Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
PLoS Biol ; 17(2): e2006409, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30759083

RESUMEN

Dysregulation of sleep and feeding has widespread health consequences. Despite extensive epidemiological evidence for interactions between sleep and metabolic function, little is known about the neural or molecular basis underlying the integration of these processes. D. melanogaster potently suppress sleep in response to starvation, and powerful genetic tools allow for mechanistic investigation of sleep-metabolism interactions. We have previously identified neurons expressing the neuropeptide leucokinin (Lk) as being required for starvation-mediated changes in sleep. Here, we demonstrate an essential role for Lk neuropeptide in metabolic regulation of sleep. The activity of Lk neurons is modulated by feeding, with reduced activity in response to glucose and increased activity under starvation conditions. Both genetic silencing and laser-mediated microablation localize Lk-dependent sleep regulation to a single pair of Lk neurons within the Lateral Horn (LHLK neurons). A targeted screen identified a role for 5' adenosine monophosphate-activated protein kinase (AMPK) in starvation-modulated changes in sleep. Knockdown of AMPK in Lk neurons suppresses sleep and increases LHLK neuron activity in fed flies, phenocopying the starvation state. Further, we find a requirement for the Lk receptor in the insulin-producing cells (IPCs), suggesting LHLK-IPC connectivity is critical for sleep regulation under starved conditions. Taken together, these findings localize feeding-state-dependent regulation of sleep to a single pair of neurons within the fruit fly brain and provide a system for investigating the cellular basis of sleep-metabolism interactions.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neuronas/metabolismo , Neuropéptidos/metabolismo , Sueño/fisiología , Adenilato Quinasa/metabolismo , Animales , Terapia por Láser , Inanición , Vigilia
2.
J Biol Chem ; 293(45): 17442-17453, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30257867

RESUMEN

L1 cell adhesion molecule (L1CAM) is well-known for its importance in nervous system development and cancer progression. In addition to its role as a plasma membrane protein in cytoskeletal organization, recent in vitro studies have revealed that both transmembrane and cytosolic fragments of proteolytically cleaved vertebrate L1CAM translocate to the nucleus. In vitro studies indicate that nuclear L1CAM affects genes with functions in DNA post-replication repair, cell cycle control, and cell migration and differentiation, but its in vivo role and how its nuclear levels are regulated is less well-understood. Here, we report that mutations in the conserved ankyrin-binding domain affect nuclear levels of the sole Drosophila homolog neuroglian (Nrg) and that it also has a noncanonical role in regulating transcript levels of the oncogene Myc in the adult nervous system. We further show that altered nuclear levels of Nrg correlate with altered transcript levels of Myc in neurons, similar to what has been reported for human glioblastoma stem cells. However, whereas previous in vitro studies suggest that increased nuclear levels of L1CAM promote tumor cell survival, we found here that elevated levels of nuclear Nrg in neurons are associated with increased sensitivity to oxidative stress and reduced life span of adult animals. We therefore conclude that these findings are of potential relevance to the management of neurodegenerative diseases associated with oxidative stress and cancer.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Secuencias de Aminoácidos , Animales , Moléculas de Adhesión Celular Neuronal/genética , Núcleo Celular/patología , Proteínas de Drosophila/genética , Drosophila melanogaster , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Molécula L1 de Adhesión de Célula Nerviosa/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Proteínas Proto-Oncogénicas c-myc/genética
3.
FASEB J ; 29(3): 1011-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25466886

RESUMEN

Nicotinic acetylcholine receptors (nAChRs) play a pivotal role in synaptic transmission of neuronal signaling pathways and are fundamentally involved in neuronal disorders, including Alzheimer's disease, Parkinson's disease, and schizophrenia. In vertebrates, cholinergic pathways can be selectively inhibited by α-conotoxins; we show that in the model organism Drosophila, the cholinergic component of the giant fiber system is inhibited by α-conotoxins MII, AuIB, BuIA, EI, PeIA, and ImI. The injection of 45 pmol/fly of each toxin dramatically decreases the response of the giant fiber to dorsal longitudinal muscle (GF-DLM) connection to 20 ± 13.9% for MII; 26 ± 13.7% for AuIB, 12 ± 9.9% for BuIA, 30 ± 11.3% for EI, 1 ± 1% for PeIA, and 34 ± 15.4% for ImI. Through bioassay-guided fractionation of the venom of Conus brunneus, we found BruIB, an α-conotoxin that inhibits Drosophila nicotinic receptors but not its vertebrate counterparts. GF-DLM responses decreased to 43.7 ± 8.02% on injection of 45 pmol/fly of BruIB. We manipulated the Dα7 nAChR to mimic the selectivity of its vertebrate counterpart by placing structurally guided point mutations in the conotoxin-binding site. This manipulation rendered vertebrate-like behavior in the Drosophila system, enhancing the suitability of Drosophila as an in vivo tool to carry out studies related to human neuronal diseases. .


Asunto(s)
Acetilcolina/farmacología , Conotoxinas/farmacología , Drosophila melanogaster/metabolismo , Antagonistas Nicotínicos/farmacología , Transmisión Sináptica/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/química , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente/genética , Animales Modificados Genéticamente/crecimiento & desarrollo , Animales Modificados Genéticamente/metabolismo , Sitios de Unión , Colinérgicos/farmacología , Caracol Conus/química , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Femenino , Células Gigantes/citología , Células Gigantes/efectos de los fármacos , Células Gigantes/metabolismo , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Mutación/genética , Oocitos/citología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Fragmentos de Péptidos/farmacología , Conformación Proteica , Homología de Secuencia de Aminoácido , Xenopus laevis/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/genética
4.
PLoS Biol ; 11(4): e1001537, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23610557

RESUMEN

The precise control of synaptic connectivity is essential for the development and function of neuronal circuits. While there have been significant advances in our understanding how cell adhesion molecules mediate axon guidance and synapse formation, the mechanisms controlling synapse maintenance or plasticity in vivo remain largely uncharacterized. In an unbiased RNAi screen we identified the Drosophila L1-type CAM Neuroglian (Nrg) as a central coordinator of synapse growth, function, and stability. We demonstrate that the extracellular Ig-domains and the intracellular Ankyrin-interaction motif are essential for synapse development and stability. Nrg binds to Ankyrin2 in vivo and mutations reducing the binding affinities to Ankyrin2 cause an increase in Nrg mobility in motoneurons. We then demonstrate that the Nrg-Ank2 interaction controls the balance of synapse growth and stability at the neuromuscular junction. In contrast, at a central synapse, transsynaptic interactions of pre- and postsynaptic Nrg require a dynamic, temporal and spatial, regulation of the intracellular Ankyrin-binding motif to coordinate pre- and postsynaptic development. Our study at two complementary model synapses identifies the regulation of the interaction between the L1-type CAM and Ankyrin as an important novel module enabling local control of synaptic connectivity and function while maintaining general neuronal circuit architecture.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/citología , Sinapsis/metabolismo , Transmisión Sináptica , Potenciales de Acción , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Ancirinas/metabolismo , Moléculas de Adhesión Celular Neuronal/química , Aumento de la Célula , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Datos de Secuencia Molecular , Unión Neuromuscular/fisiología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Sinapsis/fisiología
5.
Mol Cell Neurosci ; 64: 24-31, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25433167

RESUMEN

PTP69D is a receptor protein tyrosine phosphatase (RPTP) with two intracellular catalytic domains (Cat1 and Cat2) and has been shown to play a role in axon guidance of embryonic motoneurons as well as targeting of photoreceptor neurons in the visual system of Drosophila melanogaster. Here, we characterized the developmental role of PTP69D in the giant fiber (GF) neurons, two interneurons in the central nervous system (CNS) that control the escape response of the fly. Our studies revealed that PTP69D has a function in synaptic terminal growth in the CNS. We found that missense mutations in the first immunoglobulin (Ig) domain and in the Cat1 domain, present in Ptp69D10 and Ptp69D20 mutants, respectively, did not affect axon guidance or targeting but resulted in stunted terminal growth of the GFs. Cell autonomous rescue experiments demonstrated a function for the Cat1 and the first Ig domain of PTP69D in the GFs but not in its postsynaptic target neurons. In addition, complementation studies and structure-function analyses revealed that for GF terminal growth Cat1 function of PTP69D requires the immunoglobulin and the Cat2 domains, but not the fibronectin III or the membrane proximal region domains. In contrast, the fibronectin III but not the immunoglobulin domains were previously shown to be essential for axon targeting of photoreceptor neurons. Thus, our studies uncover a novel role for PTP69D in synaptic terminal growth in the CNS that is mechanistically distinct from its function in photoreceptor targeting.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neurogénesis , Terminales Presinápticos/metabolismo , Proteínas Tirosina Fosfatasas Similares a Receptores/metabolismo , Animales , Dominio Catalítico , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/fisiología , Fibronectinas/metabolismo , Mutación Missense , Neuronas/citología , Neuronas/metabolismo , Unión Proteica , Proteínas Tirosina Fosfatasas Similares a Receptores/química , Proteínas Tirosina Fosfatasas Similares a Receptores/genética
6.
Neurobiol Dis ; 68: 180-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24807208

RESUMEN

Aminoacyl-tRNA synthetases are ubiquitously expressed proteins that charge tRNAs with their cognate amino acids. By ensuring the fidelity of protein synthesis, these enzymes are essential for the viability of every cell. Yet, mutations in six tRNA synthetases specifically affect the peripheral nerves and cause Charcot-Marie-Tooth (CMT) disease. The CMT-causing mutations in tyrosyl- and glycyl-tRNA synthetases (YARS and GARS, respectively) alter the activity of the proteins in a range of ways (some mutations do not impact charging function, while others abrogate it), making a loss of function in tRNA charging unlikely to be the cause of disease pathology. It is currently unknown which cellular mechanisms are triggered by the mutant enzymes and how this leads to neurodegeneration. Here, by expressing two pathogenic mutations (G240R, P234KY) in Drosophila, we generated a model for GARS-associated neuropathy. We observed compromised viability, and behavioral, electrophysiological and morphological impairment in flies expressing the cytoplasmic isoform of mutant GARS. Their features recapitulated several hallmarks of CMT pathophysiology and were similar to the phenotypes identified in our previously described Drosophila model of YARS-associated neuropathy. Furthermore, CG8316 and CG15599 - genes identified in a retinal degeneration screen to modify mutant YARS, also modified the mutant GARS phenotypes. Our study presents genetic evidence for common mutant-specific interactions between two CMT-associated aminoacyl-tRNA synthetases, lending support for a shared mechanism responsible for the synthetase-induced peripheral neuropathies.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/complicaciones , Enfermedad de Charcot-Marie-Tooth/genética , Glicina-ARNt Ligasa/genética , Mutación/genética , Enfermedades del Sistema Nervioso Periférico/etiología , Tirosina-ARNt Ligasa/genética , Animales , Animales Modificados Genéticamente , Enfermedad de Charcot-Marie-Tooth/patología , Dextranos , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Femenino , Humanos , Masculino , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Fibras Nerviosas/fisiología , Neuronas/patología , Neuronas/fisiología , Enfermedades del Sistema Nervioso Periférico/genética , Retina/patología , Retina/ultraestructura , Degeneración Retiniana/diagnóstico , Degeneración Retiniana/etiología , Degeneración Retiniana/genética , Rodaminas , Alas de Animales/patología , Alas de Animales/ultraestructura
7.
Nat Commun ; 14(1): 999, 2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36890170

RESUMEN

Dominant mutations in tyrosyl-tRNA synthetase (YARS1) and six other tRNA ligases cause Charcot-Marie-Tooth peripheral neuropathy (CMT). Loss of aminoacylation is not required for their pathogenicity, suggesting a gain-of-function disease mechanism. By an unbiased genetic screen in Drosophila, we link YARS1 dysfunction to actin cytoskeleton organization. Biochemical studies uncover yet unknown actin-bundling property of YARS1 to be enhanced by a CMT mutation, leading to actin disorganization in the Drosophila nervous system, human SH-SY5Y neuroblastoma cells, and patient-derived fibroblasts. Genetic modulation of F-actin organization improves hallmark electrophysiological and morphological features in neurons of flies expressing CMT-causing YARS1 mutations. Similar beneficial effects are observed in flies expressing a neuropathy-causing glycyl-tRNA synthetase. Hence, in this work, we show that YARS1 is an evolutionary-conserved F-actin organizer which links the actin cytoskeleton to tRNA-synthetase-induced neurodegeneration.


Asunto(s)
Actinas , Tirosina-ARNt Ligasa , Animales , Humanos , Actinas/metabolismo , Enfermedad de Charcot-Marie-Tooth/genética , Drosophila/genética , Glicina-ARNt Ligasa/genética , Mutación , ARN de Transferencia , Tirosina-ARNt Ligasa/genética , Tirosina-ARNt Ligasa/metabolismo , Línea Celular Tumoral
8.
Methods Mol Biol ; 2431: 417-428, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35412290

RESUMEN

Live imaging of axons allows for the determination of motility and directionality of proteins or organelles. In Drosophila, axonal transport has been predominantly characterized in peripheral neurons, such as larval motor neurons and sensory neurons of the adult wing. As peripheral neurons and central nervous system (CNS) neurons are inherently different, we provide a method to live-image axonal transport of CNS neurons in the cervical connective using an upright or inverted microscope. The method involves dissecting and mounting an entire CNS in a glass bottom petri dish, which is suitable for imaging of nearly any axon in cervical connective. Here, we show an example for simultaneous imaging of both giant fiber axons, which are part of the fly's escape response circuitry, and due to their large diameter provide outstanding resolution.


Asunto(s)
Transporte Axonal , Drosophila , Animales , Transporte Axonal/fisiología , Axones/metabolismo , Sistema Nervioso Central , Drosophila/fisiología , Células Receptoras Sensoriales
9.
BMC Physiol ; 11: 7, 2011 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-21518451

RESUMEN

BACKGROUND: One of the hallmarks of Alzheimer's disease, and several other degenerative disorders such as Inclusion Body Myositis, is the abnormal accumulation of amyloid precursor protein (APP) and its proteolytic amyloid peptides. To better understand the pathological consequences of inappropriate APP expression on developing tissues, we generated transgenic flies that express wild-type human APP in the skeletal muscles, and then performed anatomical, electrophysiological, and behavioral analysis of the adults. RESULTS: We observed that neither muscle development nor animal longevity was compromised in these transgenic animals. However, human APP expressing adults developed age-dependent defects in both climbing and flying. We could advance or retard the onset of symptoms by rearing animals in vials with different surface properties, suggesting that human APP expression-mediated behavioral defects are influenced by muscle activity. Muscles from transgenic animals did not display protein aggregates or structural abnormalities at the light or transmission electron microscopic levels. In agreement with genetic studies performed with developing mammalian myoblasts, we observed that co-expression of the ubiquitin E3 ligase Parkin could ameliorate human APP-induced defects. CONCLUSIONS: These data suggest that: 1) ectopic expression of human APP in fruit flies leads to age- and activity-dependent behavioral defects without overt changes to muscle development or structure; 2) environmental influences can greatly alter the phenotypic consequences of human APP toxicity; and 3) genetic modifiers of APP-induced pathology can be identified and analyzed in this model.


Asunto(s)
Precursor de Proteína beta-Amiloide/fisiología , Modelos Animales de Enfermedad , Drosophila melanogaster/fisiología , Debilidad Muscular/etiología , Unión Neuromuscular/fisiopatología , Envejecimiento , Precursor de Proteína beta-Amiloide/biosíntesis , Precursor de Proteína beta-Amiloide/genética , Animales , Animales Modificados Genéticamente , Ejercicio Físico , Vuelo Animal , Ganglios de Invertebrados/fisiopatología , Vidrio , Vivienda para Animales , Humanos , Operón Lac , Neuronas Motoras/fisiología , Músculos/ultraestructura , Plásticos , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transgenes
10.
MicroPubl Biol ; 20212021.
Artículo en Inglés | MEDLINE | ID: mdl-34235405

RESUMEN

In rodents, all three paralogs of the Attractin (Atrn) transmembrane protein family exhibit strong phenotypic overlap and are implicated in the regulation of the same G-protein coupled receptors (GPCR) as E3-ligase Mahogunin ring finger 1 (Mgrn1). Recently it was shown that the highly conserved intracellular MASRPF motif in mammal Multiple epidermal growth factor-like domain 8 protein is required for binding of Mgrn1 to mediate ubiquitination of GPCR Smoothened in vitro. Here, we show that the MASRPF motif of Drosophila Distracted, the ortholog of ATRN and Attractin-like 1, is required for association with Drosophila Mgrn1 (dMgrn1) in vivo.

11.
J Neurosci ; 28(34): 8615-23, 2008 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-18716220

RESUMEN

The signaling mechanisms that allow the conversion of a growth cone into a mature and stable synapse are yet to be completely understood. Ubiquitination plays key regulatory roles in synaptic development and may be involved in this process. Previous studies identified the Drosophila ubiquitin conjugase bendless (ben) to be important for central synapse formation, but the precise role it plays has not been elucidated. Our studies indicate that Ben plays a pivotal role in synaptic growth and maturation. We have determined that an incipient synapse is present with a high penetrance in ben mutants, suggesting that Ben is required for a developmental step after target recognition. We used cell-autonomous rescue experiments to show that Ben has a presynaptic role in synapse growth. We then harnessed the TARGET system to transiently express UAS (upstream activating sequence)-ben in a ben mutant background and identified a well defined critical period for Ben function in establishing a full-grown, mature synaptic terminal. We demonstrate that the protein must be present at a time point before but not during the actual growth process. We also provide phenotypic evidence demonstrating that Ben is not a part of the signal transduction pathway involving the well characterized ubiquitin ligase highwire. We conclude that Bendless functions as a novel developmental switch that permits the transition from axonal growth and incipient synapse formation to synaptic growth and maturation in the CNS.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/crecimiento & desarrollo , Sinapsis/fisiología , Enzimas Ubiquitina-Conjugadoras/fisiología , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Drosophila/genética , Ganglios de Invertebrados/citología , Ganglios de Invertebrados/metabolismo , Mutación , Proteínas del Tejido Nervioso/fisiología , Terminales Presinápticos/fisiología , Transducción de Señal , Sinapsis/metabolismo , Factores de Tiempo , Distribución Tisular , Enzimas Ubiquitina-Conjugadoras/genética
12.
Curr Biol ; 16(1): 12-23, 2006 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-16401420

RESUMEN

BACKGROUND: Drosophila Neuroglian (Nrg) and its vertebrate homolog L1-CAM are cell-adhesion molecules (CAM) that have been well studied in early developmental processes. Mutations in the human gene result in a broad spectrum of phenotypes (the CRASH-syndrome) that include devastating neurological disorders such as spasticity and mental retardation. Although the role of L1-CAMs in neurite extension and axon pathfinding has been extensively studied, much less is known about their role in synapse formation. RESULTS: We found that a single extracellular missense mutation in nrg(849) mutants disrupted the physiological function of a central synapse in Drosophila. The identified giant neuron in nrg(849) mutants made a synaptic terminal on the appropriate target, but ultrastructural analysis revealed in the synaptic terminal a dramatic microtubule reduction, which was likely to be the cause for disrupted active zones. Our results reveal that tyrosine phosphorylation of the intracellular ankyrin binding motif was reduced in mutants, and cell-autonomous rescue experiments demonstrated the indispensability of this tyrosine in giant-synapse formation. We also show that this function in giant-synapse formation was conserved in human L1-CAM but neither in human L1-CAM with a pathological missense mutation nor in two isoforms of the paralogs NrCAM and Neurofascin. CONCLUSIONS: We conclude that Nrg has a function in synapse formation by organizing microtubules in the synaptic terminal. This novel synaptic function is conserved in human L1-CAM but is not common to all L1-type proteins. Finally, our findings suggest that some aspects of L1-CAM-related neurological disorders in humans may result from a disruption in synapse formation rather than in axon pathfinding.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Proteínas de Drosophila/fisiología , Drosophila/fisiología , Molécula L1 de Adhesión de Célula Nerviosa/fisiología , Sinapsis/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Secuencia Conservada , Drosophila/metabolismo , Drosophila/ultraestructura , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Conductividad Eléctrica , Genotipo , Humanos , Modelos Biológicos , Mutación Missense , Molécula L1 de Adhesión de Célula Nerviosa/genética , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuronas/fisiología , Fosforilación , Homología Estructural de Proteína , Sinapsis/ultraestructura
13.
J Neurogenet ; 23(1-2): 147-55, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19052954

RESUMEN

We have previously demonstrated a function for Neuroglian and Semaphorin1a in Drosophila giant fiber circuit formation. Both molecules are required for guiding the giant fibers out of the brain and have distinct functions during giant synapse formation. In this study we characterized the effects of various combinations of Neuroglian and Semaphorin1a gain and loss of function backgrounds on giant fiber circuitry formation. We found that Neuroglian and Semaphorin1a genetically interact with each other during axon guidance as well as during synapse formation. Our experiments revealed that during pathfinding of the giant fibers out of the brain, Neuroglian function seems to be dependent on Semaphorin1a. In contrast, during giant fiber synapse formation we observed that Semaphorin1a signaling as a receptor can be altered by Neuroglian in the same cell. In summary, our findings suggest that Neuroglian and Semaphorin1a can regulate each other's function in cis and that the resultant signaling output is possibly different during guidance and synapse formation.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Proteínas de Drosophila/genética , Drosophila/genética , Semaforinas/genética , Sinapsis/fisiología , Animales , Adhesión Celular/fisiología , Moléculas de Adhesión Celular Neuronal/fisiología , Drosophila/fisiología , Proteínas de Drosophila/fisiología , Mutación , Fibras Nerviosas/fisiología , Vías Nerviosas/fisiología , Semaforinas/fisiología , Transducción de Señal/fisiología
14.
Cell Mol Biol Lett ; 14(1): 57-69, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18839070

RESUMEN

L1-type cell adhesion molecules (CAMs) are important mediators of neural differentiation, including axonal outgrowth and pathfinding and also of synapse formation and maintenance. In addition, their interactions with cytoskeletal components are highly conserved and regulated. How these different aspects of CAM functionality relate to each other is not well understood. Based on results from our and other laboratories we propose that ankyrin-binding to L1-type CAMs provides a master switch. The interaction with ankyrins directs L1-type adhesive proteins into different functional contexts, either ankyrin-independent functions, such as neurite outgrowth and axonal pathfinding or into ankyrin-dependent functions, such as L1's role at axon initial segments (AIS), paranodal regions, synapses and in dendrites.


Asunto(s)
Ancirinas/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Secuencia de Aminoácidos , Animales , Axones/metabolismo , Drosophila/metabolismo , Datos de Secuencia Molecular , Molécula L1 de Adhesión de Célula Nerviosa/química , Neuroglía/metabolismo , Unión Proteica
15.
Neuron ; 36(1): 5-8, 2002 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-12367500

RESUMEN

A series of recent papers highlight a prominent role for ubiquitin in the formation and function of neural circuits. These new results focus attention on the molecular remodeling that occurs at various decision points in the life of growth cones and synapses.


Asunto(s)
Diferenciación Celular/fisiología , Sistema Nervioso Central/embriología , Conos de Crecimiento/metabolismo , Vías Nerviosas/embriología , Ubiquitina/metabolismo , Animales , Comunicación Celular/fisiología , Membrana Celular/metabolismo , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Quimiotaxis/fisiología , Conos de Crecimiento/ultraestructura , Humanos , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Sinapsis/metabolismo , Sinapsis/ultraestructura
16.
Nat Neurosci ; 5(12): 1294-301, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12436113

RESUMEN

Semaphorins have been intensively studied for their role in dendritic and axonal pathfinding, but less is known about their potential role in synapse formation. In the adult giant fiber (GF) system of fruit flies (Drosophila melanogaster), we show that transmembrane Semaphorin 1a (Sema-1a) is involved in synapse formation in addition to its role in guidance during pathfinding. Cell-autonomous rescue experiments showed that Sema-1a is involved in assembly of a central synapse and that it is required both pre- and postsynaptically. We also found that pre- but not postsynaptic gain-of-function Sema-1a was able to disrupt the GF-motor neuron synapse and that the phenotype depended on a proline-rich intracellular domain that contains a putative Enabled binding site. We suggest that transmembrane Sema-1a is part of a bi-directional signaling system that leads to the formation of the GF synapse and possibly acts as both a ligand and a receptor.


Asunto(s)
Diferenciación Celular/genética , Sistema Nervioso Central/embriología , Drosophila/embriología , Terminales Presinápticos/metabolismo , Semaforinas/deficiencia , Transducción de Señal/genética , Animales , Animales Modificados Genéticamente , Sistema Nervioso Central/citología , Sistema Nervioso Central/metabolismo , Drosophila/citología , Drosophila/metabolismo , Femenino , Masculino , Mutación/genética , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo , Fenotipo , Terminales Presinápticos/ultraestructura , Estructura Terciaria de Proteína/genética , Tiempo de Reacción/genética , Semaforinas/genética , Membranas Sinápticas/genética , Membranas Sinápticas/metabolismo
17.
PLoS One ; 12(8): e0183605, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28837701

RESUMEN

Here, we established the Drosophila Giant Fiber neurons (GF) as a novel model to study axonal trafficking of L1-type Cell Adhesion Molecules (CAM) Neuroglian (Nrg) in the adult CNS using live imaging. L1-type CAMs are well known for their importance in nervous system development and we previously demonstrated a role for Nrg in GF synapse formation. However, in the adult they have also been implicated in synaptic plasticity and regeneration. In addition, to its canonical role in organizing cytoskeletal elements at the plasma membrane, vertebrate L1CAM has also been shown to regulate transcription indirectly as well as directly via its import to the nucleus. Here, we intend to determine if the sole L1CAM homolog Nrg is retrogradley transported and thus has the potential to relay signals from the synapse to the soma. Live imaging of c-terminally tagged Nrg in the GF revealed that there are at least two populations of retrograde vesicles that differ in speed, and either move with consistent or varying velocity. To determine if endogenous Nrg is retrogradely transported, we inhibited two key regulators, Lissencephaly-1 (Lis1) and Dynactin, of the retrograde motor protein Dynein. Similar to previously described phenotypes for expression of poisonous subunits of Dynactin, we found that developmental knock down of Lis1 disrupted GF synaptic terminal growth and that Nrg vesicles accumulated inside the stunted terminals in both mutant backgrounds. Moreover, post mitotic Lis1 knock down in mature GFs by either RNAi or Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) induced mutations, resulted in normal length terminals with fully functional GF synapses which also exhibited severe accumulation of endogenous Nrg vesicles. Thus, our data suggests that accumulation of Nrg vesicles is due to failure of retrograde transport rather than a failure of terminal development. Together with the finding that post mitotic knock down of Lis1 also disrupted retrograde transport of tagged Nrg vesicles in GF axons, it demonstrates that endogenous Nrg protein is transported from the synapse to the soma in the adult central nervous system in a Lis1-dependent manner.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Sistema Nervioso Central/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Animales , Transporte Biológico , Moléculas de Adhesión Celular Neuronal/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Proteínas de Drosophila/genética , Técnicas de Silenciamiento del Gen
18.
Neuropharmacology ; 127: 253-259, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28917942

RESUMEN

α-Conotoxins inhibit nicotinic acetylcholine receptors (nAChRs) and are used as probes to study cholinergic pathways in vertebrates. Model organisms, such as Drosophila melanogaster, express nAChRs in their CNS that are suitable to investigate the neuropharmacology of α-conotoxins in vivo. Here we report the paired nanoinjection of native α-conotoxin PIA and two novel α-conotoxins, PIC and PIC[O7], from the injected venom of Conus purpurascens and electrophysiological recordings of their effects on the giant fiber system (GFS) of D. melanogaster and heterologously expressed nAChRs in Xenopus oocytes. α-PIA caused disruption of the function of giant fiber dorsal longitudinal muscle (GF-DLM) pathway by inhibiting the Dα7 nAChR a homolog to the vertebrate α7 nAChR, whereas PIC and PIC[O7] did not. PIC and PIC[O7] reversibly inhibited ACh-evoked currents mediated by vertebrate rodent (r)α1ß1δγ, rα1ß1δε and human (h)α3ß2, but not hα7 nAChR subtypes expressed in Xenopus oocytes with the following selectivity: rα1ß1δε > rα1ß1δγ ≈ hα3ß2 >> hα7. Our study emphasizes the importance of loop size and α-conotoxin sequence specificity for receptor binding. These studies can be used for the evaluation of the neuropharmacology of novel α-conotoxins that can be utilized as molecular probes for diseases such as, Alzheimer's, Parkinson's, and cancer. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'


Asunto(s)
Conotoxinas/farmacología , Caracol Conus/química , Potenciales de la Membrana/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Acetilcolina/farmacología , Animales , Cromatografía Líquida de Alta Presión , Conotoxinas/química , Relación Dosis-Respuesta a Droga , Drosophila melanogaster , Potenciales de la Membrana/genética , Microinyecciones , Modelos Moleculares , Oocitos , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Xenopus
19.
J Neurosci ; 22(8): 3117-29, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11943815

RESUMEN

The Roundabout (Robo) receptors have been intensively studied for their role in regulating axon guidance in the embryonic nervous system, whereas a role in dendritic guidance has not been explored. In the adult giant fiber system of Drosophila, we have revealed that ectopic Robo expression can regulate the growth and guidance of specific motor neuron dendrites, whereas Robo2 and Robo3 have no effect. We also show that the effect of Robo on dendritic guidance can be suppressed by Commissureless coexpression. Although we confirmed a role for all three Robo receptors in giant fiber axon guidance, the strong axon guidance alterations caused by overexpression of Robo2 or Robo3 have no effect on synaptic connectivity. In contrast, Robo overexpression in the giant fiber seems to directly interfere with synaptic function. We conclude that axon guidance, dendritic guidance, and synaptogenesis are separable processes and that the different Robo family members affect them distinctly.


Asunto(s)
Dendritas/fisiología , Proteínas de Drosophila , Fibras Nerviosas/metabolismo , Receptores Inmunológicos/biosíntesis , Sinapsis/fisiología , Secuencias de Aminoácidos/fisiología , Animales , Animales Modificados Genéticamente , Axones/fisiología , Drosophila , Estimulación Eléctrica , Potenciales Postsinápticos Excitadores/fisiología , Expresión Génica , Proteínas de Insectos/biosíntesis , Proteínas de Insectos/genética , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Neuronas Motoras/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Fenotipo , Pupa/metabolismo , Tiempo de Reacción/fisiología , Receptores Inmunológicos/genética , Proteínas Roundabout
20.
PLoS One ; 8(10): e76974, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24155914

RESUMEN

A large number of different pathological L1CAM mutations have been identified that result in a broad spectrum of neurological and non-neurological phenotypes. While many of these mutations have been characterized for their effects on homophilic and heterophilic interactions, as well as expression levels in vitro, there are only few studies on their biological consequences in vivo. The single L1-type CAM gene in Drosophila, neuroglian (nrg), has distinct functions during axon guidance and synapse formation and the phenotypes of nrg mutants can be rescued by the expression of human L1CAM. We previously showed that the highly conserved intracellular FIGQY Ankyrin-binding motif is required for L1CAM-mediated synapse formation, but not for neurite outgrowth or axon guidance of the Drosophila giant fiber (GF) neuron. Here, we use the GF as a model neuron to characterize the pathogenic L120V, Y1070C, C264Y, H210Q, E309K and R184Q extracellular L1CAM missense mutations and a L1CAM protein with a disrupted ezrin-moesin-radixin (ERM) binding site to investigate the signaling requirements for neuronal development. We report that different L1CAM mutations have distinct effects on axon guidance and synapse formation. Furthermore, L1CAM homophilic binding and signaling via the ERM motif is essential for axon guidance in Drosophila. In addition, the human pathological H210Q, R184Q and Y1070C, but not the E309K and L120V L1CAM mutations affect outside-in signaling via the FIGQY Ankyrin binding domain which is required for synapse formation. Thus, the pathological phenotypes observed in humans are likely to be caused by the disruption of signaling required for both, guidance and synaptogenesis.


Asunto(s)
Drosophila melanogaster/metabolismo , Prueba de Complementación Genética , Mutación Missense/genética , Molécula L1 de Adhesión de Célula Nerviosa/genética , Sinapsis/metabolismo , Animales , Axones/metabolismo , Fenómenos Electrofisiológicos , Humanos , Inmunohistoquímica , Proteínas Mutantes/metabolismo , Sistema Nervioso/metabolismo , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA