Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Development ; 147(10)2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32345746

RESUMO

Synapses exhibit an astonishing degree of adaptive plasticity in healthy and disease states. We have investigated whether synapses also adjust to life stages imposed by novel developmental programs for which they were never molded by evolution. Under conditions in which Drosophila larvae are terminally arrested, we have characterized synaptic growth, structure and function at the neuromuscular junction (NMJ). Although wild-type larvae transition to pupae after 5 days, arrested third instar (ATI) larvae persist for 35 days, during which time NMJs exhibit extensive overgrowth in muscle size, presynaptic release sites and postsynaptic glutamate receptors. Remarkably, despite this exuberant growth, stable neurotransmission is maintained throughout the ATI lifespan through a potent homeostatic reduction in presynaptic neurotransmitter release. Arrest of the larval stage in stathmin mutants also reveals a degree of progressive instability and neurodegeneration that was not apparent during the typical larval period. Hence, an adaptive form of presynaptic depression stabilizes neurotransmission during an extended developmental period of unconstrained synaptic growth. More generally, the ATI manipulation provides a powerful system for studying neurodegeneration and plasticity across prolonged developmental timescales.


Assuntos
Drosophila/crescimento & desenvolvimento , Drosophila/genética , Larva/crescimento & desenvolvimento , Larva/genética , Depressão Sináptica de Longo Prazo/genética , Degeneração Neural/genética , Junção Neuromuscular/crescimento & desenvolvimento , Animais , Axônios/patologia , Proteínas de Drosophila/genética , Feminino , Homeostase/genética , Masculino , Mutação , Junção Neuromuscular/metabolismo , Interferência de RNA , Proteínas Smad Reguladas por Receptor/genética , Estatmina/genética , Sinapses/metabolismo , Transmissão Sináptica/genética
2.
J Cell Sci ; 131(18)2018 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-30154211

RESUMO

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons, resulting in progressive locomotor dysfunction. Identification of genes required for the maintenance of these neurons should help to identify potential therapeutic targets. However, little is known regarding the factors that render dopaminergic neurons selectively vulnerable to PD. Here, we show that Drosophila melanogaster scarlet mutants exhibit an age-dependent progressive loss of dopaminergic neurons, along with subsequent locomotor defects and a shortened lifespan. Knockdown of Scarlet specifically within dopaminergic neurons is sufficient to produce this neurodegeneration, demonstrating a unique role for Scarlet beyond its well-characterized role in eye pigmentation. Both genetic and pharmacological manipulation of the kynurenine pathway rescued loss of dopaminergic neurons by promoting synthesis of the free radical scavenger kynurenic acid (KYNA) and limiting the production of the free radical generator 3-hydroxykynurenine (3-HK). Finally, we show that expression of wild-type Scarlet is neuroprotective in a model of PD, suggesting that manipulating kynurenine metabolism may be a potential therapeutic option in treating PD.This article has an associated First Person interview with the first author of the paper.


Assuntos
Drosophila melanogaster/metabolismo , Doenças Neurodegenerativas/genética , Doença de Parkinson/genética , Animais , Humanos , Doenças Neurodegenerativas/patologia , Doença de Parkinson/patologia
3.
Anesthesiology ; 133(4): 839-851, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32773682

RESUMO

BACKGROUND: General anesthetics influence mitochondrial homeostasis, placing individuals with mitochondrial disorders and possibly carriers of recessive mitochondrial mutations at increased risk of perioperative complications. In Drosophila, mutations in the ND23 subunit of complex I of the mitochondrial electron transport chain-analogous to mammalian NDUFS8-replicate key characteristics of Leigh syndrome, an inherited mitochondrial disorder. The authors used the ND23 mutant for testing the hypothesis that anesthetics have toxic potential in carriers of mitochondrial mutations. METHODS: The authors exposed wild-type flies and ND23 mutant flies to behaviorally equivalent doses of isoflurane or sevoflurane in 5%, 21%, or 75% oxygen. The authors used percent mortality (mean ± SD, n ≥ 3) at 24 h after exposure as a readout of toxicity and changes in gene expression to investigate toxicity mechanisms. RESULTS: Exposure of 10- to 13-day-old male ND23 flies to isoflurane in 5%, 21%, or 75% oxygen resulted in 16.0 ± 14.9% (n = 10), 48.2 ± 16.1% (n = 9), and 99.2 ± 2.0% (n = 10) mortality, respectively. Comparable mortality was observed in females. In contrast, under the same conditions, mortality was less than 5% for all male and female groups exposed to sevoflurane, except 10- to 13-day-old male ND23 flies with 9.6 ± 8.9% (n = 16) mortality. The mortality of 10- to 13-day-old ND23 flies exposed to isoflurane was rescued by neuron- or glia-specific expression of wild-type ND23. Isoflurane and sevoflurane differentially affected expression of antioxidant genes in 10- to 13-day-old ND23 flies. ND23 flies had elevated mortality from paraquat-induced oxidative stress compared with wild-type flies. The mortality of heterozygous ND23 flies exposed to isoflurane in 75% oxygen increased with age, resulting in 54.0 ± 19.6% (n = 4) mortality at 33 to 39 days old, and the percent mortality varied in different genetic backgrounds. CONCLUSIONS: Mutations in the mitochondrial complex I subunit ND23 increase susceptibility to isoflurane-induced toxicity and to oxidative stress in Drosophila. Asymptomatic flies that carry ND23 mutations are sensitized to hyperoxic isoflurane toxicity by age and genetic background.


Assuntos
Anestésicos Inalatórios/toxicidade , Complexo I de Transporte de Elétrons/genética , Isoflurano/toxicidade , Mitocôndrias/genética , Mutação/genética , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Envelhecimento/patologia , Animais , Animais Geneticamente Modificados , Drosophila , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Mutação/efeitos dos fármacos , Sevoflurano/toxicidade
4.
J Neurogenet ; : 1-5, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39039773
5.
Proc Natl Acad Sci U S A ; 112(39): E5427-33, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26351672

RESUMO

A key feature of many neurodegenerative diseases is the accumulation and subsequent aggregation of misfolded proteins. Recent studies have highlighted the transcellular propagation of protein aggregates in several major neurodegenerative diseases, although the precise mechanisms underlying this spreading and how it relates to disease pathology remain unclear. Here we use a polyglutamine-expanded form of human huntingtin (Htt) with a fluorescent tag to monitor the spreading of aggregates in the Drosophila brain in a model of Huntington's disease. Upon expression of this construct in a defined subset of neurons, we demonstrate that protein aggregates accumulate at synaptic terminals and progressively spread throughout the brain. These aggregates are internalized and accumulate within other neurons. We show that Htt aggregates cause non-cell-autonomous pathology, including loss of vulnerable neurons that can be prevented by inhibiting endocytosis in these neurons. Finally we show that the release of aggregates requires N-ethylmalemide-sensitive fusion protein 1, demonstrating that active release and uptake of Htt aggregates are important elements of spreading and disease progression.


Assuntos
Encéfalo/fisiologia , Drosophila/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Doenças Neurodegenerativas/fisiopatologia , Agregados Proteicos/fisiologia , Transcitose/fisiologia , Animais , Proteínas de Drosophila , Proteína Huntingtina , Imuno-Histoquímica , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/genética , Peptídeos/genética , Expansão das Repetições de Trinucleotídeos/genética
6.
Proc Natl Acad Sci U S A ; 110(19): E1752-60, 2013 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-23613578

RESUMO

A growing body of evidence in humans implicates chronic activation of the innate immune response in the brain as a major cause of neuropathology in various neurodegenerative conditions, although the mechanisms remain unclear. In an unbiased genetic screen for mutants exhibiting neurodegeneration in Drosophila, we have recovered a mutation of dnr1 (defense repressor 1), a negative regulator of the Imd (immune deficiency) innate immune-response pathway. dnr1 mutants exhibit shortened lifespan and progressive, age-dependent neuropathology associated with activation of the Imd pathway and elevated expression of AMP (antimicrobial peptide) genes. To test the hypothesis that overactivation of innate immune-response pathways in the brain is responsible for neurodegeneration, we demonstrated that direct bacterial infection in the brain of wild-type flies also triggers neurodegeneration. In both cases, neurodegeneration is dependent on the NF-κB transcription factor, Relish. Moreover, we found that neural overexpression of individual AMP genes is sufficient to cause neurodegeneration. These results provide a mechanistic link between innate immune responses and neurodegeneration and may have important implications for the role of neuroinflammation in human neurodegenerative diseases as well.


Assuntos
Encéfalo/patologia , Proteínas de Drosophila/genética , Drosophila melanogaster/imunologia , Imunidade Inata , Mutação , Proteínas Repressoras/genética , Envelhecimento , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Apoptose , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Encéfalo/imunologia , Encéfalo/microbiologia , Morte Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/microbiologia , Genótipo , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/patologia , Neurônios/patologia , Fenótipo , Fatores de Transcrição/metabolismo
7.
Proc Natl Acad Sci U S A ; 110(44): E4152-9, 2013 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-24127584

RESUMO

Traumatic brain injury (TBI) is a substantial health issue worldwide, yet the mechanisms responsible for its complex spectrum of pathologies remains largely unknown. To investigate the mechanisms underlying TBI pathologies, we developed a model of TBI in Drosophila melanogaster. The model allows us to take advantage of the wealth of experimental tools available in flies. Closed head TBI was inflicted with a mechanical device that subjects flies to rapid acceleration and deceleration. Similar to humans with TBI, flies with TBI exhibited temporary incapacitation, ataxia, activation of the innate immune response, neurodegeneration, and death. Our data indicate that TBI results in death shortly after a primary injury only if the injury exceeds a certain threshold and that age and genetic background, but not sex, substantially affect this threshold. Furthermore, this threshold also appears to be dependent on the same cellular and molecular mechanisms that control normal longevity. This study demonstrates the potential of flies for providing key insights into human TBI that may ultimately provide unique opportunities for therapeutic intervention.


Assuntos
Aceleração/efeitos adversos , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Drosophila melanogaster , Imunidade Inata/fisiologia , Longevidade/fisiologia , Fatores Etários , Análise de Variância , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/imunologia , Feminino , Masculino , Reação em Cadeia da Polimerase em Tempo Real , Fatores Sexuais
8.
Proc Natl Acad Sci U S A ; 109(11): E648-55, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22355119

RESUMO

Although the complexity and circuitry of nervous systems undergo evolutionary change, we lack understanding of the general principles and specific mechanisms through which it occurs. The Drosophila larval neuromuscular junction (NMJ), which has been widely used for studies of synaptic development and function, is also an excellent system for studies of synaptic evolution because the genus spans >40 Myr of evolution and the same identified synapse can be examined across the entire phylogeny. We have now characterized morphology of the NMJ on muscle 4 (NMJ4) in >20 species of Drosophila. Although there is little variation within a species, NMJ morphology and complexity vary extensively between species. We find no significant correlation between NMJ phenotypes and phylogeny for the species examined, suggesting that drift alone cannot explain the phenotypic variation and that selection likely plays an important role. However, the nature of the selective pressure is still unclear because basic parameters of synaptic function remain uniform. Whatever the mechanism, NMJ morphology is evolving rapidly in comparison with other morphological features because NMJ phenotypes differ even between several sibling species pairs. The discovery of this unexpectedly extensive divergence in NMJ morphology among Drosophila species provides unique opportunities to investigate mechanisms that regulate synaptic growth; the interrelationships between synaptic morphology, neural function, and behavior; and the evolution of nervous systems and behavior in natural populations.


Assuntos
Evolução Biológica , Drosophila/anatomia & histologia , Junção Neuromuscular/anatomia & histologia , Animais , Drosophila/genética , Deriva Genética , Larva/anatomia & histologia , Tamanho do Órgão , Fenótipo , Filogenia , Terminações Pré-Sinápticas , Especificidade da Espécie
9.
J Neurosci ; 32(40): 13776-86, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23035089

RESUMO

The Drosophila larval neuromuscular junction (NMJ) is a powerful system for the genetic and molecular analysis of neuronal excitability, synaptic transmission, and synaptic development. However, its use for studying age-dependent processes, such as maintenance of neuronal viability and synaptic stability, are temporally limited by the onset of pupariation and metamorphosis. Here we characterize larval NMJ growth, growth regulation, structure, and function in a developmental variant with an extended third instar (ETI). RNAi-knockdown of the prothoracicotropic hormone receptor, torso, in the ring gland of developing larvae leaves the timing of first and second instar molts largely unchanged, but triples duration of the third instar from 3 to 9.5 d (McBrayer et al., 2007; Rewitz et al., 2009). During this ETI period, NMJs undergo additional growth (adding >50 boutons/NMJ), and this growth remains under the control of the canonical regulators Highwire and the TGFß/BMP pathway. NMJ growth during the ETI period occurs via addition of new branches, satellite boutons, and interstitial boutons, and continues even after muscle growth levels off. Throughout the ETI, organization of synapses and active zones remains normal, and synaptic transmission is unchanged. These results establish the ETI larval system as a viable model for studying motor neuron diseases and for investigating time-dependent effects of perturbations that impair mechanisms of neuroprotection, synaptic maintenance, and response to neural injury.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Sistema Nervoso/crescimento & desenvolvimento , Junção Neuromuscular/crescimento & desenvolvimento , Terminações Pré-Sinápticas/fisiologia , Animais , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Hormônios de Inseto/fisiologia , Proteínas de Insetos/antagonistas & inibidores , Proteínas de Insetos/genética , Larva , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/fisiologia , Muda/fisiologia , Terminações Pré-Sinápticas/ultraestrutura , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/biossíntese , Receptores Proteína Tirosina Quinases/genética , Receptores de Esteroides/antagonistas & inibidores , Receptores de Esteroides/genética , Fatores de Tempo
10.
Proc Natl Acad Sci U S A ; 107(12): 5617-21, 2010 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-20212103

RESUMO

To signal properly, excitable cells must establish and maintain the correct balance of various types of ion channels that increase or decrease membrane excitability. The mechanisms by which this balance is regulated remain largely unknown. Here, we describe a regulatory mechanism uncovered by a Drosophila behavioral mutant, down and out (dao). At elevated temperatures, dao loss-of-function mutants exhibit seizures associated with spontaneous bursts of neural activity. This phenotype closely resembles that of seizure mutations, which impair activity of ether-a-go-go-related gene (Erg)-type potassium channels. Conversely, neural over-expression of wild-type Dao confers dominant temperature-sensitive paralysis with kinetics reminiscent of paralytic sodium-channel mutants. The over-expression phenotype of dao is suppressed in a seizure mutant background, suggesting that Dao acts by an effect on Erg channels. In support of this hypothesis, functional expression of Erg channels in a heterologous system is dependent on the presence of Dao. These results indicate that Dao has an important role in establishing the proper level of neuronal membrane excitability by regulating functional expression of Erg channels.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila/genética , Drosophila/fisiologia , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/fisiologia , Animais , Animais Geneticamente Modificados , Comportamento Animal , Mapeamento Cromossômico , Feminino , Expressão Gênica , Genes de Insetos , Técnicas In Vitro , Mutação , Oócitos/metabolismo , Fenótipo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
11.
Genetics ; 223(3)2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36683334

RESUMO

Traumatic brain injury (TBI) outcomes vary greatly among individuals, but most of the variation remains unexplained. Using a Drosophila melanogaster TBI model and 178 genetically diverse lines from the Drosophila Genetic Reference Panel (DGRP), we investigated the role that genetic variation plays in determining TBI outcomes. Following injury at 20-27 days old, DGRP lines varied considerably in mortality within 24 h ("early mortality"). Additionally, the disparity in early mortality resulting from injury at 20-27 vs 0-7 days old differed among DGRP lines. These data support a polygenic basis for differences in TBI outcomes, where some gene variants elicit their effects by acting on aging-related processes. Our genome-wide association study of DGRP lines identified associations between single nucleotide polymorphisms in Lissencephaly-1 (Lis-1) and Patronin and early mortality following injury at 20-27 days old. Lis-1 regulates dynein, a microtubule motor required for retrograde transport of many cargoes, and Patronin protects microtubule minus ends against depolymerization. While Patronin mutants did not affect early mortality, Lis-1 compound heterozygotes (Lis-1x/Lis-1y) had increased early mortality following injury at 20-27 or 0-7 days old compared with Lis-1 heterozygotes (Lis-1x/+), and flies that survived 24 h after injury had increased neurodegeneration but an unaltered lifespan, indicating that Lis-1 affects TBI outcomes independently of effects on aging. These data suggest that Lis-1 activity is required in the brain to ameliorate TBI outcomes through effects on axonal transport, microtubule stability, and other microtubule proteins, such as tau, implicated in chronic traumatic encephalopathy, a TBI-associated neurodegenerative disease in humans.


Assuntos
Lesões Encefálicas Traumáticas , Proteínas de Drosophila , Lisencefalia , Doenças Neurodegenerativas , Animais , Humanos , Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Estudo de Associação Genômica Ampla , Lesões Encefálicas Traumáticas/genética , Mutação , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
12.
J Neurogenet ; 26(3-4): 317-27, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22882183

RESUMO

We describe the characterization of m4, an autosomal recessive, temperature-sensitive paralytic mutant in Drosophila that is associated with shortened lifespan and neurodegeneration. Deletion mapping places the mutation in the gene encoding the glycolytic enzyme, Aldolase. The mutant enzyme contains a single amino acid substitution, which results in decreased steady-state levels of Aldolase with a consequent reduction in adenosine triphosphate (ATP) levels. Transgenic-rescue experiments with a genomic construct containing the entire Aldolase gene confirm that paralysis, reduced lifespan, and neurodegeneration all result from the same mutation. Tissue-specific rescue and RNA interference (RNAi) knockdown experiments indicate that Aldolase function (and presumably glycolysis) is important both in neurons and in glia for normal lifespan and neuronal maintenance over time. Impaired glycolysis in neurons can apparently be rescued in part by glycolytically active glia. However, this rescue may depend on the exact physiological state of the neurons and may also vary in different subsets of neurons. Further studies of m4 and related mutants in Drosophila should help elucidate the connections between energy production and utilization in glia and neurons and lead to better understanding of how metabolic defects impair neuronal function and maintenance.


Assuntos
Frutose-Bifosfato Aldolase/genética , Longevidade/genética , Mutação/genética , Degeneração Neural/genética , Paralisia/genética , Distúrbios Somatossensoriais/genética , Trifosfato de Adenosina/metabolismo , Fatores Etários , Animais , Animais Geneticamente Modificados , Clonagem Molecular , Drosophila , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Paralisia/complicações , Fenótipo , RNA Interferente Pequeno/farmacologia , Distúrbios Somatossensoriais/complicações
13.
Nature ; 434(7037): 1087-92, 2005 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-15858564

RESUMO

Most of us sleep 7-8 h per night, and if we are deprived of sleep our performance suffers greatly; however, a few do well with just 3-4 h of sleep-a trait that seems to run in families. Determining which genes underlie this phenotype could shed light on the mechanisms and functions of sleep. To do so, we performed mutagenesis in Drosophila melanogaster, because flies also sleep for many hours and, when sleep deprived, show sleep rebound and performance impairments. By screening 9,000 mutant lines, we found minisleep (mns), a line that sleeps for one-third of the wild-type amount. We show that mns flies perform normally in a number of tasks, have preserved sleep homeostasis, but are not impaired by sleep deprivation. We then show that mns flies carry a point mutation in a conserved domain of the Shaker gene. Moreover, after crossing out genetic modifiers accumulated over many generations, other Shaker alleles also become short sleepers and fail to complement the mns phenotype. Finally, we show that short-sleeping Shaker flies have a reduced lifespan. Shaker, which encodes a voltage-dependent potassium channel controlling membrane repolarization and transmitter release, may thus regulate sleep need or efficiency.


Assuntos
Drosophila melanogaster/metabolismo , Mutação Puntual/genética , Canais de Potássio/metabolismo , Privação do Sono/genética , Privação do Sono/fisiopatologia , Sequência de Aminoácidos , Animais , Comportamento Animal/fisiologia , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Sequência Conservada , Cruzamentos Genéticos , Escuridão , Proteínas de Drosophila , Drosophila melanogaster/genética , Feminino , Genes Recessivos/genética , Teste de Complementação Genética , Homeostase , Humanos , Luz , Longevidade/genética , Longevidade/fisiologia , Masculino , Mamíferos/fisiologia , Dados de Sequência Molecular , Atividade Motora/fisiologia , Fenótipo , Canais de Potássio/química , Canais de Potássio/genética , Estrutura Terciária de Proteína , Superfamília Shaker de Canais de Potássio , Sono/genética , Sono/fisiologia , Fatores de Tempo , Cromossomo X/genética
14.
Proc Natl Acad Sci U S A ; 105(20): 7327-32, 2008 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-18480253

RESUMO

Although deficiencies in the retromer sorting pathway have been linked to late-onset Alzheimer's disease, whether these deficiencies underlie the disease remains unknown. Here we characterized two genetically modified animal models to test separate but related questions about the effects that retromer deficiency has on the brain. First, testing for cognitive defects, we investigated retromer-deficient mice and found that they develop hippocampal-dependent memory and synaptic dysfunction, which was associated with elevations in endogenous Abeta peptide. Second, testing for neurodegeneration and amyloid deposits, we investigated retromer-deficient flies expressing human wild-type amyloid precursor protein (APP) and human beta-site APP-cleaving enzyme (BACE) and found that they develop neuronal loss and human Abeta aggregates. By recapitulating features of the disease, these animal models suggest that retromer deficiency observed in late-onset Alzheimer's disease can contribute to disease pathogenesis.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/química , Hipocampo/patologia , Doenças Neurodegenerativas/patologia , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Drosophila/metabolismo , Eletrofisiologia , Heterozigoto , Hipocampo/metabolismo , Humanos , Camundongos , Camundongos Knockout , Modelos Biológicos , Doenças Neurodegenerativas/metabolismo
15.
MicroPubl Biol ; 20212021.
Artigo em Inglês | MEDLINE | ID: mdl-34278243

RESUMO

Blunt force injuries are a significant cause of disability and death worldwide. Here, we describe a Drosophila melanogaster model of blunt force injury that can be used to investigate cellular and molecular mechanisms that underlie the short-term and long-term effects of injuries sustained at a juvenile stage of development. Injuries inflicted in late third-instar larvae using the spring-based High-Impact Trauma (HIT) device robustly activated the humoral defense response process of melanization and caused larval and pupal lethality. Additionally, adults that developed from injured larvae had reduced lifespans, indicating that cellular and molecular mechanisms activated by blunt force injuries in larvae persist through metamorphosis and adult development. Previously, the HIT device has been used to investigate genetic and environmental factors underlying mechanisms that contribute to consequences of blunt force injuries incurred in adult flies. This work expands use of the HIT device to a juvenile stage of development, offering the opportunity to investigate whether the consequences of blunt force injuries involve different factors and mechanisms at different stages of development.

16.
MicroPubl Biol ; 20212021.
Artigo em Inglês | MEDLINE | ID: mdl-34723144

RESUMO

Traumatic brain injury (TBI) frequently leads to non-neurological consequences such as intestinal permeability. The beta-blocker drug labetalol, which inhibits binding of catecholamine neurotransmitters to adrenergic receptors, reduces intestinal permeability in a rat TBI model. Using a Drosophila melanogaster TBI model, we previously found a strong positive correlation between intestinal permeability and mortality within 24 hours of TBI in a standard laboratory line (w1118 ) and across genetically diverse inbred lines from the Drosophila Genetic Reference Panel (DGRP). Here, we report that feeding injured w1118 flies the beta-blockers labetalol and metoprolol reduced intestinal permeability and mortality. Additionally, metoprolol reduced intestinal permeability when 18 DGRP fly lines were analyzed in aggregate, but neither beta-blocker affected mortality. These data indicate that the mechanism underlying disruption of the intestinal barrier by adrenergic signaling following TBI is conserved between humans and flies and that mortality following TBI in flies is not strictly dependent on disruption of the intestinal barrier. Thus, the fly TBI model is useful for shedding light on the mechanism and consequences of adrenergic signaling between the brain and intestine following TBI in humans.

17.
PLoS One ; 16(10): e0258873, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34699541

RESUMO

Traumatic brain injury (TBI) is a common neurological disorder whose outcomes vary widely depending on a variety of environmental factors, including diet. Using a Drosophila melanogaster TBI model that reproduces key aspects of TBI in humans, we previously found that the diet consumed immediately following a primary brain injury has a substantial effect on the incidence of mortality within 24 h (early mortality). Flies that receive equivalent primary injuries have a higher incidence of early mortality when fed high-carbohydrate diets versus water. Here, we report that flies fed high-fat ketogenic diet (KD) following TBI exhibited early mortality that was equivalent to that of flies fed water and that flies protected from early mortality by KD continued to show survival benefits weeks later. KD also has beneficial effects in mammalian TBI models, indicating that the mechanism of action of KD is evolutionarily conserved. To probe the mechanism, we examined the effect of KD in flies mutant for Eip75B, an ortholog of the transcription factor PPARγ (peroxisome proliferator-activated receptor gamma) that contributes to the mechanism of action of KD and has neuroprotective effects in mammalian TBI models. We found that the incidence of early mortality of Eip75B mutant flies was higher when they were fed KD than when they were fed water following TBI. These data indicate that Eip75B/PPARγ is necessary for the beneficial effects of KD following TBI. In summary, this work provides the first evidence that KD activates PPARγ to reduce deleterious outcomes of TBI and it demonstrates the utility of the fly TBI model for dissecting molecular pathways that contribute to heterogeneity in TBI outcomes.


Assuntos
Lesões Encefálicas Traumáticas/terapia , Proteínas de Ligação a DNA/metabolismo , Dieta Cetogênica , Proteínas de Drosophila/metabolismo , Fatores de Transcrição/metabolismo , Animais , Lesões Encefálicas Traumáticas/metabolismo , Modelos Animais de Doenças , Drosophila melanogaster
18.
G3 (Bethesda) ; 10(9): 3109-3119, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32631949

RESUMO

Neuroinflammation is a major pathophysiological feature of traumatic brain injury (TBI). Early and persistent activation of innate immune response signaling pathways by primary injuries is associated with secondary cellular injuries that cause TBI outcomes to change over time. We used a Drosophila melanogaster model to investigate the role of antimicrobial peptides (AMPs) in acute and chronic outcomes of closed-head TBI. AMPs are effectors of pathogen and stress defense mechanisms mediated by the evolutionarily conserved Toll and Immune-deficiency (Imd) innate immune response pathways that activate Nuclear Factor kappa B (NF-κB) transcription factors. Here, we analyzed the effect of null mutations in 10 of the 14 known Drosophila AMP genes on TBI outcomes. We found that mutation of Metchnikowin (Mtk) was unique in protecting flies from mortality within the 24 h following TBI under two diet conditions that produce different levels of mortality. In addition, Mtk mutants had reduced behavioral deficits at 24 h following TBI and increased lifespan either in the absence or presence of TBI. Using a transcriptional reporter of gene expression, we found that TBI increased Mtk expression in the brain. Quantitative analysis of mRNA in whole flies revealed that expression of other AMPs in the Toll and Imd pathways as well as NF-κB transcription factors were not altered in Mtk mutants. Overall, these results demonstrate that Mtk plays an infection-independent role in the fly nervous system, and TBI-induced expression of Mtk in the brain activates acute and chronic secondary injury pathways that are also activated during normal aging.


Assuntos
Lesões Encefálicas Traumáticas , Proteínas de Drosophila , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Lesões Encefálicas Traumáticas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas Citotóxicas Formadoras de Poros
19.
Genetics ; 216(4): 1117-1136, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33109529

RESUMO

Traumatic brain injury (TBI) pathologies are caused by primary and secondary injuries. Primary injuries result from physical damage to the brain, and secondary injuries arise from cellular responses to primary injuries. A characteristic cellular response is sustained activation of inflammatory pathways commonly mediated by nuclear factor-κB (NF-κB) transcription factors. Using a Drosophila melanogaster TBI model, we previously found that the main proximal transcriptional response to primary injuries is triggered by activation of Toll and Imd innate immune response pathways that engage NF-κB factors Dif and Relish (Rel), respectively. Here, we found by mass spectrometry that Rel protein level increased in fly heads at 4-8 hr after TBI. To investigate the necessity of Rel for secondary injuries, we generated a null allele, Reldel , by CRISPR/Cas9 editing. When heterozygous but not homozygous, the Reldel mutation reduced mortality at 24 hr after TBI and increased the lifespan of injured flies. Additionally, the effect of heterozygosity for Reldel on mortality was modulated by genetic background and diet. To identify genes that facilitate effects of Reldel on TBI outcomes, we compared genome-wide mRNA expression profiles of uninjured and injured +/+, +/Reldel , and Reldel /Reldel flies at 4 hr following TBI. Only a few genes changed expression more than twofold in +/Reldel flies relative to +/+ and Reldel /Reldel flies, and they were not canonical innate immune response genes. Therefore, Rel is necessary for TBI-induced secondary injuries but in complex ways involving Rel gene dose, genetic background, diet, and possibly small changes in expression of innate immune response genes.


Assuntos
Lesões Encefálicas Traumáticas/genética , Proteínas de Drosophila/genética , Fatores de Transcrição/genética , Animais , Lesões Encefálicas Traumáticas/imunologia , Drosophila melanogaster , Patrimônio Genético , Heterozigoto , Imunidade Inata , Mutação , Transcriptoma
20.
Genetics ; 178(2): 947-56, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18245348

RESUMO

Genetic factors are known to contribute to seizure susceptibility, although the long-term effects of these predisposing factors on neuronal viability remain unclear. To examine the consequences of genetic factors conferring increased seizure susceptibility, we surveyed a class of Drosophila mutants that exhibit seizures and paralysis following mechanical stimulation. These bang-sensitive seizure mutants exhibit shortened life spans and age-dependent neurodegeneration. Because the increased seizure susceptibility in these mutants likely results from altered metabolism and since the Na(+)/K(+) ATPase consumes the majority of ATP in neurons, we examined the effect of ATPalpha mutations in combination with bang-sensitive mutations. We found that double mutants exhibit strikingly reduced life spans and age-dependent uncoordination and inactivity. These results emphasize the importance of proper cellular metabolism in maintaining both the activity and viability of neurons.


Assuntos
Drosophila/genética , Predisposição Genética para Doença , Mutação , Convulsões/genética , Animais , Drosophila/crescimento & desenvolvimento , Longevidade , Sistema Nervoso/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA