Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Hum Genet ; 110(8): 1356-1376, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37421948

RESUMO

By converting physical forces into electrical signals or triggering intracellular cascades, stretch-activated ion channels allow the cell to respond to osmotic and mechanical stress. Knowledge of the pathophysiological mechanisms underlying associations of stretch-activated ion channels with human disease is limited. Here, we describe 17 unrelated individuals with severe early-onset developmental and epileptic encephalopathy (DEE), intellectual disability, and severe motor and cortical visual impairment associated with progressive neurodegenerative brain changes carrying ten distinct heterozygous variants of TMEM63B, encoding for a highly conserved stretch-activated ion channel. The variants occurred de novo in 16/17 individuals for whom parental DNA was available and either missense, including the recurrent p.Val44Met in 7/17 individuals, or in-frame, all affecting conserved residues located in transmembrane regions of the protein. In 12 individuals, hematological abnormalities co-occurred, such as macrocytosis and hemolysis, requiring blood transfusions in some. We modeled six variants (p.Val44Met, p.Arg433His, p.Thr481Asn, p.Gly580Ser, p.Arg660Thr, and p.Phe697Leu), each affecting a distinct transmembrane domain of the channel, in transfected Neuro2a cells and demonstrated inward leak cation currents across the mutated channel even in isotonic conditions, while the response to hypo-osmotic challenge was impaired, as were the Ca2+ transients generated under hypo-osmotic stimulation. Ectopic expression of the p.Val44Met and p.Gly580Cys variants in Drosophila resulted in early death. TMEM63B-associated DEE represents a recognizable clinicopathological entity in which altered cation conductivity results in a severe neurological phenotype with progressive brain damage and early-onset epilepsy associated with hematological abnormalities in most individuals.


Assuntos
Encefalopatias , Deficiência Intelectual , Humanos , Encefalopatias/genética , Canais Iônicos/genética , Encéfalo , Deficiência Intelectual/genética , Fenótipo
2.
Hum Mol Genet ; 32(9): 1524-1538, 2023 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-36611008

RESUMO

Drosophila is an excellent model organism for studying human neurodegenerative diseases (NDs). However, there is still almost no experimental system that could directly observe the degeneration of neurons and automatically quantify axonal degeneration. In this study, we created MeDUsA (a 'method for the quantification of degeneration using fly axons'), a standalone executable computer program based on Python that combines a pre-trained deep-learning masking tool with an axon terminal counting tool. This software automatically quantifies the number of retinal R7 axons in Drosophila from a confocal z-stack image series. Using this software, we were able to directly demonstrate that axons were degenerated by the representative causative genes of NDs for the first time in Drosophila. The fly retinal axon is an excellent experimental system that is capable of mimicking the pathology of axonal degeneration in human NDs. MeDUsA rapidly and accurately quantifies axons in Drosophila photoreceptor neurons. It enables large-scale research into axonal degeneration, including screening to identify genes or drugs that mediate axonal toxicity caused by ND proteins and diagnose the pathological significance of novel variants of human genes in axons.


Assuntos
Proteínas de Drosophila , Doenças Neurodegenerativas , Animais , Humanos , Drosophila/genética , Drosophila/metabolismo , Doenças Neurodegenerativas/metabolismo , Axônios/metabolismo , Neurônios/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
3.
J Neurosci ; 42(24): 4937-4952, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35534228

RESUMO

In human neurodegenerative diseases, neurons undergo axonal degeneration months to years before they die. Here, we developed a system modeling early degenerative events in Drosophila adult photoreceptor cells. Thanks to the stereotypy of their axonal projections, this system delivers quantitative data on sporadic and progressive axonal degeneration of photoreceptor cells. Using this method, we show that exposure of adult female flies to a constant light stimulation for several days overcomes the intrinsic resilience of R7 photoreceptors and leads to progressive axonal degeneration. This was not associated with apoptosis. We furthermore provide evidence that loss of synaptic integrity between R7 and a postsynaptic partner preceded axonal degeneration, thus recapitulating features of human neurodegenerative diseases. Finally, our experiments uncovered a role of postsynaptic partners of R7 to initiate degeneration, suggesting that postsynaptic cells signal back to the photoreceptor to maintain axonal structure. This model can be used to dissect cellular and circuit mechanisms involved in the early events of axonal degeneration, allowing for a better understanding of how neurons cope with stress and lose their resilience capacities.SIGNIFICANCE STATEMENT Neurons can be active and functional for several years. In the course of aging and in disease conditions leading to neurodegeneration, subsets of neurons lose their resilience and start dying. What initiates this turning point at the cellular level is not clear. Here, we developed a model allowing to systematically describe this phase. The loss of synapses and axons represents an early and functionally relevant event toward degeneration. Using the ordered distribution of Drosophila photoreceptor axon terminals, we assembled a system to study sporadic initiation of axon loss and delineated a role for non-cell-autonomous activity regulation in the initiation of axon degeneration. This work will help shed light on key steps in the etiology of nonfamilial cases of neurodegenerative diseases.


Assuntos
Proteínas de Drosophila , Doenças Neurodegenerativas , Animais , Axônios/fisiologia , Drosophila/fisiologia , Proteínas de Drosophila/genética , Feminino , Sinapses/fisiologia
4.
Hum Mol Genet ; 31(1): 69-81, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34346499

RESUMO

An optimal Golgi transport system is important for mammalian cells. The adenosine diphosphate (ADP) ribosylation factors (ARF) are key proteins for regulating cargo sorting at the Golgi network. In this family, ARF3 mainly works at the trans-Golgi network (TGN), and no ARF3-related phenotypes have yet been described in humans. We here report the clinical and genetic evaluations of two unrelated children with de novo pathogenic variants in the ARF3 gene: c.200A > T (p.Asp67Val) and c.296G > T (p.Arg99Leu). Although the affected individuals presented commonly with developmental delay, epilepsy and brain abnormalities, there were differences in severity, clinical course and brain lesions. In vitro subcellular localization assays revealed that the p.Arg99Leu mutant localized to Golgi apparatus, similar to the wild-type, whereas the p.Asp67Val mutant tended to show a disperse cytosolic pattern together with abnormally dispersed Golgi localization, similar to that observed in a known dominant negative variant (p.Thr31Asn). Pull-down assays revealed that the p.Asp67Val had a loss-of-function effect and the p.Arg99Leu variant had increased binding of the adaptor protein, Golgi-localized, γ-adaptin ear-containing, ARF-binding protein 1 (GGA1), supporting the gain of function. Furthermore, in vivo studies revealed that p.Asp67Val transfection led to lethality in flies. In contrast, flies expressing p.Arg99Leu had abnormal rough eye, as observed in the gain-of-function variant p.Gln71Leu. These data indicate that two ARF3 variants, the possibly loss-of-function p.Asp67Val and the gain-of-function p.Arg99Leu, both impair the Golgi transport system. Therefore, it may not be unreasonable that they showed different clinical features like diffuse brain atrophy (p.Asp67Val) and cerebellar hypoplasia (p.Arg99Leu).


Assuntos
Fatores de Ribosilação do ADP , Transtornos do Neurodesenvolvimento , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Encéfalo/metabolismo , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Mamíferos/metabolismo , Transtornos do Neurodesenvolvimento/metabolismo
5.
Dev Biol ; 421(2): 233-244, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27908785

RESUMO

Axonal branching is one of the key processes within the enormous complexity of the nervous system to enable a single neuron to send information to multiple targets. However, the molecular mechanisms that control branch formation are poorly understood. In particular, previous studies have rarely addressed the mechanisms underlying axonal bifurcation, in which axons form new branches via splitting of the growth cone. We demonstrate that DISCO Interacting Protein 2 (DIP2) is required for precise axonal bifurcation in Drosophila mushroom body (MB) neurons by suppressing ectopic bifurcation and regulating the guidance of sister axons. We also found that DIP2 localize to the plasma membrane. Domain function analysis revealed that the AMP-synthetase domains of DIP2 are essential for its function, which may involve exerting a catalytic activity that modifies fatty acids. Genetic analysis and subsequent biochemical analysis suggested that DIP2 is involved in the fatty acid metabolization of acyl-CoA. Taken together, our results reveal a function of DIP2 in the developing nervous system and provide a potential functional relationship between fatty acid metabolism and axon morphogenesis.


Assuntos
Orientação de Axônios , Axônios/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Corpos Pedunculados/inervação , Corpos Pedunculados/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Animais Geneticamente Modificados , Células Clonais , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Ácidos Graxos/metabolismo , Metabolismo dos Lipídeos , Modelos Biológicos , Mutação/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Domínios Proteicos , Interferência de RNA , Deleção de Sequência , Homologia de Sequência de Aminoácidos
6.
Biochem Biophys Res Commun ; 487(4): 898-902, 2017 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-28465232

RESUMO

Axonal branching is a fundamental requirement for sending electrical signals to multiple targets. However, despite the importance of axonal branching in neural development and function, the molecular mechanisms that control branch formation are poorly understood. Previous studies have hardly addressed the intracellular signaling cascade of axonal bifurcation characterized by growth cone splitting. Recently we reported that DISCO interacting protein 2 (DIP2) regulates bifurcation of mushroom body axons in Drosophila melanogaster. DIP2 mutant displays ectopic bifurcations in α/ß neurons. Taking advantage of this phenomenon, we tried to identify genes involved in branching formation by comparing the transcriptome of wild type with that of DIP2 RNAi flies. After the microarray analysis, Glaikit (Gkt), a member of the phospholipase D superfamily, was identified as a downstream target of DIP2 by RNAi against gkt and qRT-PCR experiment. Single cell MARCM analysis of gkt mutant phenocopied the ectopic axonal branches observed in DIP2 mutant. Furthermore, a genetic analysis between gkt and DIP2 revealed that gkt potentially acts in parallel with DIP2. In conclusion, we identified a novel gene underlying the axonal bifurcation process.


Assuntos
Axônios/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Perfilação da Expressão Gênica , Genoma de Inseto/genética , Corpos Pedunculados/metabolismo , Proteínas do Tecido Nervoso/genética , Animais , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Mutação , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Análise Serial de Proteínas , Interferência de RNA
7.
Biochem Biophys Res Commun ; 487(1): 116-121, 2017 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-28396149

RESUMO

Precisely controlled axon guidance for complex neuronal wiring is essential for appropriate neuronal function. c-Jun N-terminal kinase (JNK) was found to play a role in axon guidance recently as well as in cell proliferation, protection and apoptosis. In spite of many genetic and molecular studies on these biological processes regulated by JNK, how JNK regulates axon guidance accurately has not been fully explained thus far. To address this question, we use the Drosophila mushroom body (MB) as a model since the α/ß axons project in two distinct directions. Here we show that DISCO interacting protein 2 (DIP2) is required for the accurate direction of axonal guidance. DIP2 expression is under the regulation of Basket (Bsk), the Drosophila homologue of JNK. We additionally found that the Bsk/DIP2 pathway is independent from the AP-1 transcriptional factor complex pathway, which is directly activated by Bsk. In conclusion, our findings revealed DIP2 as a novel effector downstream of Bsk modulating the direction of axon projection.


Assuntos
Orientação de Axônios/fisiologia , Axônios/fisiologia , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Morfogênese/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Animais , Axônios/ultraestrutura , Células Cultivadas , Drosophila/fisiologia , Corpos Pedunculados/fisiologia
8.
Cell Rep ; 43(2): 113798, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38381608

RESUMO

Neurons establish specific synapses based on the adhesive properties of cell-surface proteins while also retaining the ability to form synapses in a relatively non-selective manner. However, comprehensive understanding of the underlying mechanism reconciling these opposing characteristics remains incomplete. Here, we have identified Side-IV/Beat-IIb, members of the Drosophila immunoglobulin superfamily, as a combination of cell-surface recognition molecules inducing synapse formation. The Side-IV/Beat-IIb combination transduces bifurcated signaling with Side-IV's co-receptor, Kirre, and a synaptic scaffold protein, Dsyd-1. Genetic experiments and subcellular protein localization analyses showed the Side-IV/Beat-IIb/Kirre/Dsyd-1 complex to have two essential functions. First, it narrows neuronal binding specificity through Side-IV/Beat-IIb extracellular interactions. Second, it recruits synapse formation factors, Kirre and Dsyd-1, to restrict synaptic loci and inhibit miswiring. This dual function explains how the combinations of cell-surface molecules enable the ranking of preferred interactions among neuronal pairs to achieve synaptic specificity in complex circuits in vivo.


Assuntos
Comunicação Celular , Imunoglobulinas , Animais , Imunoglobulinas/genética , Membrana Celular , Drosophila , Proteínas de Membrana
9.
Front Genet ; 15: 1383176, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38601074

RESUMO

Introduction: RRAS2, a member of the R-Ras subfamily of Ras-like low-molecular-weight GTPases, is considered to regulate cell proliferation and differentiation via the RAS/MAPK signaling pathway. Seven RRAS2 pathogenic variants have been reported in patients with Noonan syndrome; however, few functional analyses have been conducted. Herein, we report two patients who presented with a Noonan-like phenotype with recurrent and novel RRAS2 pathogenic variants (p.Gly23Val and p.Gly24Glu, respectively) and the results of their functional analysis. Materials and methods: Wild-type (WT) and mutant RRAS2 genes were transiently expressed in Human Embryonic Kidney293 cells. Expression of RRAS2 and phosphorylation of ERK1/2 were confirmed by Western blotting, and the RAS signaling pathway activity was measured using a reporter assay system with the serum response element-luciferase construct. WT and p.Gly23Val RRAS2 were expressed in Drosophila eye using the glass multiple reporter-Gal4 driver. Mutant mRNA microinjection into zebrafish embryos was performed, and the embryo jaws were observed. Results: No obvious differences in the expression of proteins WT, p.Gly23Val, and p.Gly24Glu were observed. The luciferase reporter assay showed that the activity of p.Gly23Val was 2.45 ± 0.95-fold higher than WT, and p.Gly24Glu was 3.06 ± 1.35-fold higher than WT. For transgenic flies, the p.Gly23Val expression resulted in no adults flies emerging, indicating lethality. For mutant mRNA-injected zebrafish embryos, an oval shape and delayed jaw development were observed compared with WT mRNA-injected embryos. These indicated hyperactivity of the RAS signaling pathway. Discussion: Recurrent and novel RRAS2 variants that we reported showed increased in vitro or in vivo RAS signaling pathway activity because of gain-of-function RRAS2 variants. Clinical features are similar to those previously reported, suggesting that RRAS2 gain-of-function variants cause this disease in patients.

10.
Nat Commun ; 15(1): 4696, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38824133

RESUMO

Age-related microangiopathy, also known as small vessel disease (SVD), causes damage to the brain, retina, liver, and kidney. Based on the DNA damage theory of aging, we reasoned that genomic instability may underlie an SVD caused by dominant C-terminal variants in TREX1, the most abundant 3'-5' DNA exonuclease in mammals. C-terminal TREX1 variants cause an adult-onset SVD known as retinal vasculopathy with cerebral leukoencephalopathy (RVCL or RVCL-S). In RVCL, an aberrant, C-terminally truncated TREX1 mislocalizes to the nucleus due to deletion of its ER-anchoring domain. Since RVCL pathology mimics that of radiation injury, we reasoned that nuclear TREX1 would cause DNA damage. Here, we show that RVCL-associated TREX1 variants trigger DNA damage in humans, mice, and Drosophila, and that cells expressing RVCL mutant TREX1 are more vulnerable to DNA damage induced by chemotherapy and cytokines that up-regulate TREX1, leading to depletion of TREX1-high cells in RVCL mice. RVCL-associated TREX1 mutants inhibit homology-directed repair (HDR), causing DNA deletions and vulnerablility to PARP inhibitors. In women with RVCL, we observe early-onset breast cancer, similar to patients with BRCA1/2 variants. Our results provide a mechanistic basis linking aberrant TREX1 activity to the DNA damage theory of aging, premature senescence, and microvascular disease.


Assuntos
Dano ao DNA , Exodesoxirribonucleases , Fosfoproteínas , Animais , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Camundongos , Reparo de DNA por Recombinação , Fenótipo , Mutação , Drosophila/genética , Envelhecimento/genética , Envelhecimento/metabolismo , Feminino , Drosophila melanogaster/genética , Masculino , Doenças Retinianas , Doenças Vasculares , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central
11.
J Am Chem Soc ; 135(33): 12208-11, 2013 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-23914951

RESUMO

Revisiting Murahashi coupling, we found that it effectively allows polymerization of lithiated (hetero)arenes by nickel(II)-catalyzed polycondensation. Deprotonative polymerization of 2-chloro-3-substituted thiophene with n-butyllithium gave head-to-tail-type poly(3-substituted thiophene). Poly(1,4-arylene)s were obtained by the reaction of the corresponding dibromides through lithium-bromine exchange. A lithiated thiophene derivative obtained via deprotonative halogen dance also underwent polymerization to afford a bromo-substituted polythiophene.

12.
Development ; 137(19): 3193-203, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20724446

RESUMO

During neurogenesis in the medulla of the Drosophila optic lobe, neuroepithelial cells are programmed to differentiate into neuroblasts at the medial edge of the developing optic lobe. The wave of differentiation progresses synchronously in a row of cells from medial to the lateral regions of the optic lobe, sweeping across the entire neuroepithelial sheet; it is preceded by the transient expression of the proneural gene lethal of scute [l(1)sc] and is thus called the proneural wave. We found that the epidermal growth factor receptor (EGFR) signaling pathway promotes proneural wave progression. EGFR signaling is activated in neuroepithelial cells and induces l(1)sc expression. EGFR activation is regulated by transient expression of Rhomboid (Rho), which is required for the maturation of the EGF ligand Spitz. Rho expression is also regulated by the EGFR signal. The transient and spatially restricted expression of Rho generates sequential activation of EGFR signaling and assures the directional progression of the differentiation wave. This study also provides new insights into the role of Notch signaling. Expression of the Notch ligand Delta is induced by EGFR, and Notch signaling prolongs the proneural state. Notch signaling activity is downregulated by its own feedback mechanism that permits cells at proneural states to subsequently develop into neuroblasts. Thus, coordinated sequential action of the EGFR and Notch signaling pathways causes the proneural wave to progress and induce neuroblast formation in a precisely ordered manner.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Receptores ErbB/metabolismo , Lobo Óptico de Animais não Mamíferos/metabolismo , Receptores de Peptídeos de Invertebrados/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Ativação Enzimática , Receptores ErbB/genética , Regulação da Expressão Gênica no Desenvolvimento , Janus Quinases/genética , Janus Quinases/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Lobo Óptico de Animais não Mamíferos/embriologia , Receptores de Peptídeos de Invertebrados/genética , Receptores Notch/genética , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo
13.
Development ; 137(19): 3303-13, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20724453

RESUMO

Topographic maps, which maintain the spatial order of neurons in the order of their axonal connections, are found in many parts of the nervous system. Here, we focus on the communication between retinal axons and their postsynaptic partners, lamina neurons, in the first ganglion of the Drosophila visual system, as a model for the formation of topographic maps. Post-mitotic lamina precursor cells differentiate upon receiving Hedgehog signals delivered through newly arriving retinal axons and, before maturing to extend neurites, extend short processes toward retinal axons to create the lamina column. The lamina column provides the cellular basis for establishing stereotypic synapses between retinal axons and lamina neurons. In this study, we identified two cell-adhesion molecules: Hibris, which is expressed in post-mitotic lamina precursor cells; and Roughest, which is expressed on retinal axons. Both proteins belong to the nephrin/NEPH1 family. We provide evidence that recognition between post-mitotic lamina precursor cells and retinal axons is mediated by interactions between Hibris and Roughest. These findings revealed mechanisms by which axons of presynaptic neurons deliver signals to induce the development of postsynaptic partners at the target area. Postsynaptic partners then recognize the presynaptic axons to make ensembles, thus establishing a topographic map along the anterior/posterior axis.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Proteínas do Olho/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Moléculas de Adesão Celular Neuronais/genética , Proteínas de Drosophila/genética , Proteínas do Olho/genética , Proteínas de Membrana/genética , Proteínas Nucleares/metabolismo , Transdução de Sinais
14.
Development ; 137(10): 1755-64, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20430750

RESUMO

The Wingless (Wg)/Wnt signaling pathway is highly conserved throughout many multicellular organisms. It directs the development of diverse tissues and organs by regulating important processes such as proliferation, polarity and the specification of cell fates. Upon activation of the Wg/Wnt signaling pathway, Armadillo (Arm)/beta-catenin is stabilized and interacts with the TCF family of transcription factors, which in turn activate Wnt target genes. We show here that Arm interacts with a novel BED (BEAF and Dref) finger protein that we have termed Sunspot (Ssp). Ssp transactivates Drosophila E2F-1 (dE2F-1) and PCNA expression, and positively regulates the proliferation of imaginal disc cells and the endoreplication of salivary gland cells. Wg negatively regulates the function of Ssp by changing its subcellular localization in the salivary gland. In addition, Ssp was found not to be involved in the signaling pathway mediated by Arm associated with dTCF. Our findings indicate that Arm controls development in part by regulating the function of Ssp.


Assuntos
Proliferação de Células , Proteínas de Drosophila/fisiologia , Drosophila/crescimento & desenvolvimento , Fatores de Transcrição/fisiologia , Proteína Wnt1/fisiologia , Animais , Animais Geneticamente Modificados , Proteínas do Domínio Armadillo/metabolismo , Células Cultivadas , Drosophila/genética , Drosophila/metabolismo , Drosophila/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Ligação Proteica , Glândulas Salivares/crescimento & desenvolvimento , Glândulas Salivares/metabolismo , Glândulas Salivares/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Distribuição Tecidual , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
15.
Chemistry ; 19(5): 1658-65, 2013 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-23255398

RESUMO

The synthesis of linear and branched oligothiophenes of well-defined structures is performed with regioselective deprotonation of 3-substituted thiophenes and nickel-catalyzed cross-coupling of the thus formed metalated species with a bromothiophene. The reaction of 3-hexylthiophene with EtMgCl and 2,2,6,6-tetramethylpiperidine (TMP-H, 10 mol%) induces the metalation selectively at the 5-position by use of the catalytically generated hindered magnesium amide (TMPMgCl) and the subsequent reaction of a 2-halo-3-hexylthiophene (bromide or chloride) in the presence of a nickel catalyst affords a head-to-tail (HT)-type dimer. By repeating the same sequence, the linear oligothiophene up to a 4-mer is synthesized in good yield. The reaction of 3-hexylthiophene with 2,3-dibromothiophene also takes place to afford a branched oligothiophene 3-mer in quantitative yield. The obtained 3-mer is also metalated at the sterically less-hindered position in a regioselective manner furnishing a 7-mer in >99% yield after a further coupling reaction with 2,3-dibromothiophene. These dendrimers react with several multifunctionalized organic electrophiles, leading to a variety of branched oligothiophenes.

16.
Genes Genet Syst ; 97(6): 297-309, 2023 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-36878557

RESUMO

Neural activity-dependent synaptic plasticity is an important physiological phenomenon underlying environmental adaptation, memory and learning. However, its molecular basis, especially in presynaptic neurons, is not well understood. Previous studies have shown that the number of presynaptic active zones in the Drosophila melanogaster photoreceptor R8 is reversibly changed in an activity-dependent manner. During reversible synaptic changes, both synaptic disassembly and assembly processes were observed. Although we have established a paradigm for screening molecules involved in synaptic stability and several genes have been identified, genes involved in stimulus-dependent synaptic assembly are still elusive. Therefore, the aim of this study was to identify genes regulating stimulus-dependent synaptic assembly in Drosophila using an automated synapse quantification system. To this end, we performed RNAi screening against 300 memory-defective, synapse-related or transmembrane molecules in photoreceptor R8 neurons. Candidate genes were narrowed down to 27 genes in the first screen using presynaptic protein aggregation as a sign of synaptic disassembly. In the second screen, we directly quantified the decreasing synapse number using a GFP-tagged presynaptic protein marker. We utilized custom-made image analysis software, which automatically locates synapses and counts their number along individual R8 axons, and identified cirl as a candidate gene responsible for synaptic assembly. Finally, we present a new model of stimulus-dependent synaptic assembly through the interaction of cirl and its possible ligand, ten-a. This study demonstrates the feasibility of using the automated synapse quantification system to explore activity-dependent synaptic plasticity in Drosophila R8 photoreceptors in order to identify molecules involved in stimulus-dependent synaptic assembly.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/genética , Drosophila melanogaster/genética , Sinapses/metabolismo , Axônios/metabolismo , Neurônios/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Receptores de Peptídeos/metabolismo
17.
Eur J Med Genet ; 66(8): 104804, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37369308

RESUMO

DExH-box helicases are involved in unwinding of RNA and DNA. Among the 16 DExH-box genes, monoallelic variants of DHX16, DHX30, DHX34, and DHX37 are known to be associated with neurodevelopmental disorders. In particular, DHX30 is well established as a causative gene for neurodevelopmental disorders. Germline variants of DHX9, the closest homolog of DHX30, have not been reported until now as being associated with congenital disorders in humans, except that one de novo heterozygous variant, p.(Arg1052Gln) of the gene was identified during comprehensive screening in a patient with autism; unfortunately, the phenotypic details of this individual are unknown. Herein, we report a patients with a heterozygous de novo missense variant, p.(Gly414Arg) of DHX9 who presented with a short stature, intellectual disability, and ventricular non-compaction cardiomyopathy. The variant was located in the glycine codon of the ATP-binding site, G-C-G-K-T. To assess the pathogenicity of these variants, we generated transgenic Drosophila lines expressing human wild-type and mutant DHX9 proteins: 1) the mutant proteins showed aberrant localization both in the nucleus and the cytoplasm; 2) ectopic expression of wild-type protein in the visual system led to the rough eye phenotype, whereas expression of the mutant proteins had minimal effect; 3) overexpression of the wild-type protein in the retina led to a reduction in axonal numbers, whereas expression of the mutant proteins had a less pronounced effect. Furthermore, in a gene-editing experiment of Dhx9 G416 to R416, corresponding to p.(Gly414Arg) in humans, heterozygous mice showed a reduced body size, reduced emotionality, and cardiac conduction abnormality. In conclusion, we established that heterozygosity for a loss-of-function variant of DHX9 can lead to a new neurodevelopmental disorder.


Assuntos
Deficiência Intelectual , Transtornos do Neurodesenvolvimento , Animais , Humanos , Camundongos , RNA Helicases DEAD-box/genética , Genética Humana , Deficiência Intelectual/genética , Proteínas de Neoplasias/genética , Transtornos do Neurodesenvolvimento/genética , RNA/genética , RNA Helicases
18.
Sci Rep ; 13(1): 975, 2023 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-36653413

RESUMO

The Drosophila behavior/human splicing protein family is involved in numerous steps of gene regulation. In humans, this family consists of three proteins: SFPQ, PSPC1, and NONO. Hemizygous loss-of-function (LoF) variants in NONO cause a developmental delay with several complications (e.g., distinctive facial features, cardiac symptoms, and skeletal symptoms) in an X-linked recessive manner. Most of the reported variants have been LoF variants, and two missense variants have been reported as likely deleterious but with no functional validation. We report three individuals from two families harboring an identical missense variant that is located in the nuclear localization signal, NONO: NM_001145408.2:c.1375C > G p.(Pro459Ala). All of them were male and the variant was inherited from their asymptomatic mothers. Individual 1 was diagnosed with developmental delay and cardiac phenotypes (ventricular tachycardia and dilated cardiomyopathy), which overlapped with the features of reported individuals having NONO LoF variants. Individuals 2 and 3 were monozygotic twins. Unlike in Individual 1, developmental delay with autistic features was the only symptom found in them. A fly experiment and cell localization experiment showed that the NONO variant impaired its proper intranuclear localization, leading to mild LoF. Our findings suggest that deleterious NONO missense variants should be taken into consideration when whole-exome sequencing is performed on male individuals with developmental delay with or without cardiac symptoms.


Assuntos
Cardiomiopatia Dilatada , Proteínas de Ligação a DNA , Coração , Mutação de Sentido Incorreto , Proteínas de Ligação a RNA , Feminino , Humanos , Masculino , Cardiomiopatia Dilatada/genética , Proteínas de Ligação a DNA/genética , Fenótipo , Proteínas de Ligação a RNA/genética
19.
Chemistry ; 18(22): 6959-71, 2012 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-22529058

RESUMO

A family of seven cationic gold complexes that contain both an alkyl substituted π-allene ligand and an electron-rich, sterically hindered supporting ligand was isolated in >90% yield and characterized by spectroscopy and, in three cases, by X-ray crystallography. Solution-phase and solid-state analysis of these complexes established preferential binding of gold to the less substituted C=C bond of the allene and to the allene π face trans to the substituent on the uncomplexed allenyl C=C bond. Kinetic analysis of intermolecular allene exchange established two-term rate laws of the form rate=k(1)[complex]+k(2)[complex][allene] consistent with allene-independent and allene-dependent exchange pathways with energy barriers of ΔG(≠)(1)=17.4-18.8 and ΔG(≠)(2)=15.2-17.6 kcal mol(-1), respectively. Variable temperature (VT) NMR analysis revealed fluxional behavior consistent with facile (ΔG(≠)=8.9-11.4 kcal mol(-1)) intramolecular exchange of the allene π faces through η(1)-allene transition states and/or intermediates that retain a staggered arrangement of the allene substituents. VT NMR/spin saturation transfer analysis of [{P(tBu)(2)o-binaphthyl}Au(η(2)-4,5-nonadiene)](+)SbF(6)(-) (5), which contains elements of chirality in both the phosphine and allene ligands, revealed no epimerization of the allene ligand below the threshold for intermolecular allene exchange (ΔG(≠)(298K)=17.4 kcal mol(-1)), which ruled out the participation of a η(1)-allylic cation species in the low-energy π-face exchange process for this complex.

20.
Fly (Austin) ; 16(1): 275-298, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35765969

RESUMO

The use of Drosophila in neurodegenerative disease research has contributed to the identification of modifier genes for the pathology. The basis for neurodegenerative disease occurrence in Drosophila is the conservation of genes across species and the ability to perform rapid genetic analysis using a compact brain. Genetic findings previously discovered in Drosophila can reveal molecular pathologies involved in human neurological diseases in later years. Disease models using Drosophila began to be generated during the development of genetic engineering. In recent years, results of reverse translational research using Drosophila have been reported. In this review, we discuss research on neurodegenerative diseases; moreover, we introduce various methods for quantifying neurodegeneration in Drosophila.


Assuntos
Drosophila , Doenças Neurodegenerativas , Animais , Modelos Animais de Doenças , Drosophila/genética , Doenças Neurodegenerativas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA