadynamics ,General Medicine ,molecular dynamics simulations ,Ligand (biochemistry) ,0104 chemical sciences ,Computer Science Applications ,Molecular Docking Simulation ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Alprenolol ,G protein-coupled receptors (GPCRs) ,Algorithms ,Protein Binding ,clustering - Abstract
G protein-coupled receptors (GPCRs) are a main drug target and therefore a hot topic in pharmaceutical research. One important prerequisite to understand how a certain ligand affects a GPCR is precise knowledge about its binding mode and the specific underlying interactions. If no crystal structure of the respective complex is available, computational methods can be used to deduce the binding site. One of them are metadynamics simulations which have the advantage of an enhanced sampling compared to conventional molecular dynamics simulations. However, the enhanced sampling of higher-energy states hampers identification of the preferred binding mode. Here, we present a novel protocol based on clustering of multiple walker metadynamics simulations which allows identifying the preferential binding mode from such conformational ensembles. We tested this strategy for three different model systems namely the histamine H1 receptor in combination with its physiological ligand histamine, as well as the &beta, 2 adrenoceptor with its agonist adrenaline and its antagonist alprenolol. For all three systems, the proposed protocol was able to reproduce the correct binding mode known from the literature suggesting that the approach can more generally be applied to the prediction of GPCR ligand binding in future.
- Published
- 2019
50. Further corroboration of distinct functional features in SCN2A variants causing intellectual disability or epileptic phenotypes
- Author
-
Mario A. Acuña, Lucia Abela, Hanns Ulrich Zeilhofer, Markus Zweier, Yushi Inoue, Anaïs Begemann, Anita Rauch, Annette Hackenberg, Ruxandra Bachmann-Gagescu, Kazuhiro Yamakawa, Katharina Steindl, Judith Kroell-Seger, Barbara Plecko, Alessandra Baumer, Reza Asadollahi, Marie Vincent, Heinrich Sticht, and University of Zurich
- Subjects
0301 basic medicine ,10039 Institute of Medical Genetics ,Intellectual disability ,10050 Institute of Pharmacology and Toxicology ,Gating ,Epilepsy ,0302 clinical medicine ,Channelopathy ,Medizinische Fakultät ,Missense mutation ,lcsh:QD415-436 ,Child ,Genetics (clinical) ,Genetics ,NAV1.2 Voltage-Gated Sodium Channel ,Epileptic encephalopathy ,Phenotype ,3. Good health ,Electrophysiology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Research Article ,2716 Genetics (clinical) ,Adolescent ,SCN2A ,Nav1.2 ,Patch-clamp ,Structural modelling ,610 Medicine & health ,Biology ,lcsh:Biochemistry ,03 medical and health sciences ,Young Adult ,1311 Genetics ,1312 Molecular Biology ,medicine ,Humans ,ddc:610 ,Molecular Biology ,QH426 ,Genetic Association Studies ,Sodium channel ,lcsh:RM1-950 ,medicine.disease ,Epilepsy, Benign Neonatal ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,HEK293 Cells ,10036 Medical Clinic ,1313 Molecular Medicine ,570 Life sciences ,biology ,Autism ,Epileptic Syndromes - Abstract
Background Deleterious variants in the voltage-gated sodium channel type 2 (Nav1.2) lead to a broad spectrum of phenotypes ranging from benign familial neonatal-infantile epilepsy (BFNIE), severe developmental and epileptic encephalopathy (DEE) and intellectual disability (ID) to autism spectrum disorders (ASD). Yet, the underlying mechanisms are still incompletely understood. Methods To further elucidate the genotype-phenotype correlation of SCN2A variants we investigated the functional effects of six variants representing the phenotypic spectrum by whole-cell patch-clamp studies in transfected HEK293T cells and in-silico structural modeling. Results The two variants p.L1342P and p.E1803G detected in patients with early onset epileptic encephalopathy (EE) showed profound and complex changes in channel gating, whereas the BFNIE variant p.L1563V exhibited only a small gain of channel function. The three variants identified in ID patients without seizures, p.R937C, p.L611Vfs*35 and p.W1716*, did not produce measurable currents. Homology modeling of the missense variants predicted structural impairments consistent with the electrophysiological findings. Conclusions Our findings support the hypothesis that complete loss-of-function variants lead to ID without seizures, small gain-of-function variants cause BFNIE and EE variants exhibit variable but profound Nav1.2 gating changes. Moreover, structural modeling was able to predict the severity of the variant impact, supporting a potential role of structural modeling as a prognostic tool. Our study on the functional consequences of SCN2A variants causing the distinct phenotypes of EE, BFNIE and ID contributes to the elucidation of mechanisms underlying the broad phenotypic variability reported for SCN2A variants. Electronic supplementary material The online version of this article (10.1186/s10020-019-0073-6) contains supplementary material, which is available to authorized users.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.