Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Genes (Basel) ; 13(12)2022 12 10.
Article in English | MEDLINE | ID: mdl-36553593

ABSTRACT

To keep pace with the rapid advancements in molecular genetics and rare diseases research, we have updated the list of ectodermal dysplasias based on the latest classification approach that was adopted in 2017 by an international panel of experts. For this purpose, we searched the databases PubMed and OMIM for the term "ectodermal dysplasia", referring mainly to changes in the last 5 years. We also tried to obtain information about those diseases on which the last scientific report appeared more than 15 years ago by contacting the authors of the most recent publication. A group of experts, composed of researchers who attended the 8th International Conference on Ectodermal Dysplasias and additional members of the previous classification panel, reviewed the proposed amendments and agreed on a final table listing all 49 currently known ectodermal dysplasias for which the molecular genetic basis has been clarified, including 15 new entities. A newly reported ectodermal dysplasia, linked to the gene LRP6, is described here in more detail. These ectodermal dysplasias, in the strict sense, should be distinguished from syndromes with features of ectodermal dysplasia that are related to genes extraneous to the currently known pathways involved in ectodermal development. The latter group consists of 34 syndromes which had been placed on the previous list of ectodermal dysplasias, but most if not all of them could actually be classified elsewhere. This update should streamline the classification of ectodermal dysplasias, provide guidance to the correct diagnosis of rare disease entities, and facilitate the identification of individuals who could benefit from novel treatment options.


Subject(s)
Ectodermal Dysplasia , Humans , Ectodermal Dysplasia/diagnosis , Ectodermal Dysplasia/genetics , Syndrome , PubMed , Rare Diseases
2.
Genes (Basel) ; 13(11)2022 11 15.
Article in English | MEDLINE | ID: mdl-36421794

ABSTRACT

We report on a cohort of 204 children referred between January 2017 and January 2022 to the German Center for Ectodermal Dysplasias, Erlangen. The most frequent reasons for referral were tooth malformations and lack of multiple teeth leading to the suspicion of an ectodermal dysplasia. Many patients also suffered from being unable to perspire. Nail abnormalities, in contrast, represented a much rarer finding, albeit the impact on some individuals was large. As ectodermal dysplasias are congenital genetic conditions affecting the development and/or homeostasis of two or more ectodermal derivatives, including hair, teeth, nails, and certain glands, we analyzed congenital nail disorders detected in these patients. Dystrophic or otherwise abnormal nails were evident in 17 of 18 subjects with pathogenic WNT10A or GJB6 variants but in none of 161 children with EDA variants underlying X-linked hypohidrotic ectodermal dysplasia. However, 2 of 17 children who carry mutations in EDAR or EDARADD, two other genes involved in the ectodysplasin A signaling pathway, showed nail abnormalities, such as brittle or hypoplastic nails. TP63 variants were regularly associated with nail disorders. In one girl, anonychia congenita caused by a compound heterozygous variant of the R-spondin-4 gene (RSPO4) was diagnosed. Thus, nail dysplasia is rarer among patients with ectodermal dysplasia than commonly thought.


Subject(s)
Ectodermal Dysplasia , Limb Deformities, Congenital , Nails, Malformed , Child , Female , Humans , Nails , Ectodermal Dysplasia/genetics , Nails, Malformed/genetics , Ectoderm
3.
EMBO J ; 40(7): e106745, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33491228

ABSTRACT

Circadian rhythms are a pervasive property of mammalian cells, tissues and behaviour, ensuring physiological adaptation to solar time. Models of cellular timekeeping revolve around transcriptional feedback repression, whereby CLOCK and BMAL1 activate the expression of PERIOD (PER) and CRYPTOCHROME (CRY), which in turn repress CLOCK/BMAL1 activity. CRY proteins are therefore considered essential components of the cellular clock mechanism, supported by behavioural arrhythmicity of CRY-deficient (CKO) mice under constant conditions. Challenging this interpretation, we find locomotor rhythms in adult CKO mice under specific environmental conditions and circadian rhythms in cellular PER2 levels when CRY is absent. CRY-less oscillations are variable in their expression and have shorter periods than wild-type controls. Importantly, we find classic circadian hallmarks such as temperature compensation and period determination by CK1δ/ε activity to be maintained. In the absence of CRY-mediated feedback repression and rhythmic Per2 transcription, PER2 protein rhythms are sustained for several cycles, accompanied by circadian variation in protein stability. We suggest that, whereas circadian transcriptional feedback imparts robustness and functionality onto biological clocks, the core timekeeping mechanism is post-translational.


Subject(s)
Circadian Rhythm , Cryptochromes/metabolism , Animals , Cells, Cultured , Cryptochromes/deficiency , Cryptochromes/genetics , Drosophila melanogaster , Female , Locomotion , Male , Mice , Mice, Inbred C57BL , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism
4.
Sci Rep ; 7: 40304, 2017 01 13.
Article in English | MEDLINE | ID: mdl-28084307

ABSTRACT

Sleep is a highly conserved and essential behaviour in many species, including the fruit fly Drosophila melanogaster. In the wild, sensory signalling encoding environmental information must be integrated with sleep drive to ensure that sleep is not initiated during detrimental conditions. However, the molecular and circuit mechanisms by which sleep timing is modulated by the environment are unclear. Here we introduce a novel behavioural paradigm to study this issue. We show that in male fruit flies, onset of the daytime siesta is delayed by ambient temperatures above 29 °C. We term this effect Prolonged Morning Wakefulness (PMW). We show that signalling through the TrpA1 thermo-sensor is required for PMW, and that TrpA1 specifically impacts siesta onset, but not night sleep onset, in response to elevated temperatures. We identify two critical TrpA1-expressing circuits and show that both contact DN1p clock neurons, the output of which is also required for PMW. Finally, we identify the circadian blue-light photoreceptor CRYPTOCHROME as a molecular regulator of PMW, and propose a model in which the Drosophila nervous system integrates information encoding temperature, light, and time to dynamically control when sleep is initiated. Our results provide a platform to investigate how environmental inputs co-ordinately regulate sleep plasticity.


Subject(s)
Circadian Rhythm/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Sleep/genetics , TRPA1 Cation Channel/genetics , Animals , Drosophila melanogaster/physiology , Humans , Ion Channels , Light , Models, Animal , Motor Activity/genetics , Neurons/metabolism , Neurons/physiology , Sleep/physiology , Temperature , Wakefulness/genetics , Wakefulness/physiology
5.
Nat Commun ; 7: 10165, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26836814

ABSTRACT

The bed bug, Cimex lectularius, has re-established itself as a ubiquitous human ectoparasite throughout much of the world during the past two decades. This global resurgence is likely linked to increased international travel and commerce in addition to widespread insecticide resistance. Analyses of the C. lectularius sequenced genome (650 Mb) and 14,220 predicted protein-coding genes provide a comprehensive representation of genes that are linked to traumatic insemination, a reduced chemosensory repertoire of genes related to obligate hematophagy, host-symbiont interactions, and several mechanisms of insecticide resistance. In addition, we document the presence of multiple putative lateral gene transfer events. Genome sequencing and annotation establish a solid foundation for future research on mechanisms of insecticide resistance, human-bed bug and symbiont-bed bug associations, and unique features of bed bug biology that contribute to the unprecedented success of C. lectularius as a human ectoparasite.


Subject(s)
Bedbugs/genetics , Ectoparasitic Infestations , Feeding Behavior , Gene Transfer, Horizontal/genetics , Host-Parasite Interactions/genetics , Insecticide Resistance/genetics , Insecticides , Animals , Genome , Humans , Sequence Analysis, DNA
6.
PLoS One ; 11(1): e0146571, 2016.
Article in English | MEDLINE | ID: mdl-26741981

ABSTRACT

Cryptochrome (CRY) is the primary photoreceptor of Drosophila's circadian clock. It resets the circadian clock by promoting light-induced degradation of the clock protein Timeless (TIM) in the proteasome. Under constant light, the clock stops because TIM is absent, and the flies become arrhythmic. In addition to TIM degradation, light also induces CRY degradation. This depends on the interaction of CRY with several proteins such as the E3 ubiquitin ligases Jetlag (JET) and Ramshackle (BRWD3). However, CRY can seemingly also be stabilized by interaction with the kinase Shaggy (SGG), the GSK-3 beta fly orthologue. Consequently, flies with SGG overexpression in certain dorsal clock neurons are reported to remain rhythmic under constant light. We were interested in the interaction between CRY, Ramshackle and SGG and started to perform protein interaction studies in S2 cells. To our surprise, we were not able to replicate the results, that SGG overexpression does stabilize CRY, neither in S2 cells nor in the relevant clock neurons. SGG rather does the contrary. Furthermore, flies with SGG overexpression in the dorsal clock neurons became arrhythmic as did wild-type flies. Nevertheless, we could reproduce the published interaction of SGG with TIM, since flies with SGG overexpression in the lateral clock neurons shortened their free-running period. We conclude that SGG does not directly interact with CRY but rather with TIM. Furthermore we could demonstrate, that an unspecific antibody explains the observed stabilization effects on CRY.


Subject(s)
Cryptochromes/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/enzymology , Eye Proteins/metabolism , Glycogen Synthase Kinase 3/physiology , Animals , Cell Line , Circadian Clocks , Protein Stability , Proteolysis
7.
J Biol Rhythms ; 29(2): 75-86, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24682202

ABSTRACT

Many organisms change their activity on moonlit nights. Even the fruit fly Drosophila melanogaster responds to moonlight with a shift of activity into the night, at least under laboratory conditions. The compound eyes have been shown to be essential for the perception of moonlight, but it is unknown which of the 5 rhodopsins in the eyes are responsible for the observed moonlight effects. Here, we show that the outer (R1-R6) and inner (R7 and R8) photoreceptor cells in a fly's ommatidium interact in a complex manner to provoke the moonlight effects on locomotor activity. The shift of the evening activity peak into the night depends on several rhodopsins in the inner and outer photoreceptor cells. The increase in relative nocturnal activity in response to moonlight is mainly mediated by the rhodopsin 6-expressing inner photoreceptor cell R8 together with the rhodopsin 1-expressing outer receptor cells (R1-R6), whereas just rhodopsin 1 of R1 to R6 seems necessary for increasing nocturnal activity in response to increasing daylight intensity.


Subject(s)
Biological Clocks/physiology , Drosophila melanogaster/physiology , Eye/radiation effects , Light , Photoreceptor Cells, Invertebrate/physiology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eye/cytology , Eye/metabolism , Immunohistochemistry , Microscopy, Confocal , Moon , Motor Activity/physiology , Mutation , Ocular Physiological Phenomena/radiation effects , Photoreceptor Cells, Invertebrate/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism
8.
J Biol Rhythms ; 27(1): 37-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22306972

ABSTRACT

The ability to adapt to different environmental conditions including seasonal changes is a key feature of the circadian clock. Here, we compared the ability of 3 Drosophila melanogaster wild-type strains to adapt rhythmic activity to long photoperiods simulated in the laboratory. Fruit flies are predominantly crepuscular with activity bouts in the morning (M) and evening (E). The M peak follows dawn and the E peak follows dusk when the photoperiod is extended. We show that this ability is restricted to a certain extension of the phase angle between M and E peaks, such that the E peak does not delay beyond a certain phase under long days. We demonstrate that this ability is significantly improved by simulated twilight and that it depends additionally on the genetic background and the ambient temperature. At 20 °C, the laboratory strain CantonS had the most flexible phase angle between M and E peaks, a Northern wild-type strain had an intermediate one, and a Southern wild-type strain had the lowest flexibility. Furthermore, we found that the 3 strains differed in clock light sensitivity, with the CantonS and the Northern strains more light sensitive than the Southern strain. These results are generally in accord with the recently discovered polymorphisms in the timeless gene (tim) that affect clock light sensitivity.


Subject(s)
Adaptation, Physiological/genetics , Motor Activity/radiation effects , Photoperiod , Adaptation, Physiological/radiation effects , Animals , Circadian Rhythm , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Light , Male , Temperature
9.
Curr Biol ; 21(9): 719-29, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21530261

ABSTRACT

BACKGROUND: Circadian clocks are synchronized to the solar day via visual and specialized photoreceptors. In Drosophila, CRYPTOCHROME (CRY) is a major photoreceptor that mediates resetting of the circadian clock via light-dependent degradation of the clock protein TIMELESS (TIM). However, in the absence of CRY, this TIM-mediated resetting still occurs in some pacemaker neurons, resulting in synchronized behavioral rhythms when flies are exposed to light-dark cycles. Even in the additional absence of visual photoreception, partial molecular and behavioral light synchronization persists. Therefore, other important clock-related photoreceptive and synchronization mechanisms must exist. RESULTS: We identified a novel clock-controlled gene (quasimodo) that encodes a light-responsive and membrane-anchored Zona Pellucida domain protein that supports light-dependent TIM degradation. Whereas wild-type flies become arrhythmic in constant light (LL), quasimodo mutants elicit rhythmic expression of clock proteins and behavior in LL. QUASIMODO (QSM) can function independently of CRY and is predominantly expressed within CRY-negative clock neurons. Interestingly, downregulation of qsm in the clock circuit restores LL clock protein rhythms in qsm-negative neurons, indicating that qsm-mediated light input is not entirely cell autonomous and can be accessed by the clock circuit. CONCLUSIONS: Our findings indicate that QSM constitutes part of a novel and CRY-independent light input to the circadian clock. Like CRY, this pathway targets the clock protein TIM. QSM's light-responsive character in conjunction with the predicted localization at the outer neuronal membrane suggests that its function is linked to a yet unidentified membrane-bound photoreceptor.


Subject(s)
Brain/metabolism , Circadian Clocks/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , GPI-Linked Proteins/metabolism , Light Signal Transduction/physiology , Zona Pellucida/metabolism , Animals , Animals, Genetically Modified , Blotting, Western , Cloning, Molecular , DNA Primers , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , GPI-Linked Proteins/genetics , Genotype , Immunohistochemistry , Light Signal Transduction/genetics , Microscopy, Fluorescence , Neurons/metabolism , Photoperiod , Polymerase Chain Reaction , RNA Interference
10.
FEBS Lett ; 585(10): 1435-42, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21354415

ABSTRACT

Nowadays humans mainly rely on external, unnatural clocks such as of cell phones and alarm clocks--driven by circuit boards and electricity. Nevertheless, our body is under the control of another timer firmly anchored in our genes. This evolutionary very old biological clock drives most of our physiology and behavior. The genes that control our internal clock are conserved among most living beings. One organism that shares this ancient clock mechanism with us humans is the fruitfly Drosophila melanogaster. Since it turned out that Drosophila is an excellent model, it is no surprise that its clock is very well and intensely investigated. In the following review we want to display an overview of the current understanding of Drosophila's circadian clock.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Drosophila melanogaster/physiology , Adaptation, Physiological , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Neurons/cytology
11.
Curr Biol ; 19(3): 241-7, 2009 Feb 10.
Article in English | MEDLINE | ID: mdl-19185492

ABSTRACT

Circadian clocks regulate daily fluctuations of many physiological and behavioral aspects in life. They are synchronized with the environment via light or temperature cycles [1]. Natural fluctuations of the day length (photoperiod) and temperature necessitate a daily reset of the circadian clock on the molecular level. In Drosophila, the blue-light photoreceptor Cryptochrome (Cry) mediates a rapid light-dependent degradation of the clock protein Timeless (Tim) via the F box protein Jetlag (Jet) and the proteasome, which initiates the resetting of the molecular clock [2, 3]. Cry is also degraded in the light but whereas the degradation of Tim is well characterized [4-8], the mechanism for light-dependent degradation of Cry is mostly unknown. Until now it was believed that these two degradation pathways are distinct [4, 9]. Here we reveal that Jetlag also interacts with Cry in a light-dependent manner. After illumination, Jetlag induces massive degradation of Cry, which can be prevented in vitro and in vivo by adding Tim as an antagonist. We show that the affinity of Tim for Cry and Jetlag determines the sequential order of Tim and Cry degradation and thus reveal an intimate connection between the light-dependent degradation of these two proteins by the same proteasomal pathway.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/radiation effects , Drosophila Proteins/metabolism , Drosophila/physiology , Eye Proteins/metabolism , F-Box Proteins/metabolism , Light , Receptors, G-Protein-Coupled/metabolism , Animals , Blotting, Western , Cryptochromes , Drosophila/genetics , Immunoprecipitation , Luciferases , Models, Biological , Oligonucleotides/genetics , Protein Binding/radiation effects , Two-Hybrid System Techniques
12.
Proc Natl Acad Sci U S A ; 103(46): 17313-8, 2006 Nov 14.
Article in English | MEDLINE | ID: mdl-17068124

ABSTRACT

Organisms use the daily cycles of light and darkness to synchronize their internal circadian clocks with the environment. Because they optimize physiological processes and behavior, properly synchronized circadian clocks are thought to be important for the overall fitness. In Drosophila melanogaster, the circadian clock is synchronized with the natural environment by light-dependent degradation of the clock protein Timeless, mediated by the blue-light photoreceptor Cryptochrome (Cry). Here we report identification of a genetic variant, Veela, which severely disrupts this process, because these genetically altered flies maintain behavioral and molecular rhythmicity under constant-light conditions that usually stop the clock. We show that the Veela strain carries a natural timeless allele (ls-tim), which encodes a less-light-sensitive form of Timeless in combination with a mutant variant of the F-box protein Jetlag. However, neither the ls-tim nor the jetlag genetic variant alone is sufficient to disrupt light input into the central pacemaker. We show a strong interaction between Veela and cryptochrome genetic variants, demonstrating that the Jetlag, Timeless, and Cry proteins function in the same pathway. Veela also reveals a function for the two natural variants of timeless, which differ in their sensitivity to light. In combination with the complex array of retinal and extraretinal photoreceptors known to signal light to the pacemaker, this previously undescribed molecular component of photic sensitivity mediated by the two Timeless proteins reveals that an unexpectedly rich complexity underlies modulation of this process.


Subject(s)
Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Flavoproteins/metabolism , Light , Signal Transduction , Alleles , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cryptochromes , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , F-Box Proteins/chemistry , F-Box Proteins/genetics , F-Box Proteins/metabolism , Flavoproteins/genetics , Molecular Sequence Data , Motor Activity , Mutation/genetics , Neuroglia/metabolism , Neurons/metabolism , Polymorphism, Genetic/genetics , Renin/metabolism , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...