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








Base de dados
Intervalo de ano de publicação
1.
Front Genet ; 14: 881638, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36968586

RESUMO

Transactive response DNA binding protein-43 (TDP-43) is known to mediate neurodegeneration associated with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). The exact mechanism by which TDP-43 exerts toxicity in the brains, spinal cord, and lower motor neurons of affected patients remains unclear. In a novel Drosophila melanogaster model, we report gain-of-function phenotypes due to misexpression of insect codon-optimized version of human wild-type TDP-43 (CO-TDP-43) using both the binary GAL4/UAS system and direct promoter fusion constructs. The CO-TDP-43 model showed robust tissue specific phenotypes in the adult eye, wing, and bristles in the notum. Compared to non-codon optimized transgenic flies, the CO-TDP-43 flies produced increased amount of high molecular weight protein, exhibited pathogenic phenotypes, and showed cytoplasmic aggregation with both nuclear and cytoplasmic expression of TDP-43. Further characterization of the adult retina showed a disruption in the morphology and function of the photoreceptor neurons with the presence of acidic vacuoles that are characteristic of autophagy. Based on our observations, we propose that TDP-43 has the propensity to form toxic protein aggregates via a gain-of-function mechanism, and such toxic overload leads to activation of protein degradation pathways such as autophagy. The novel codon optimized TDP-43 model is an excellent resource that could be used in genetic screens to identify and better understand the exact disease mechanism of TDP-43 proteinopathies and find potential therapeutic targets.

2.
STAR Protoc ; 3(2): 101430, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35664260

RESUMO

The adult Drosophila compound eye is an ideal in vivo model for studying biological questions. However, light microscopy of this tissue requires cumbersome embedding and sectioning. Here, we document detailed whole-mount procedures for immunolabeling the adult retina, enabling high-quality studies of fluorescent-tagged targets with straightforward preparations. We describe the steps for visualizing the nuclear lamina, membrane-associated protein, and actin-rich rhabdomere, but this robust protocol can apply to other cellular structures and target proteins. For complete details on the use and execution of this protocol, please refer to Chang et al. (2021).


Assuntos
Drosophila , Técnicas Histológicas , Actinas , Animais , Técnicas Histológicas/métodos , Microscopia , Retina/diagnóstico por imagem
3.
Nat Commun ; 12(1): 4258, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34253734

RESUMO

The maintenance of constant karyoplasmic ratios suggests that nuclear size has physiological significance. Nuclear size anomalies have been linked to malignant transformation, although the mechanism remains unclear. By expressing dominant-negative TER94 mutants in Drosophila photoreceptors, here we show disruption of VCP (valosin-containing protein, human TER94 ortholog), a ubiquitin-dependent segregase, causes progressive nuclear size increase. Loss of VCP function leads to accumulations of MDC1 (mediator of DNA damage checkpoint protein 1), connecting DNA damage or associated responses to enlarged nuclei. TER94 can interact with MDC1 and decreases MDC1 levels, suggesting that MDC1 is a VCP substrate. Our evidence indicates that MDC1 accumulation stabilizes p53A, leading to TER94K2A-associated nuclear size increase. Together with a previous report that p53A disrupts autophagic flux, we propose that the stabilization of p53A in TER94K2A-expressing cells likely hinders the removal of nuclear content, resulting in aberrant nuclear size increase.


Assuntos
Autofagia , Tamanho do Núcleo Celular , Núcleo Celular/metabolismo , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteína com Valosina/metabolismo , Animais , Biomarcadores/metabolismo , Olho Composto de Artrópodes , Reparo do DNA , Mitose , Transdução de Sinais , Fatores de Tempo , Proteínas Ubiquitinadas/metabolismo
4.
Sci Rep ; 10(1): 13482, 2020 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-32778728

RESUMO

Hyperphosphorylated and truncated tau variants are enriched in neuropathological aggregates in diseases known as tauopathies. However, whether the interaction of these posttranslational modifications affects tau toxicity as a whole remains unresolved. By expressing human tau with disease-related Ser/Thr residues to simulate hyperphosphorylation, we show that despite severe neurodegeneration in full-length tau, with the truncation at Asp421, the toxicity is ameliorated. Cytological and biochemical analyses reveal that hyperphosphorylated full-length tau distributes in the soma, the axon, and the axonal terminal without evident distinction, whereas the Asp421-truncated version is mostly restricted from the axonal terminal. This discrepancy is correlated with the fact that fly expressing hyperphosphorylated full-length tau, but not Asp421-cleaved one, develops axonopathy lesions, including axonal spheroids and aberrant actin accumulations. The reduced presence of hyperphosphorylated tau in the axonal terminal is corroborated with the observation that flies expressing Asp421-truncated variants showed less motor deficit, suggesting synaptic function is preserved. The Asp421 cleavage of tau is a proteolytic product commonly found in the neurofibrillary tangles. Our finding suggests the coordination of different posttranslational modifications on tau may have an unexpected impact on the protein subcellular localization and cytotoxicity, which may be valuable when considering tau for therapeutic purposes.


Assuntos
Fosforilação/genética , Proteínas tau/genética , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Animais , Axônios/metabolismo , Modelos Animais de Doenças , Drosophila , Feminino , Humanos , Masculino , Emaranhados Neurofibrilares/metabolismo , Neurônios/metabolismo , Processamento de Proteína Pós-Traducional , Tauopatias/metabolismo
5.
Int J Mol Sci ; 19(10)2018 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-30304824

RESUMO

Neuronal cell death in the central nervous system has always been a challenging process to decipher. In normal physiological conditions, neuronal cell death is restricted in the adult brain, even in aged individuals. However, in the pathological conditions of various neurodegenerative diseases, cell death and shrinkage in a specific region of the brain represent a fundamental pathological feature across different neurodegenerative diseases. In this review, we will briefly go through the general pathways of cell death and describe evidence for cell death in the context of individual common neurodegenerative diseases, discussing our current understanding of cell death by connecting with renowned pathogenic proteins, including Tau, amyloid-beta, alpha-synuclein, huntingtin and TDP-43.


Assuntos
Morte Celular , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Animais , Apoptose , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Doenças Neurodegenerativas/patologia , Neurônios/patologia
6.
J Biomed Sci ; 25(1): 54, 2018 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-29991349

RESUMO

Tau is a microtubule-associated protein that mainly localizes to the axon to stabilize axonal microtubule structure and neuronal connectivity. Tau pathology is one of the most common proteinopathies that associates with age-dependent neurodegenerative diseases including Alzheimer's disease (AD), and various Parkinsonism. Tau protein undergoes a plethora of intra-molecular modifications and some altered forms promote the production of toxic oligomeric tau and paired helical filaments, and through which further assemble into neurofibrillary tangles, also known as tauopathy. In this review, we will discuss the recent advances of the tauopathy research, primarily focusing on its association with the early axonal manifestation of axonal transport defect, axonal mitochondrial stress, autophagic vesicle accumulation and the proceeding of axon destruction, and the pathogenic Tau spreading across the synapse. Two alternative strategies either by targeting tau protein itself or by improving the age-related physiological decline are currently racing to find the hopeful treatment for tauopathy. Undoubtedly, more studies are needed to combat this devastating condition that has already affected millions of people in our aging population.


Assuntos
Doença de Alzheimer/genética , Transtornos Parkinsonianos/genética , Tauopatias/genética , Proteínas tau/genética , Doença de Alzheimer/patologia , Axônios/metabolismo , Axônios/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Mitocôndrias/genética , Mitocôndrias/patologia , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Transtornos Parkinsonianos/patologia , Sinapses/metabolismo , Sinapses/patologia , Tauopatias/patologia
7.
J Cardiovasc Dev Dis ; 3(2)2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27500162

RESUMO

Valosin-containing protein (VCP) is a highly conserved mechanoenzyme that helps maintain protein homeostasis in all cells and serves specialized functions in distinct cell types. In skeletal muscle, it is critical for myofibrillogenesis and atrophy. However, little is known about VCP's role(s) in the heart. Its functional diversity is determined by differential binding of distinct cofactors/adapters, which is likely disrupted during disease. VCP mutations cause multisystem proteinopathy (MSP), a pleiotropic degenerative disorder that involves inclusion body myopathy. MSP patients display progressive muscle weakness. They also exhibit cardiomyopathy and die from cardiac and respiratory failure, which are consistent with critical myocardial roles for the enzyme. Nonetheless, efficient models to interrogate VCP in cardiac muscle remain underdeveloped and poorly studied. Here, we investigated the significance of VCP and mutant VCP in the Drosophila heart. Cardiac-restricted RNAi-mediated knockdown of TER94, the Drosophila VCP homolog, severely perturbed myofibrillar organization and heart function in adult flies. Furthermore, expression of MSP disease-causing alleles engendered cardiomyopathy in adults and structural defects in embryonic hearts. Drosophila may therefore serve as a valuable model for examining role(s) of VCP in cardiogenesis and for identifying novel heart-specific VCP interactions, which when disrupted via mutation, contribute to or elicit cardiac pathology.

8.
PLoS Genet ; 10(9): e1004675, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25255315

RESUMO

Mutations in VCP (Valosin-containing protein), an AAA ATPase critical for ER-associated degradation, are linked to IBMPFD (Inclusion body myopathy with Paget disease and frontotemporal dementia). Using a Drosophila IBMPFD model, we have identified the ER protein Derlin-1 as a modifier of pathogenic TER94 (the fly VCP homolog) mutants. Derlin-1 binds to TER94 directly, and this interaction is essential for Derlin-1 overexpression to suppress the pathogenic TER94-induced neurodegeneration. Derlin-1 overexpression reduces the elevated ATPase activity of pathogenic TER94, implying that IBMPFD is caused by ATPase hyper-activation. Under physiological condition, Derlin-1 expression is increased upon ER stress to recruit TER94 to the ER. However, in response to severe ER stress, Derlin-1 is required for activating apoptosis to eliminate damaged cells. This pro-apoptotic response is mimicked by Derlin-1 overexpression, which elicits acute ER stress and triggers apoptosis via a novel C-terminal motif (α). As this Derlin-1-dependent cell death is negated by TER94 overexpression, we propose that while Derlin-1 and VCP work cooperatively in ER stress response, their imbalance has a role in removing cells suffering prolonged ER stress.


Assuntos
Adenosina Trifosfatases/genética , Apoptose/genética , Proteínas de Drosophila/genética , Estresse do Retículo Endoplasmático/genética , Proteínas de Membrana/genética , Mutação , Animais , Caspases/metabolismo , Modelos Animais de Doenças , Drosophila , Proteínas de Drosophila/metabolismo , Degradação Associada com o Retículo Endoplasmático/genética , Ativação Enzimática , Expressão Gênica , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mitocôndrias/ultraestrutura , Modelos Moleculares , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Fenótipo , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteína com Valosina
9.
Mol Cell Biol ; 34(9): 1594-606, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24550005

RESUMO

The rapid removal of larval midgut is a critical developmental process directed by molting hormone ecdysone during Drosophila metamorphosis. To date, it remains unclear how the stepwise events can link the onset of ecdysone signaling to the destruction of larval midgut. This study investigated whether ecdysone-induced expression of receptor protein tyrosine phosphatase PTP52F regulates this process. The mutation of the Ptp52F gene caused significant delay in larval midgut degradation. Transitional endoplasmic reticulum ATPase (TER94), a regulator of ubiquitin proteasome system, was identified as a substrate and downstream effector of PTP52F in the ecdysone signaling. The inducible expression of PTP52F at the puparium formation stage resulted in dephosphorylation of TER94 on its Y800 residue, ensuring the rapid degradation of ubiquitylated proteins. One of the proteins targeted by dephosphorylated TER94 was found to be Drosophila inhibitor of apoptosis 1 (DIAP1), which was rapidly proteolyzed in cells with significant expression of PTP52F. Importantly, the reduced level of DIAP1 in response to inducible PTP52F was essential not only for the onset of apoptosis but also for the initiation of autophagy. This study demonstrates a novel function of PTP52F in regulating ecdysone-directed metamorphosis via enhancement of autophagic and apoptotic cell death in doomed Drosophila midguts.


Assuntos
Apoptose , Autofagia , Proteínas de Drosophila/metabolismo , Drosophila/crescimento & desenvolvimento , Ecdisona/metabolismo , Metamorfose Biológica , Proteínas Tirosina Fosfatases/metabolismo , Animais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Drosophila/citologia , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Inibidoras de Apoptose/metabolismo , Larva/citologia , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Proteínas Tirosina Fosfatases/genética , Transdução de Sinais , Ubiquitinação , Proteína com Valosina
10.
Mol Biosyst ; 8(6): 1730-41, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22481368

RESUMO

IBMPFD, Inclusion body myopathy associated with Paget's disease of bone and frontotemporal dementia, is a hereditary degenerative disorder due to single missense mutations in VCP (Valosin-Containing Protein). The mechanisms of how mutations of VCP lead to IBMPFD remain mysterious. Here we utilize two-dimensional difference gel electrophoresis (2D-DIGE) combined with mass spectrometry to study the IBMPFD disorder at the protein level. With this set-up, we are able to employ comparative proteomics to analyze IBMPFD disease using Drosophila melanogaster as our disease model organism. Head proteome of transgenic D. melanogaster expressing wild type VCP is compared, respectively, with the head proteome of transgenic mutant type VCPs that correspond to human IBMPFD disease alleles (TER94(A229E), TER94(R188Q), and TER94(R152H)). Of all the proteins identified, a significant fraction of proteins altered in TER94(A229E) and TER94(R188Q) mutants belong to the same functional categories, i.e. apoptosis and metabolism. Among these, Drosophila transferrin is observed to be significantly up-regulated in mutant flies expressing TER94(A229E). A knock-down experiment suggests that fly transferrin might be a potential modifier in IBMPFD disease. The molecular analysis of IBMPFD disease may benefit from the proteomics approach which combines the advantages of high throughput analysis and the focus on protein levels.


Assuntos
Drosophila melanogaster/fisiologia , Demência Frontotemporal/metabolismo , Miosite de Corpos de Inclusão/metabolismo , Osteíte Deformante/metabolismo , Proteoma/análise , Actinas/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Apoptose/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Modelos Animais de Doenças , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Eletroforese em Gel Bidimensional , Metabolismo Energético , Feminino , Demência Frontotemporal/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Masculino , Mutação de Sentido Incorreto , Miosite de Corpos de Inclusão/genética , Osteíte Deformante/genética , Proteoma/genética , Proteoma/metabolismo , Proteômica , Espécies Reativas de Oxigênio/metabolismo , Proteína com Valosina
11.
Aging Cell ; 11(1): 93-103, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22040003

RESUMO

Aging and age-related diseases can be viewed as the result of the lifelong accumulation of stress insults. The identification of mutant strains and genes that are responsive to stress and can alter longevity profiles provides new therapeutic targets for age-related diseases. Here we reported that a Drosophila strain with reduced expression of ribose-5-phosphate isomerase (rpi), EP2456, exhibits increased resistance to oxidative stress and enhanced lifespan. In addition, the strain also displays higher levels of NADPH. The knockdown of rpi in neurons by double-stranded RNA interference recapitulated the lifespan extension and oxidative stress resistance in Drosophila. This manipulation was also found to ameliorate the effects of genetic manipulations aimed at creating a model for studying Huntington's disease by overexpression of polyglutamine in the eye, suggesting that modulating rpi levels could serve as a treatment for normal aging as well as for polyglutamine neurotoxicity.


Assuntos
Envelhecimento/metabolismo , Aldose-Cetose Isomerases/genética , Proteínas de Drosophila/genética , Via de Pentose Fosfato/genética , Peptídeos/metabolismo , Envelhecimento/genética , Aldose-Cetose Isomerases/deficiência , Animais , Modelos Animais de Doenças , Proteínas de Drosophila/deficiência , Drosophila melanogaster , Técnicas de Silenciamento de Genes , Humanos , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Longevidade , Masculino , NADP/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo , Peptídeos/genética , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo
12.
PLoS Genet ; 7(2): e1001288, 2011 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-21304887

RESUMO

Inclusion body myopathy with Paget's disease of bone and frontotemporal dementia (IBMPFD) is caused by mutations in Valosin-containing protein (VCP), a hexameric AAA ATPase that participates in a variety of cellular processes such as protein degradation, organelle biogenesis, and cell-cycle regulation. To understand how VCP mutations cause IBMPFD, we have established a Drosophila model by overexpressing TER94 (the sole Drosophila VCP ortholog) carrying mutations analogous to those implicated in IBMPFD. Expression of these TER94 mutants in muscle and nervous systems causes tissue degeneration, recapitulating the pathogenic phenotypes in IBMPFD patients. TER94-induced neurodegenerative defects are enhanced by elevated expression of wild-type TER94, suggesting that the pathogenic alleles are dominant active mutations. This conclusion is further supported by the observation that TER94-induced neurodegenerative defects require the formation of hexamer complex, a prerequisite for a functional AAA ATPase. Surprisingly, while disruptions of the ubiquitin-proteasome system (UPS) and the ER-associated degradation (ERAD) have been implicated as causes for VCP-induced tissue degeneration, these processes are not significantly affected in our fly model. Instead, the neurodegenerative defect of TER94 mutants seems sensitive to the level of cellular ATP. We show that increasing cellular ATP by independent mechanisms could suppress the phenotypes of TER94 mutants. Conversely, decreasing cellular ATP would enhance the TER94 mutant phenotypes. Taken together, our analyses have defined the nature of IBMPFD-causing VCP mutations and made an unexpected link between cellular ATP level and IBMPFD pathogenesis.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Ciclo Celular/genética , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Demência Frontotemporal/genética , Miosite de Corpos de Inclusão/genética , Osteíte Deformante/genética , Adenosina Trifosfatases/genética , Trifosfato de Adenosina/genética , Alelos , Animais , Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Demência Frontotemporal/metabolismo , Humanos , Mutação , Miosite de Corpos de Inclusão/metabolismo , Osteíte Deformante/metabolismo , Ubiquitina/metabolismo , Proteína com Valosina
13.
Hum Mol Genet ; 18(1): 164-77, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18930955

RESUMO

Hyperphosphorylation of tau at multiple sites has been implicated in the formation of neurofibrillary tangles in Alzheimer's disease; however, the relationship between toxicity and phosphorylation of tau has not been clearly elucidated. Putative tau kinases that play a role in such phosphorylation events include the proline-directed kinases glycogen synthase kinase-3beta (GSK-3beta) and cyclin-dependent kinase 5 (Cdk5), as well as nonproline-directed kinases such as microtubule affinity-regulating kinase (MARK)/PAR-1; however, whether the cascade of events linking tau phosphorylation and neurodegeneration involves sequential action of kinases as opposed to parallel pathways is still a matter of controversy. Here, we employed a well-characterized Drosophila model of tauopathy to investigate the interdependence of tau kinases in regulating the phosphorylation and toxicity of tau in vivo. We found that tau mutants resistant to phosphorylation by MARK/PAR-1 were indeed less toxic than wild-type tau; however, this was not due to their resistance to phosphorylation by GSK-3beta/Shaggy. On the contrary, a tau mutant resistant to phosphorylation by GSK-3beta/Shaggy retained substantial toxicity and was found to have increased affinity for microtubules compared with wild-type tau. The fly homologs of Cdk5/p35 did not have major effects on tau toxicity or phosphorylation in this model. These data suggest that, in addition to tau phosphorylation, microtubule binding plays a crucial role in the regulation of tau toxicity when misexpressed. These data have important implications for the understanding and interpretation of animal models of tauopathy.


Assuntos
Doença de Alzheimer/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas tau/metabolismo , Proteínas tau/toxicidade , Doença de Alzheimer/genética , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/metabolismo , Quinase 5 Dependente de Ciclina/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Olho/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Humanos , Microtúbulos/metabolismo , Mutação de Sentido Incorreto , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Proteínas tau/genética
14.
J Neurosci ; 27(5): 981-92, 2007 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-17267552

RESUMO

Mutations in human parkin have been identified in familial Parkinson's disease and in some sporadic cases. Here, we report that expression of mutant but not wild-type human parkin in Drosophila causes age-dependent, selective degeneration of dopaminergic (DA) neurons accompanied by a progressive motor impairment. Overexpression or knockdown of the Drosophila vesicular monoamine transporter, which regulates cytosolic DA homeostasis, partially rescues or exacerbates, respectively, the degenerative phenotypes caused by mutant human parkin. These results support a model in which the vulnerability of DA neurons to parkin-induced neurotoxicity results from the interaction of mutant parkin with cytoplasmic dopamine.


Assuntos
Dopamina/fisiologia , Proteínas de Drosophila/fisiologia , Mutação , Degeneração Neural/patologia , Neurônios/patologia , Ubiquitina-Proteína Ligases/fisiologia , Fatores Etários , Animais , Animais Geneticamente Modificados , Encéfalo/patologia , Contagem de Células , Modelos Animais de Doenças , Dopamina/genética , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/toxicidade , Regulação da Expressão Gênica/fisiologia , Humanos , Degeneração Neural/induzido quimicamente , Degeneração Neural/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/toxicidade
15.
Neuron ; 51(5): 549-60, 2006 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-16950154

RESUMO

Neurofibrillary tangles (NFT) containing tau are a hallmark of neurodegenerative diseases, including Alzheimer's disease (AD). NFT burden correlates with cognitive decline and neurodegeneration in AD. However, little is known about mechanisms that protect against tau-induced neurodegeneration. We used a cross species functional genomic approach to analyze gene expression in multiple brain regions in mouse, in parallel with validation in Drosophila, to identify tau modifiers, including the highly conserved protein puromycin-sensitive aminopeptidase (PSA/Npepps). PSA protected against tau-induced neurodegeneration in vivo, whereas PSA loss of function exacerbated neurodegeneration. We further show that human PSA directly proteolyzes tau in vitro. These data highlight the utility of using both evolutionarily distant species for genetic screening and functional assessment to identify modifiers of neurodegeneration. Further investigation is warranted in defining the role of PSA and other genes identified here as potential therapeutic targets in tauopathy.


Assuntos
Aminopeptidases/metabolismo , Encéfalo/enzimologia , Degeneração Neural/enzimologia , Tauopatias/genética , Proteínas tau/metabolismo , Animais , Northern Blotting , Western Blotting , Encéfalo/patologia , Drosophila , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Degeneração Neural/patologia , Emaranhados Neurofibrilares/enzimologia , Emaranhados Neurofibrilares/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Tauopatias/enzimologia , Tauopatias/patologia , Proteínas tau/genética
16.
Hum Mol Genet ; 14(11): 1475-88, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15843405

RESUMO

Hydrolethalus syndrome (HLS) is an autosomal recessive lethal malformation syndrome characterized by multiple developmental defects of fetus. We have earlier mapped and restricted the HLS region to a critical 1 cM interval on 11q23-25. The linkage disequilibrium (LD) and haplotype analyses of single nucleotide polymorphism (SNP) markers helped to further restrict the HLS locus to 476 kb between genes PKNOX2 and DDX25. An HLS associated mutation was identified in a novel regional transcript (GenBank accession no. FLJ32915), referred to here as the HYLS1 gene. The identified A to G transition results in a D211G change in the 299 amino acid polypeptide with unknown function. The HYLS1 gene shows alternative splicing and the transcript is found in multiple tissues during fetal development. In situ hybridization shows spatial and temporal distributions of transcripts in good agreement with the tissue phenotype of HLS patients. Immunostaining of in vitro expressed polypeptides from wild-type (WT) cDNA revealed cytoplasmic staining, whereas mutant polypeptides became localized in distinct nuclear structures, implying a disturbed cellular localization of the mutant protein. The Drosophila melanogaster model confirmed these findings and provides evidence for the significance of the mutation both in vitro and in vivo.


Assuntos
Anormalidades Múltiplas/genética , Genes Letais , Mutação de Sentido Incorreto , Proteínas/genética , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Sequência de Bases , Células COS , Chlorocebus aethiops , DNA Complementar , Drosophila melanogaster/genética , Desenvolvimento Fetal , Genes Recessivos , Humanos , Hibridização In Situ , Desequilíbrio de Ligação , Camundongos , Dados de Sequência Molecular , Proteínas/química , Proteínas/metabolismo , RNA Mensageiro/genética , Homologia de Sequência de Aminoácidos , Frações Subcelulares/metabolismo
17.
Hum Mol Genet ; 14(3): 357-72, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15590702

RESUMO

Huntington's disease (HD) is caused by expansion of a polyglutamine tract near the N-terminal of huntingtin. Mutant huntingtin forms aggregates in striatum and cortex, where extensive cell death occurs. We used a Drosophila polyglutamine peptide model to assess the role of specific cell death regulators in polyglutamine-induced cell death. Here, we report that polyglutamine-induced cell death was dramatically suppressed in flies lacking Dark, the fly homolog of human Apaf-1, a key regulator of apoptosis. Dark appeared to play a role in the accumulation of polyglutamine-containing aggregates. Suppression of cell death, caspase activation and aggregate formation were also observed when mutant huntingtin exon 1 was expressed in homozygous dark mutant animals. Expanded polyglutamine induced a marked increase in expression of Dark, and Dark was observed to colocalize with ubiquitinated protein aggregates. Apaf-1 also was found to colocalize with huntingtin-containing aggregates in a murine model and HD brain, suggesting a common role for Dark/Apaf-1 in polyglutamine pathogenesis in invertebrates, mice and man. These findings suggest that limiting Apaf-1 activity may alleviate both pathological protein aggregation and neuronal cell death in HD.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Olho/patologia , Peptídeos/metabolismo , Animais , Fator Apoptótico 1 Ativador de Proteases , Encéfalo/metabolismo , Encéfalo/patologia , Caspase 3 , Caspases/metabolismo , Morte Celular , Drosophila/genética , Proteínas de Drosophila/genética , Éxons , Olho/inervação , Olho/metabolismo , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Camundongos , Camundongos Mutantes , Microscopia Eletrônica , Mutação , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas/genética , Proteínas/metabolismo
18.
NeuroRx ; 2(3): 438-46, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16389307

RESUMO

Over the last two decades, a number of mutations have been identified that give rise to neurodegenerative disorders, including familial forms of Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Although in most cases sporadic cases vastly outnumber familial forms of such diseases, study of such inherited forms has the potential to provide powerful clues regarding the pathophysiological basis of neurodegeneration. One powerful approach to analyzing disease mechanisms is the development of transgenic animal models, most notably in the mouse. However, development and analysis of such models can be costly and time consuming. Development of improved transgenic technologies have contributed to the development of Drosophila models of a number of neurodegenerative disorders that have shown striking similarities to the human diseases. Moreover, genetic screens using such models have begun to unravel aspects of the pathophysiological basis of neurodegenerative disorders. Here, we provide a general overview of fly models pertinent to trinucleotide repeat expansion disorders, Alzheimer's, and Parkinson's diseases, and highlight key genetic modifiers that have been identified to date using such models.


Assuntos
Drosophila/genética , Drosophila/fisiologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Modelos Animais de Doenças , Olho/patologia , Humanos , Doença de Parkinson/genética , Doença de Parkinson/patologia , Repetições de Trinucleotídeos
19.
Neuron ; 34(4): 509-19, 2002 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-12062036

RESUMO

Pathologic alterations in the microtubule-associated protein tau have been implicated in a number of neurodegenerative disorders, including Alzheimer's disease (AD), progressive supranuclear palsy (PSP), and frontotemporal dementia (FTD). Here, we show that tau overexpression, in combination with phosphorylation by the Drosophila glycogen synthase kinase-3 (GSK-3) homolog and wingless pathway component (Shaggy), exacerbated neurodegeneration induced by tau overexpression alone, leading to neurofibrillary pathology in the fly. Furthermore, manipulation of other wingless signaling molecules downstream from shaggy demonstrated that components of the Wnt signaling pathway modulate neurodegeneration induced by tau pathology in vivo but suggested that tau phosphorylation by GSK-3beta differs from canonical Wnt effects on beta-catenin stability and TCF activity. The genetic system we have established provides a powerful reagent for identification of novel modifiers of tau-induced neurodegeneration that may serve as future therapeutic targets.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster/crescimento & desenvolvimento , Anormalidades do Olho/genética , Proteínas de Insetos/genética , Malformações do Sistema Nervoso/genética , Emaranhados Neurofibrilares/genética , Células Fotorreceptoras de Invertebrados/anormalidades , Transativadores , Fatores de Transcrição , Proteínas tau/genética , Animais , Animais Geneticamente Modificados , Apoptose/genética , Proteínas do Domínio Armadillo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestrutura , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Quinase 3 da Glicogênio Sintase , Quinases da Glicogênio Sintase , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Grupo de Alta Mobilidade/metabolismo , Humanos , Proteínas Inibidoras de Apoptose , Proteínas de Insetos/metabolismo , Proteínas de Insetos/ultraestrutura , Mutação/genética , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/patologia , Emaranhados Neurofibrilares/patologia , Emaranhados Neurofibrilares/ultraestrutura , Fenótipo , Células Fotorreceptoras de Invertebrados/patologia , Células Fotorreceptoras de Invertebrados/ultraestrutura , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transgenes/genética , beta Catenina , Proteínas tau/metabolismo , Proteínas tau/ultraestrutura
20.
Development ; 129(1): 143-54, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11782408

RESUMO

Starting with a mutation impacting photoreceptor morphogenesis, we identify here a Drosophila gene, eyes closed (eyc), as a fly homolog of p47, a protein co-factor of the p97 ATPase implicated in membrane fusion. Temporal misexpression of Eyc during rhabdomere extension early in pupal life results in inappropriate retention of normally transient adhesions between developing rhabdomeres. Later Eyc misexpression results in endoplasmic reticulum proliferation and inhibits rhodopsin transport to the developing photosensitive membrane. Loss of Eyc function results in a lethal failure of nuclear envelope assembly in early zygotic divisions. Phenotypes resulting from eyc mutations provide the first in vivo evidence for a role for p47 in membrane biogenesis.


Assuntos
Proteínas de Transporte/genética , Proteínas de Drosophila/genética , Drosophila/genética , Proteínas de Membrana/genética , Células Fotorreceptoras de Invertebrados/embriologia , Células Fotorreceptoras de Invertebrados/fisiologia , Proteínas de Transporte Vesicular , Sequência de Aminoácidos , Animais , Drosophila/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Dados de Sequência Molecular , Morfogênese/genética , Mutação , Homologia de Sequência de Aminoácidos , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA