Network


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

Hotspot


Dive into the research topics where Michaela Thoenes is active.

Publication


Featured researches published by Michaela Thoenes.


Orphanet Journal of Rare Diseases | 2014

The supposed tumor suppressor gene WWOX is mutated in an early lethal microcephaly syndrome with epilepsy, growth retardation and retinal degeneration

Ghada Mh Abdel-Salam; Michaela Thoenes; Hanan H. Afifi; Friederike Körber; Daniel Swan; Hanno J. Bolz

BackgroundWWOX, encoding WW domain-containing oxidoreductase, spans FRA16D, the second most common chromosomal fragile site frequently altered in cancers. It is therefore considered a tumor suppressor gene, but its direct implication in cancerogenesis remains controversial.Methods and resultsBy whole-exome sequencing, we identified a homozygous WWOX nonsense mutation, p.Arg54*, in a girl from a consanguineous family with a severe syndrome of growth retardation, microcephaly, epileptic seizures, retinopathy and early death, a phenotype highly similar to the abormalities reported in lde/lde rats with a spontaneous functional null mutation of Wwox. As in rats, no tumors were observed in the patient or heterozygous mutation carriers.ConclusionsOur finding, a homozygous loss-of-function germline mutation in WWOX in a patient with a lethal autosomal recessive syndrome, supports an alternative role of WWOX and indicates its importance for human viability.


Human Mutation | 2016

Mutation Update for Kabuki Syndrome Genes KMT2D and KDM6A and Further Delineation of X-Linked Kabuki Syndrome Subtype 2.

Nina Bögershausen; Vincent Gatinois; Vera Riehmer; Hülya Kayserili; Jutta Becker; Michaela Thoenes; Pelin Ozlem Simsek-Kiper; Mouna Barat-Houari; Nursel Elcioglu; Dagmar Wieczorek; Sigrid Tinschert; Guillaume Sarrabay; Tim M. Strom; Aurelie Fabre; Gareth Baynam; Elodie Sanchez; Gudrun Nürnberg; Umut Altunoglu; Yline Capri; Bertrand Isidor; Didier Lacombe; Carole Corsini; Valérie Cormier-Daire; Damien Sanlaville; Fabienne Giuliano; Kim-Hanh Le Quan Sang; Honorine Kayirangwa; Peter Nürnberg; Thomas Meitinger; Koray Boduroglu

Kabuki syndrome (KS) is a rare but recognizable condition that consists of a characteristic face, short stature, various organ malformations, and a variable degree of intellectual disability. Mutations in KMT2D have been identified as the main cause for KS, whereas mutations in KDM6A are a much less frequent cause. Here, we report a mutation screening in a case series of 347 unpublished patients, in which we identified 12 novel KDM6A mutations (KS type 2) and 208 mutations in KMT2D (KS type 1), 132 of them novel. Two of the KDM6A mutations were maternally inherited and nine were shown to be de novo. We give an up‐to‐date overview of all published mutations for the two KS genes and point out possible mutation hot spots and strategies for molecular genetic testing. We also report the clinical details for 11 patients with KS type 2, summarize the published clinical information, specifically with a focus on the less well‐defined X‐linked KS type 2, and comment on phenotype–genotype correlations as well as sex‐specific phenotypic differences. Finally, we also discuss a possible role of KDM6A in Kabuki‐like Turner syndrome and report a mutation screening of KDM6C (UTY) in male KS patients.


Human Mutation | 2014

Mutation of POC1B in a Severe Syndromic Retinal Ciliopathy

Bodo B. Beck; Jennifer B. Phillips; Malte P. Bartram; Jeremy Wegner; Michaela Thoenes; Andrea Pannes; Josephina Sampson; Raoul Heller; Heike Göbel; Friederike Koerber; Antje Neugebauer; Andrea M. Hedergott; Gudrun Nürnberg; Peter Nürnberg; Holger Thiele; Janine Altmüller; Mohammad R. Toliat; Simon Staubach; Kym M. Boycott; Enza Maria Valente; Andreas R. Janecke; Tobias Eisenberger; Carsten Bergmann; Lars Tebbe; Yang Wang; Yun-Dong Wu; Andrew M. Fry; Monte Westerfield; Uwe Wolfrum; Hanno J. Bolz

We describe a consanguineous Iraqi family with Leber congenital amaurosis (LCA), Joubert syndrome (JBTS), and polycystic kidney disease (PKD). Targeted next‐generation sequencing for excluding mutations in known LCA and JBTS genes, homozygosity mapping, and whole‐exome sequencing identified a homozygous missense variant, c.317G>C (p.Arg106Pro), in POC1B, a gene essential for ciliogenesis, basal body, and centrosome integrity. In silico modeling suggested a requirement of p.Arg106 for the formation of the third WD40 repeat and a protein interaction interface. In human and mouse retina, POC1B localized to the basal body and centriole adjacent to the connecting cilium of photoreceptors and in synapses of the outer plexiform layer. Knockdown of Poc1b in zebrafish caused cystic kidneys and retinal degeneration with shortened and reduced photoreceptor connecting cilia, compatible with the human syndromic ciliopathy. A recent study describes homozygosity for p.Arg106ProPOC1B in a family with nonsyndromic cone‐rod dystrophy. The phenotype associated with homozygous p.Arg106ProPOC1B may thus be highly variable, analogous to homozygous p.Leu710Ser in WDR19 causing either isolated retinitis pigmentosa or Jeune syndrome. Our study indicates that POC1B is required for retinal integrity, and we propose POC1B mutations as a probable cause for JBTS with severe PKD.


eLife | 2015

TALPID3 controls centrosome and cell polarity and the human ortholog KIAA0586 is mutated in Joubert syndrome (JBTS23)

Louise Stephen; Hasan Tawamie; Gemma M. Davis; Lars Tebbe; Peter Nürnberg; Gudrun Nürnberg; Holger Thiele; Michaela Thoenes; Eugen Boltshauser; Steffen Uebe; Oliver Rompel; André Reis; Arif B. Ekici; Lynn McTeir; Amy M Fraser; Emma A. Hall; Pleasantine Mill; Nicolas Daudet; Courtney E. Cross; Uwe Wolfrum; Rami Abou Jamra; Megan Davey; Hanno J. Bolz

Joubert syndrome (JBTS) is a severe recessive neurodevelopmental ciliopathy which can affect several organ systems. Mutations in known JBTS genes account for approximately half of the cases. By homozygosity mapping and whole-exome sequencing, we identified a novel locus, JBTS23, with a homozygous splice site mutation in KIAA0586 (alias TALPID3), a known lethal ciliopathy locus in model organisms. Truncating KIAA0586 mutations were identified in two additional patients with JBTS. One mutation, c.428delG (p.Arg143Lysfs*4), is unexpectedly common in the general population and may be a major contributor to JBTS. We demonstrate KIAA0586 protein localization at the basal body in human and mouse photoreceptors, as is common for JBTS proteins, and also in pericentriolar locations. We show that loss of TALPID3 (KIAA0586) function in animal models causes abnormal tissue polarity, centrosome length and orientation, and centriolar satellites. We propose that JBTS and other ciliopathies may in part result from cell polarity defects. DOI: http://dx.doi.org/10.7554/eLife.08077.001


European Journal of Human Genetics | 2014

Autosomal dominant SCA5 and autosomal recessive infantile SCA are allelic conditions resulting from SPTBN2 mutations

Solaf M. Elsayed; Raoul Heller; Michaela Thoenes; Maha S. Zaki; Daniel Swan; Ezzat Elsobky; Christine Zühlke; Inga Ebermann; Gudrun Nürnberg; Peter Nürnberg; Hanno J. Bolz

Although many genes have been identified for the autosomal recessive cerebellar ataxias (ARCAs), several patients are unlinked to the respective loci, suggesting further genetic heterogeneity. We combined homozygosity mapping and exome sequencing in a consanguineous Egyptian family with congenital ARCA, mental retardation and pyramidal signs. A homozygous 5-bp deletion in SPTBN2, the gene whose in-frame mutations cause autosomal dominant spinocerebellar ataxia type 5, was shown to segregate with ataxia in the family. Our findings are compatible with the concept of truncating SPTBN2 mutations acting recessively, which is supported by disease expression in homozygous, but not heterozygous, knockout mice, ataxia in Beagle dogs with a homozygous frameshift mutation and, very recently, a homozygous SPTBN2 nonsense mutation underlying infantile ataxia and psychomotor delay in a human family. As there was no evidence for mutations in 23 additional consanguineous families, SPTBN2-related ARCA is probably rare.


Orphanet Journal of Rare Diseases | 2015

OSBPL2 encodes a protein of inner and outer hair cell stereocilia and is mutated in autosomal dominant hearing loss (DFNA67).

Michaela Thoenes; Ulrike Zimmermann; Inga Ebermann; Martin Ptok; Morag A. Lewis; Holger Thiele; Susanne Morlot; Markus Hess; Andreas Gal; Tobias Eisenberger; Carsten Bergmann; Gudrun Nürnberg; Peter Nürnberg; Karen P. Steel; Marlies Knipper; Hanno J. Bolz

BackgroundEarly-onset hearing loss is mostly of genetic origin. The complexity of the hearing process is reflected by its extensive genetic heterogeneity, with probably many causative genes remaining to be identified. Here, we aimed at identifying the genetic basis for autosomal dominant non-syndromic hearing loss (ADNSHL) in a large German family.MethodsA panel of 66 known deafness genes was analyzed for mutations by next-generation sequencing (NGS) in the index patient. We then conducted genome-wide linkage analysis, and whole-exome sequencing was carried out with samples of two patients. Expression of Osbpl2 in the mouse cochlea was determined by immunohistochemistry. Because Osbpl2 has been proposed as a target of miR-96, we investigated homozygous Mir96 mutant mice for its upregulation.ResultsOnset of hearing loss in the investigated ADNSHL family is in childhood, initially affecting the high frequencies and progressing to profound deafness in adulthood. However, there is considerable intrafamilial variability. We mapped a novel ADNSHL locus, DFNA67, to chromosome 20q13.2-q13.33, and subsequently identified a co-segregating heterozygous frameshift mutation, c.141_142delTG (p.Arg50Alafs*103), in OSBPL2, encoding a protein known to interact with the DFNA1 protein, DIAPH1. In mice, Osbpl2 was prominently expressed in stereocilia of cochlear outer and inner hair cells. We found no significant Osbpl2 upregulation at the mRNA level in homozygous Mir96 mutant mice.ConclusionThe function of OSBPL2 in the hearing process remains to be determined. Our study and the recent description of another frameshift mutation in a Chinese ADNSHL family identify OSBPL2 as a novel gene for progressive deafness.


Human Mutation | 2016

PEX6 is Expressed in Photoreceptor Cilia and Mutated in Deafblindness with Enamel Dysplasia and Microcephaly

Maha S. Zaki; Raoul Heller; Michaela Thoenes; Gudrun Nürnberg; Gabi Stern-Schneider; Peter Nürnberg; Srikanth Karnati; Daniel Swan; Ekram Fateen; Kerstin Nagel-Wolfrum; Mostafa I. Mostafa; Holger Thiele; Uwe Wolfrum; Eveline Baumgart-Vogt; Hanno J. Bolz

Deafblindness is part of several genetic disorders. We investigated a consanguineous Egyptian family with two siblings affected by congenital hearing loss and retinal degeneration, initially diagnosed as Usher syndrome type 1. At teenage, severe enamel dysplasia, developmental delay, and microcephaly became apparent. Genome‐wide homozygosity mapping and whole‐exome sequencing detected a homozygous missense mutation, c.1238G>T (p.Gly413Val), affecting a highly conserved residue of peroxisomal biogenesis factor 6, PEX6. Biochemical profiling of the siblings revealed abnormal and borderline plasma phytanic acid concentration, and cerebral imaging revealed white matter disease in both. We show that Pex6 localizes to the apical extensions of secretory ameloblasts and differentiated odontoblasts at early stages of dentin synthesis in mice, and to cilia of retinal photoreceptor cells. We propose PEX6, and possibly other peroxisomal genes, as candidate for the rare cooccurrence of deafblindness and enamel dysplasia. Our study for the first time links peroxisome biogenesis disorders to retinal ciliopathies.


Frontiers in Genetics | 2017

Recurrent and Prolonged Infections in a Child with a Homozygous IFIH1 Nonsense Mutation

Maha S. Zaki; Michaela Thoenes; Peter Nürnberg; Rolf Kaiser; Raoul Heller; Hanno J. Bolz

In an Egyptian girl born to consanguineous parents, whole-exome sequencing (WES) identified a homozygous mutation in PHGDH, c.1273G>A (p.Val425Met), indicating 3-phosphoglycerate dehydrogenase deficiency. This diagnosis was compatible with the patient’s microcephaly, severe psychomotor retardation, seizures and cataracts. However, she additionally suffered from recurrent (at least monthly) episodes of prolonged and severe chest infections requiring hospitalization, suggesting a secondary, predisposing and potentially Mendelian, condition. A local reactivation of an EBV infection in the respiratory tract was detected after a recent chest infection, likely representing an opportunistic infection based on a compromised immune system. Further inspection of WES data revealed a homozygous nonsense mutation, c.2665A>T (p.Lys889∗), in IFIH1, encoding MDA5. MDA5 detects long viral double-stranded RNA that is generated during replication of picorna viruses, and thereby activates the type I interferon signaling pathway. The results of Western blot analysis of protein from cultured fibroblasts of the patient indicates absence of wild type MDA5/IFIH1, compatible with NMD. We propose that, analogous to the severe course of primary influenza infection due to biallelic deficiency of a downstream effector, IRF7, homozygous loss of IFIH1 defines a novel Mendelian immunodeficiency disorder that increases susceptibility to severe viral infections. This is contrasted to heterozygous gain-of-function IFIH1 mutations in autoimmune diseases. Our findings highlight the potential of comprehensive genomic investigations in patients from consanguineous families to identify monogenic predispositions to severe infections.


Human Molecular Genetics | 2017

A TUBB6 mutation is associated with autosomal dominant non-progressive congenital facial palsy, bilateral ptosis and velopharyngeal dysfunction

Walid Fazeli; Peter Herkenrath; Barbara Stiller; Antje Neugebauer; Julia Fricke; Ruth Lang-Roth; Gudrun Nürnberg; Michaela Thoenes; Jutta Becker; Janine Altmüller; A. Volk; Christian Kubisch; Raoul Heller

&NA; Congenital cranial dysinnervation disorders (CCDDs) comprise a heterogeneous spectrum of diseases characterized by congenital, non‐progressive impairment of eye, eyelid and/or facial movements including Möbius syndrome, Duane retraction syndrome, congenital ptosis, and congenital fibrosis of the extraocular muscles. Over the last 20 years, several CCDDs have been identified as neurodevelopmental disorders that are caused by mutations of genes involved in brain and cranial nerve development, e.g. KIF21A and TUBB3 that each plays a pivotal role for microtubule function. In a five‐generation pedigree, we identified a heterozygous mutation of TUBB6, a gene encoding a class V tubulin which has not been linked to a human hereditary disease so far. The missense mutation (p.Phe394Ser) affects an amino acid residue highly conserved in evolution, and co‐segregates with a phenotype characterized by congenital non‐progressive bilateral facial palsy and congenital velopharyngeal dysfunction presenting with varying degrees of hypomimia, rhinophonia, impaired gag reflex and bilateral ptosis. Expression of the mutated protein in yeast led to an impaired viability compared to wildtype cells when exposed to the microtubule‐poison benomyl. Our findings enlarge the spectrum of tubulinopathies and emphasize that mutations of TUBB6 should be considered in patients with congenital non‐progressive facial palsy. Further studies are needed to verify whether this phenotype is indeed part of the CCDD spectrum.


Human Genetics | 2011

A mutation screen in patients with Kabuki syndrome

Yun Li; Nina Bögershausen; Yasemin Alanay; Pelin Özlem Simsek Kiper; Nadine Plume; Katharina Keupp; Esther Pohl; Barbara Pawlik; Martin Rachwalski; Esther Milz; Michaela Thoenes; Beate Albrecht; Eva-Christina Prott; Margret Lehmkühler; Stephanie Demuth; Gülen Eda Utine; Koray Boduroglu; Katja Frankenbusch; Guntram Borck; Gabriele Gillessen-Kaesbach; Gökhan Yigit; Dagmar Wieczorek; Bernd Wollnik

Collaboration


Dive into the Michaela Thoenes's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maha S. Zaki

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge