Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Mol Psychiatry ; 28(1): 191-201, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36434053

RESUMEN

An obvious consequence of the coronavirus disease (COVID-19) pandemic is the worldwide reduction in social interaction, which is associated with many adverse effects on health in humans from babies to adults. Although social development under normal or isolated environments has been studied since the 1940s, the mechanism underlying social isolation (SI)-induced brain dysfunction remains poorly understood, possibly due to the complexity of SI in humans and translational gaps in findings from animal models. Herein, we present a systematic review that focused on brain changes at the molecular, cellular, structural and functional levels induced by SI at different ages and in different animal models. SI studies in humans and animal models revealed common socioemotional and cognitive deficits caused by SI in early life and an increased occurrence of depression and anxiety induced by SI during later stages of life. Altered neurotransmission and neural circuitry as well as abnormal development and function of glial cells in specific brain regions may contribute to the abnormal emotions and behaviors induced by SI. We highlight distinct alterations in oligodendrocyte progenitor cell differentiation and oligodendrocyte maturation caused by SI in early life and later stages of life, respectively, which may affect neural circuit formation and function and result in diverse brain dysfunctions. To further bridge animal and human SI studies, we propose alternative animal models with brain structures and complex social behaviors similar to those of humans.


Asunto(s)
COVID-19 , Control de Infecciones , Pandemias , Aislamiento Social , Animales , Humanos , Conducta Animal , Encéfalo , Emociones
2.
Mol Psychiatry ; 28(9): 3751-3759, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37848709

RESUMEN

Pupillary response, an important process in visual perception and social and emotional cognition, has been widely studied for understanding the neural mechanisms of neuropsychiatric disorders. However, there have been few studies on pupil response to social and non-social stimuli in animal models of neurodevelopmental disorders including autism spectrum disorder (ASD) and attention deficit hyperactivity disorder. Here, we developed a pupilometer using a robust eye feature-detection algorithm for real-time pupillometry in dogs. In a pilot study, we found that a brief light flash induced a less-pronounced and slower pupil dilation response in gene-edited dogs carrying mutations in Shank3; mutations of its ortholog in humans were repeatedly identified in ASD patients. We further found that obnoxious, loud firecracker sound of 120 dB induced a stronger and longer pupil dilation response in Shank3 mutant dogs, whereas a high reward food induced a weaker pupillary response in Shank3 mutants than in wild-type control dogs. In addition, we found that Shank3 mutants showed compromised pupillary synchrony during dog-human interaction. These findings of altered pupil response in Shank3 mutant dogs recapitulate the altered sensory responses in ASD patients. Thus, this study demonstrates the validity and value of the pupilometer for dogs, and provides an effective paradigm for studying the underlying neural mechanisms of ASD and potentially other psychiatric disorders.


Asunto(s)
Trastorno del Espectro Autista , Humanos , Perros , Animales , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/psicología , Pupila/fisiología , Proyectos Piloto , Emociones , Modelos Animales de Enfermedad
3.
Mol Psychiatry ; 28(9): 3739-3750, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37848710

RESUMEN

Despite intensive studies in modeling neuropsychiatric disorders especially autism spectrum disorder (ASD) in animals, many challenges remain. Genetic mutant mice have contributed substantially to the current understanding of the molecular and neural circuit mechanisms underlying ASD. However, the translational value of ASD mouse models in preclinical studies is limited to certain aspects of the disease due to the apparent differences in brain and behavior between rodents and humans. Non-human primates have been used to model ASD in recent years. However, a low reproduction rate due to a long reproductive cycle and a single birth per pregnancy, and an extremely high cost prohibit a wide use of them in preclinical studies. Canine model is an appealing alternative because of its complex and effective dog-human social interactions. In contrast to non-human primates, dog has comparable drug metabolism as humans and a high reproduction rate. In this study, we aimed to model ASD in experimental dogs by manipulating the Shank3 gene as SHANK3 mutations are one of most replicated genetic defects identified from ASD patients. Using CRISPR/Cas9 gene editing, we successfully generated and characterized multiple lines of Beagle Shank3 (bShank3) mutants that have been propagated for a few generations. We developed and validated a battery of behavioral assays that can be used in controlled experimental setting for mutant dogs. bShank3 mutants exhibited distinct and robust social behavior deficits including social withdrawal and reduced social interactions with humans, and heightened anxiety in different experimental settings (n = 27 for wild-type controls and n = 44 for mutants). We demonstrate the feasibility of producing a large number of mutant animals in a reasonable time frame. The robust and unique behavioral findings support the validity and value of a canine model to investigate the pathophysiology and develop treatments for ASD and potentially other psychiatric disorders.


Asunto(s)
Trastorno del Espectro Autista , Animales , Perros , Humanos , Trastorno del Espectro Autista/genética , Sistemas CRISPR-Cas/genética , Modelos Animales de Enfermedad , Edición Génica , Proteínas de Microfilamentos/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo
4.
Cereb Cortex ; 33(20): 10546-10557, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37585733

RESUMEN

Both enhanced discrimination of low-level features of auditory stimuli and mutations of SHANK3 (a gene that encodes a synaptic scaffolding protein) have been identified in autism spectrum disorder patients. However, experimental evidence regarding whether SHANK3 mutations lead to enhanced neural processing of low-level features of auditory stimuli is lacking. The present study investigated this possibility by examining effects of Shank3 mutations on early neural processing of pitch (tone frequency) in dogs. We recorded electrocorticograms from wild-type and Shank3 mutant dogs using an oddball paradigm in which deviant tones of different frequencies or probabilities were presented along with other tones in a repetitive stream (standards). We found that, relative to wild-type dogs, Shank3 mutant dogs exhibited larger amplitudes of early neural responses to deviant tones and greater sensitivity to variations of deviant frequencies within 100 ms after tone onsets. In addition, the enhanced early neural responses to deviant tones in Shank3 mutant dogs were observed independently of the probability of deviant tones. Our findings highlight an essential functional role of Shank3 in modulations of early neural detection of novel sounds and offer new insights into the genetic basis of the atypical auditory information processing in autism patients.

5.
Mol Cell Proteomics ; 21(8): 100261, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35738554

RESUMEN

Brain development and function are governed by precisely regulated protein expressions in different regions. To date, multiregional brain proteomes have been systematically analyzed only for adult human and mouse brains. To understand the underpinnings of brain development and function, we generated proteomes from six regions of the postnatal brain at three developmental stages of domestic dogs (Canis familiaris), which are special among animals in terms of their remarkable human-like social cognitive abilities. Quantitative analysis of the spatiotemporal proteomes identified region-enriched synapse types at different developmental stages and differential myelination progression in different brain regions. Through integrative analysis of inter-regional expression patterns of orthologous proteins and genome-wide cis-regulatory element frequencies, we found that proteins related with myelination and hippocampus were highly correlated between dog and human but not between mouse and human, although mouse is phylogenetically closer to human. Moreover, the global expression patterns of neurodegenerative disease and autism spectrum disorder-associated proteins in dog brain more resemble human brain than in mouse brain. The high similarity of myelination and hippocampus-related pathways in dog and human at both proteomic and genetic levels may contribute to their shared social cognitive abilities. The inter-regional expression patterns of disease-associated proteins in the brain of different species provide important information to guide mechanistic and translational study using appropriate animal models.


Asunto(s)
Trastorno del Espectro Autista , Enfermedades Neurodegenerativas , Adulto , Animales , Encéfalo , Perros , Humanos , Ratones , Proteoma , Proteómica
6.
Development ; 147(24)2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33234716

RESUMEN

The balance among different subtypes of glutamate receptors (GluRs) is crucial for synaptic function and plasticity at excitatory synapses. However, the mechanisms balancing synaptic GluR subtypes remain unclear. Herein, we show that the two subtypes of GluRs (A and B) expressed at Drosophila neuromuscular junction synapses mutually antagonize each other in terms of their relative synaptic levels and affect subsynaptic localization of each other, as shown by super-resolution microscopy. Upon temperature shift-induced neuromuscular junction plasticity, GluR subtype A increased but subtype B decreased with a timecourse of hours. Inhibition of the activity of GluR subtype A led to imbalance of GluR subtypes towards more GluRIIA. To gain a better understanding of the signalling pathways underlying the balance of GluR subtypes, we performed an RNA interference screen of candidate genes and found that postsynaptic-specific knockdown of dunce, which encodes cAMP phosphodiesterase, increased levels of GluR subtype A but decreased subtype B. Furthermore, bidirectional alterations of postsynaptic cAMP signalling resulted in the same antagonistic regulation of the two GluR subtypes. Our findings thus identify a direct role of postsynaptic cAMP signalling in control of the plasticity-related balance of GluRs.


Asunto(s)
Proteínas de Drosophila/genética , Plasticidad Neuronal/genética , Receptores Ionotrópicos de Glutamato/genética , Sinapsis/genética , Animales , AMP Cíclico/genética , Drosophila melanogaster/genética , Unión Neuromuscular/genética , Unión Neuromuscular/crecimiento & desarrollo , Receptores de Glutamato/genética , Transducción de Señal/genética , Transmisión Sináptica/genética
7.
Mamm Genome ; 34(2): 262-269, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36243810

RESUMEN

Cardiovascular diseases (CVD) are one of the leading causes of death worldwide. Eighty-five percent of CVD-associated deaths are due to heart attacks and stroke. Atherosclerosis leads to heart attack and stroke through a slow progression of lesion formation and luminal narrowing of arteries. Dogs are similar to humans in terms of their cardiovascular physiology, size, and anatomy. Dog models have been developed to recapitulate the complex phenotype of human patients and understand the underlying mechanism of CVD. Different methods, including high-fat, high-cholesterol diet and genetic modification, have been used to generate dog models of human CVD. Remarkably, the location and severity of atherosclerotic lesions in the coronary arteries and branches of the carotid arteries of dog models closely resemble those of human CVD patients. Overt clinical manifestations such as stroke caused by plaque rupture and thrombosis were observed in dog models. Thus, dog models can help define the pathophysiological mechanisms of atherosclerosis and develop potential strategy for preventing and treating CVD. In this review, we summarize the progress in generating and characterizing canine models to investigate CVD and discuss the advantages and limitations of canine CVD models.


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Infarto del Miocardio , Placa Aterosclerótica , Accidente Cerebrovascular , Humanos , Perros , Animales , Enfermedades Cardiovasculares/genética , Aterosclerosis/genética , Placa Aterosclerótica/patología , Factores de Riesgo
8.
J Neurosci ; 40(14): 2817-2827, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32122953

RESUMEN

Perturbations to postsynaptic glutamate receptors (GluRs) trigger retrograde signaling to precisely increase presynaptic neurotransmitter release, maintaining stable levels of synaptic strength, a process referred to as homeostatic regulation. However, the structural change of homeostatic regulation remains poorly defined. At wild-type Drosophila neuromuscular junction synapse, there is one Bruchpilot (Brp) ring detected by superresolution microscopy at active zones (AZs). In the present study, we report multiple Brp rings (i.e., multiple T-bars seen by electron microscopy) at AZs of both male and female larvae when GluRs are reduced. At GluRIIC-deficient neuromuscular junctions, quantal size was reduced but quantal content was increased, indicative of homeostatic presynaptic potentiation. Consistently, multiple Brp rings at AZs were observed in the two classic synaptic homeostasis models (i.e., GluRIIA mutant and pharmacological blockade of GluRIIA activity). Furthermore, postsynaptic overexpression of the cell adhesion protein Neuroligin 1 partially rescued multiple Brp rings phenotype. Our study thus supports that the formation of multiple Brp rings at AZs might be a structural basis for synaptic homeostasis.SIGNIFICANCE STATEMENT Synaptic homeostasis is a conserved fundamental mechanism to maintain efficient neurotransmission of neural networks. Active zones (AZs) are characterized by an electron-dense cytomatrix, which is largely composed of Bruchpilot (Brp) at the Drosophila neuromuscular junction synapses. It is not clear how the structure of AZs changes during homeostatic regulation. To address this question, we examined the structure of AZs by superresolution microscopy and electron microscopy during homeostatic regulation. Our results reveal multiple Brp rings at AZs of glutamate receptor-deficient neuromuscular junction synapses compared with single Brp ring at AZs in wild type (WT). We further show that Neuroligin 1-mediated retrograde signaling regulates multiple Brp ring formation at glutamate receptor-deficient synapses. This study thus reveals a regulatory mechanism for synaptic homeostasis.


Asunto(s)
Homeostasis/fisiología , Unión Neuromuscular/fisiología , Unión Neuromuscular/ultraestructura , Sinapsis/metabolismo , Sinapsis/ultraestructura , Transmisión Sináptica/fisiología , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Drosophila , Proteínas de Drosophila/metabolismo , Femenino , Masculino , Receptores de Glutamato/metabolismo
9.
Hum Mol Genet ; 28(4): 561-571, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30329048

RESUMEN

Monogenic mutations in the SHANK3 gene, which encodes a postsynaptic scaffold protein, play a causative role in autism spectrum disorder (ASD). Although a number of mouse models with Shank3 mutations have been valuable for investigating the pathogenesis of ASD, species-dependent differences in behaviors and brain structures post considerable challenges to use small animals to model ASD and to translate experimental therapeutics to the clinic. We have used clustered regularly interspersed short palindromic repeat/CRISPR-associated nuclease 9 to generate a cynomolgus monkey model by disrupting SHANK3 at exons 6 and 12. Analysis of the live mutant monkey revealed the core behavioral abnormalities of ASD, including impaired social interaction and repetitive behaviors, and reduced brain network activities detected by positron-emission computed tomography (PET). Importantly, these abnormal behaviors and brain activities were alleviated by the antidepressant fluoxetine treatment. Our findings provide the first demonstration that the genetically modified non-human primate can be used for translational research of therapeutics for ASD.


Asunto(s)
Trastorno del Espectro Autista/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Fluoxetina/administración & dosificación , Proteínas del Tejido Nervioso/genética , Animales , Trastorno del Espectro Autista/diagnóstico por imagen , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Conducta Animal/efectos de los fármacos , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Sistemas CRISPR-Cas/genética , Modelos Animales de Enfermedad , Exones , Humanos , Relaciones Interpersonales , Macaca fascicularis/genética , Ratones , Mutación
10.
J Neurosci ; 39(15): 2776-2791, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30705102

RESUMEN

Calpains are calcium-dependent, cytosolic proteinases active at neutral pH. They do not degrade but cleave substrates at limited sites. Calpains are implicated in various pathologies, such as ischemia, injuries, muscular dystrophy, and neurodegeneration. Despite so, the physiological function of calpains remains to be clearly defined. Using the neuromuscular junction of Drosophila of both sexes as a model, we performed RNAi screening and uncovered that calpains negatively regulated protein levels of the glutamate receptor GluRIIA but not GluRIIB. We then showed that calpains enrich at the postsynaptic area, and the calcium-dependent activation of calpains induced cleavage of GluRIIA at Q788 of its C terminus. Further genetic and biochemical experiments revealed that different calpains genetically and physically interact to form a protein complex. The protein complex was required for the proteinase activation to downregulate GluRIIA. Our data provide a novel insight into the mechanisms by which different calpains act together as a complex to specifically control GluRIIA levels and consequently synaptic function.SIGNIFICANCE STATEMENT Calpain has been implicated in neural insults and neurodegeneration. However, the physiological function of calpains in the nervous system remains to be defined. Here, we show that calpain enriches at the postsynaptic area and negatively and specifically regulates GluRIIA, but not IIB, level during development. Calcium-dependent activation of calpain cleaves GluRIIA at Q788 of its C terminus. Different calpains constitute an active protease complex to cleave its target. This study reveals a critical role of calpains during development to specifically cleave GluRIIA at synapses and consequently regulate synaptic function.


Asunto(s)
Calpaína/genética , Calpaína/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Receptores Ionotrópicos de Glutamato/genética , Receptores Ionotrópicos de Glutamato/metabolismo , Animales , Señalización del Calcio/genética , Regulación hacia Abajo/genética , Femenino , Inmunohistoquímica , Masculino , Músculos/metabolismo , Optogenética , Péptido Hidrolasas/metabolismo , Interferencia de ARN , Especificidad por Sustrato , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/fisiología
11.
PLoS Genet ; 12(5): e1006062, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27232889

RESUMEN

Altered expression of the E3 ubiquitin ligase UBE3A, which is involved in protein degradation through the proteasome-mediated pathway, is associated with neurodevelopmental and behavioral defects observed in Angelman syndrome (AS) and autism. However, little is known about the neuronal function of UBE3A and the pathogenesis of UBE3A-associated disorders. To understand the in vivo function of UBE3A in the nervous system, we generated multiple mutations of ube3a, the Drosophila ortholog of UBE3A. We found a significantly increased number of total boutons and satellite boutons in conjunction with compromised endocytosis in the neuromuscular junctions (NMJs) of ube3a mutants compared to the wild type. Genetic and biochemical analysis showed upregulation of bone morphogenetic protein (BMP) signaling in the nervous system of ube3a mutants. An immunochemical study revealed a specific increase in the protein level of Thickveins (Tkv), a type I BMP receptor, but not other BMP receptors Wishful thinking (Wit) and Saxophone (Sax), in ube3a mutants. Ube3a was associated with and specifically ubiquitinated lysine 227 within the cytoplasmic tail of Tkv, and promoted its proteasomal degradation in Schneider 2 cells. Negative regulation of Tkv by Ube3a was conserved in mammalian cells. These results reveal a critical role for Ube3a in regulating NMJ synapse development by repressing BMP signaling. This study sheds new light onto the neuronal functions of UBE3A and provides novel perspectives for understanding the pathogenesis of UBE3A-associated disorders.


Asunto(s)
Síndrome de Angelman/genética , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Receptores de Superficie Celular/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Síndrome de Angelman/patología , Animales , Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Morfogenéticas Óseas/genética , Modelos Animales de Enfermedad , Drosophila/genética , Endocitosis/genética , Regulación de la Expresión Génica/genética , Humanos , Unión Neuromuscular/genética , Unión Neuromuscular/patología , Neuronas/patología , Proteínas Serina-Treonina Quinasas/genética , Receptores de Superficie Celular/genética , Transducción de Señal , Sinapsis/genética , Ubiquitina-Proteína Ligasas/biosíntesis
12.
J Neurosci ; 37(48): 11592-11604, 2017 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-29074576

RESUMEN

Human genetic studies support that loss-of-function mutations in the SH3 domain and ankyrin repeat containing family proteins (SHANK1-3), the large synaptic scaffolding proteins enriched at the postsynaptic density of excitatory synapses, are causative for autism spectrum disorder and other neuropsychiatric disorders in humans. To better understand the in vivo functions of Shank and facilitate dissection of neuropathology associated with SHANK mutations in human, we generated multiple mutations in the Shank gene, the only member of the SHANK family in Drosophila melanogaster Both male and female Shank null mutants were fully viable and fertile with no apparent morphological or developmental defects. Expression analysis revealed apparent enrichment of Shank in the neuropils of the CNS. Specifically, Shank coexpressed with another PSD scaffold protein, Homer, in the calyx of mushroom bodies in the brain. Consistent with high expression in mushroom body calyces, Shank mutants show an abnormal calyx structure and reduced olfactory acuity. These morphological and functional phenotypes were fully rescued by pan-neuronal reexpression of Shank, and only partially rescued by presynaptic but no rescue by postsynaptic reexpression of Shank. Our findings thus establish a previously unappreciated presynaptic function of Shank.SIGNIFICANCE STATEMENT Mutations in SHANK family genes are causative for idiopathic autism spectrum disorder. To understand the neural function of Shank, a large scaffolding protein enriched at the postsynaptic densities, we examined the role of Drosophila Shank in synapse development at the peripheral neuromuscular junctions and the central mushroom body calyx. Our results demonstrate that, in addition to its conventional postsynaptic function, Shank also acts presynaptically in synapse development in the brain. This study offers novel insights into the synaptic role of Shank.


Asunto(s)
Proteínas del Tejido Nervioso/fisiología , Proteínas del Tejido Nervioso/ultraestructura , Terminales Presinápticos/fisiología , Terminales Presinápticos/ultraestructura , Animales , Animales Modificados Genéticamente , Drosophila , Femenino , Masculino , Cuerpos Pedunculados/fisiología , Cuerpos Pedunculados/ultraestructura , Unión Neuromuscular/fisiología , Unión Neuromuscular/ultraestructura
13.
J Cell Sci ; 129(21): 4034-4045, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27656110

RESUMEN

Nervous system development and function are tightly regulated by metabolic processes, including the metabolism of lipids such as fatty acids. Mutations in long-chain acyl-CoA synthetase 4 (ACSL4) are associated with non-syndromic intellectual disabilities. We previously reported that Acsl, the Drosophila ortholog of mammalian ACSL3 and ACSL4, inhibits neuromuscular synapse growth by suppressing bone morphogenetic protein (BMP) signaling. Here, we report that Acsl regulates the composition of fatty acids and membrane lipids, which in turn affects neuromuscular junction (NMJ) synapse development. Acsl mutant brains had a decreased abundance of C16:1 fatty acyls; restoration of Acsl expression abrogated NMJ overgrowth and the increase in BMP signaling. A lipidomic analysis revealed that Acsl suppressed the levels of three lipid raft components in the brain, including mannosyl glucosylceramide (MacCer), phosphoethanolamine ceramide and ergosterol. The MacCer level was elevated in Acsl mutant NMJs and, along with sterol, promoted NMJ overgrowth, but was not associated with the increase in BMP signaling in the mutants. These findings suggest that Acsl inhibits NMJ growth by stimulating C16:1 fatty acyl production and concomitantly suppressing raft-associated lipid levels.


Asunto(s)
Coenzima A Ligasas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Discapacidad Intelectual/metabolismo , Lípidos/química , Homología de Secuencia de Aminoácido , Sinapsis/metabolismo , Animales , Encéfalo/metabolismo , Ceramidas/metabolismo , Retículo Endoplásmico/metabolismo , Ácido Graso Desaturasas/metabolismo , Ácidos Grasos/metabolismo , Microdominios de Membrana/metabolismo , Mutación/genética , Unión Neuromuscular/metabolismo , Neuronas/metabolismo , Peroxisomas/metabolismo , Esteroles/metabolismo
14.
PLoS Genet ; 11(3): e1004984, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25748449

RESUMEN

Synaptic connections must be precisely controlled to ensure proper neural circuit formation. In Drosophila melanogaster, bone morphogenetic protein (BMP) promotes growth of the neuromuscular junction (NMJ) by binding and activating the BMP ligand receptors wishful thinking (Wit) and thickveins (Tkv) expressed in motor neurons. We report here that an evolutionally conserved, previously uncharacterized member of the S6 kinase (S6K) family S6K like (S6KL) acts as a negative regulator of BMP signaling. S6KL null mutants were viable and fertile but exhibited more satellite boutons, fewer and larger synaptic vesicles, larger spontaneous miniature excitatory junctional potential (mEJP) amplitudes, and reduced synaptic endocytosis at the NMJ terminals. Reducing the gene dose by half of tkv in S6KL mutant background reversed the NMJ overgrowth phenotype. The NMJ phenotypes of S6KL mutants were accompanied by an elevated level of Tkv protein and phosphorylated Mad, an effector of the BMP signaling pathway, in the nervous system. In addition, Tkv physically interacted with S6KL in cultured S2 cells. Furthermore, knockdown of S6KL enhanced Tkv expression, while S6KL overexpression downregulated Tkv in cultured S2 cells. This latter effect was blocked by the proteasome inhibitor MG132. Our results together demonstrate for the first time that S6KL regulates synaptic development and function by facilitating proteasomal degradation of the BMP receptor Tkv.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas de Drosophila/genética , Unión Neuromuscular/crecimiento & desarrollo , Proteínas Serina-Treonina Quinasas/genética , Receptores de Superficie Celular/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Animales , Animales Modificados Genéticamente , Receptores de Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Endocitosis/genética , Neuronas Motoras/metabolismo , Unión Neuromuscular/metabolismo , Plasticidad Neuronal/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteolisis , Receptores de Superficie Celular/metabolismo , Proteínas Quinasas S6 Ribosómicas/genética , Transducción de Señal/genética , Transmisión Sináptica/genética
15.
Development ; 141(5): 1064-74, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24550114

RESUMEN

Microtubules (MTs) are crucial for diverse biological processes including cell division, cell growth and motility, intracellular transport and the maintenance of cell shape. MT abnormalities are associated with neurodevelopmental and neurodegenerative diseases such as hereditary spastic paraplegia. Among many MT regulators, katanin was the first identified MT-severing protein, but its neuronal functions have not yet been examined in a multicellular organism. Katanin consists of two subunits; the catalytic subunit katanin 60 contains an AAA (ATPases associated with a variety of cellular activities) domain and breaks MT fibers while hydrolyzing ATP, whereas katanin 80 is a targeting and regulatory subunit. To dissect the in vivo functions of Katanin, we generated mutations in Drosophila Katanin 60 and manipulated its expression in a tissue-specific manner. Null mutants of Katanin 60 are pupal lethal, demonstrating that it is essential for viability. Loss-of-function mutants of Katanin 60 showed excess satellite boutons, reduced neurotransmission efficacy, and more enlarged cisternae at neuromuscular junctions. In peripheral sensory neurons, loss of Katanin 60 led to increased elaboration of dendrites, whereas overexpression of Katanin 60 resulted in the opposite. Genetic interaction analyses indicated that increased levels of MT acetylation increase its susceptibility to Katanin-mediated severing in neuronal and non-neuronal systems. Taken together, our results demonstrate for the first time that Katanin 60 is required for the normal development of neuromuscular synapses and dendrites.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Drosophila/metabolismo , Microtúbulos/metabolismo , Unión Neuromuscular/metabolismo , Animales , Dendritas/metabolismo , Drosophila , Histona Desacetilasa 6 , Histona Desacetilasas/metabolismo , Katanina , Unión Neuromuscular/embriología
16.
Nucleic Acids Res ; 42(9): 5765-75, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24728990

RESUMEN

Post-translational modifications (such as ubiquitination) of clock proteins are critical in maintaining the precision and robustness of the evolutionarily conserved circadian clock. Ubiquitination of the core clock transcription factor BMAL1 (brain and muscle Arnt-like 1) has recently been reported. However, it remains unknown whether BMAL1 ubiquitination affects circadian pacemaking and what ubiquitin ligase(s) is involved. Here, we show that activating UBE3A (by expressing viral oncogenes E6/E7) disrupts circadian oscillations in mouse embryonic fibroblasts, measured using PER2::Luc dynamics, and rhythms in endogenous messenger ribonucleic acid and protein levels of BMAL1. Over-expression of E6/E7 reduced the level of BMAL1, increasing its ubiquitination and proteasomal degradation. UBE3A could bind to and degrade BMAL1 in a ubiquitin ligase-dependent manner. This occurred both in the presence and absence of E6/E7. We provide in vitro (knockdown/over-expression in mammalian cells) and in vivo (genetic manipulation in Drosophila) evidence for an endogenous role of UBE3A in regulating circadian dynamics and rhythmic locomotor behaviour. Together, our data reveal an essential and conserved role of UBE3A in the regulation of the circadian system in mammals and flies and identify a novel mechanistic link between oncogene E6/E7-mediated cell transformation and circadian (BMAL1) disruption.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Relojes Circadianos , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitinación , Animales , Proteínas de Drosophila/fisiología , Drosophila melanogaster , Expresión Génica , Regulación de la Expresión Génica , Ratones , Células 3T3 NIH , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis
17.
Proc Natl Acad Sci U S A ; 110(12): 4604-9, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23487739

RESUMEN

Neurons from the brains of Alzheimer's disease (AD) and related tauopathy patients contain neurofibrillary tangles composed of hyperphosphorylated tau protein. Tau normally stabilizes microtubules (MTs); however, tau hyperphosphorylation leads to loss of this function with consequent MT destabilization and neuronal dysfunction. Accordingly, MT-stabilizing drugs such as paclitaxel and epothilone D have been shown as possible therapies for AD and related tauopathies. However, MT-stabilizing drugs have common side effects such as neuropathy and neutropenia. To find previously undescribed suppressors of tau-induced MT defects, we established a Drosophila model ectopically expressing human tau in muscle cells, which allow for clear visualization of the MT network. Overexpressed tau was hyperphosphorylated and resulted in decreased MT density and greater fragmentation, consistent with previous reports in AD patients and mouse models. From a genetic screen, we found that a histone deacetylase 6 (HDAC6) null mutation rescued tau-induced MT defects in both muscles and neurons. Genetic and pharmacological inhibition of the tubulin-specific deacetylase activity of HDAC6 indicates that the rescue effect may be mediated by increased MT acetylation. These findings reveal HDAC6 as a unique potential drug target for AD and related tauopathies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas de Drosophila/metabolismo , Histona Desacetilasas/metabolismo , Mutación , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/terapia , Animales , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster , Epotilonas/farmacología , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Ratones , Ratones Noqueados , Microtúbulos/genética , Microtúbulos/metabolismo , Microtúbulos/patología , Células Musculares/metabolismo , Células Musculares/patología , Neuronas/metabolismo , Neuronas/patología , Paclitaxel/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Moduladores de Tubulina/farmacología , Proteínas tau/genética
18.
PLoS Genet ; 9(4): e1003450, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23593037

RESUMEN

The formation of synapses and the proper construction of neural circuits depend on signaling pathways that regulate cytoskeletal structure and dynamics. After the mutual recognition of a growing axon and its target, multiple signaling pathways are activated that regulate cytoskeletal dynamics to determine the morphology and strength of the connection. By analyzing Drosophila mutations in the cytoplasmic FMRP interacting protein Cyfip, we demonstrate that this component of the WAVE complex inhibits the assembly of filamentous actin (F-actin) and thereby regulates key aspects of synaptogenesis. Cyfip regulates the distribution of F-actin filaments in presynaptic neuromuscular junction (NMJ) terminals. At cyfip mutant NMJs, F-actin assembly was accelerated, resulting in shorter NMJs, more numerous satellite boutons, and reduced quantal content. Increased synaptic vesicle size and failure to maintain excitatory junctional potential amplitudes under high-frequency stimulation in cyfip mutants indicated an endocytic defect. cyfip mutants exhibited upregulated bone morphogenetic protein (BMP) signaling, a major growth-promoting pathway known to be attenuated by endocytosis at the Drosophila NMJ. We propose that Cyfip regulates synapse development and endocytosis by inhibiting actin assembly.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Citoesqueleto , Proteínas de Drosophila , Drosophila melanogaster , Sinapsis , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Mutación , Unión Neuromuscular/metabolismo , Transducción de Señal , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/fisiología
19.
J Neurosci ; 34(8): 2785-96, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24553921

RESUMEN

Fatty acid metabolism plays an important role in brain development and function. Mutations in acyl-CoA synthetase long-chain family member 4 (ACSL4), which converts long-chain fatty acids to acyl-CoAs, result in nonsyndromic X-linked mental retardation. ACSL4 is highly expressed in the hippocampus, a structure critical for learning and memory. However, the underlying mechanism by which mutations of ACSL4 lead to mental retardation remains poorly understood. We report here that dAcsl, the Drosophila ortholog of ACSL4 and ACSL3, inhibits synaptic growth by attenuating BMP signaling, a major growth-promoting pathway at neuromuscular junction (NMJ) synapses. Specifically, dAcsl mutants exhibited NMJ overgrowth that was suppressed by reducing the doses of the BMP pathway components, accompanied by increased levels of activated BMP receptor Thickveins (Tkv) and phosphorylated mothers against decapentaplegic (Mad), the effector of the BMP signaling at NMJ terminals. In addition, Rab11, a small GTPase involved in endosomal recycling, was mislocalized in dAcsl mutant NMJs, and the membrane association of Rab11 was reduced in dAcsl mutant brains. Consistently, the BMP receptor Tkv accumulated in early endosomes but reduced in recycling endosomes in dAcsl mutant NMJs. dAcsl was also required for the recycling of photoreceptor rhodopsin in the eyes, implying a general role for dAcsl in regulating endocytic recycling of membrane receptors. Importantly, expression of human ACSL4 rescued the endocytic trafficking and NMJ phenotypes of dAcsl mutants. Together, our results reveal a novel mechanism whereby dAcsl facilitates Rab11-dependent receptor recycling and provide insights into the pathogenesis of ACSL4-related mental retardation.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Coenzima A Ligasas/farmacología , Sinapsis/efectos de los fármacos , Vesículas Transportadoras/efectos de los fármacos , Animales , Western Blotting , Proteínas Morfogenéticas Óseas/efectos de los fármacos , Drosophila , Proteínas de Drosophila/metabolismo , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Microscopía Electrónica , Músculos/metabolismo , Mutación/genética , Mutación/fisiología , Unión Neuromuscular/efectos de los fármacos , Células Fotorreceptoras de Invertebrados/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Presinapticos/efectos de los fármacos , Rodopsina/metabolismo , Transducción de Señal/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Proteínas de Unión al GTP rab/metabolismo
20.
J Neurosci ; 33(30): 12352-63, 2013 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-23884941

RESUMEN

The precise regulation of synaptic growth is critical for the proper formation and plasticity of functional neural circuits. Identification and characterization of factors that regulate synaptic growth and function have been under intensive investigation. Here we report that brain tumor (brat), which was identified as a translational repressor in multiple biological processes, plays a crucial role at Drosophila neuromuscular junction (NMJ) synapses. Immunohistochemical analysis demonstrated that brat mutants exhibited synaptic overgrowth characterized by excess satellite boutons at NMJ terminals, whereas electron microscopy revealed increased synaptic vesicle size but reduced density at active zones compared with wild-types. Spontaneous miniature excitatory junctional potential amplitudes were larger and evoked quantal content was lower at brat mutant NMJs. In agreement with the morphological and physiological phenotypes, loss of Brat resulted in reduced FM1-43 uptake at the NMJ terminals, indicating that brat regulates synaptic endocytosis. Genetic analysis revealed that the actions of Brat at synapses are mediated through mothers against decapentaplegic (Mad), the signal transduction effector of the bone morphogenetic protein (BMP) signaling pathway. Furthermore, biochemical analyses showed upregulated levels of Mad protein but normal mRNA levels in the larval brains of brat mutants, suggesting that Brat suppresses Mad translation. Consistently, knockdown of brat by RNA interference in Drosophila S2 cells also increased Mad protein level. These results together reveal an important and previously unidentified role for Brat in synaptic development and endocytosis mediated by suppression of BMP signaling.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Endocitosis/fisiología , Unión Neuromuscular/metabolismo , Plasticidad Neuronal/fisiología , Sinapsis/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Colorantes Fluorescentes/farmacocinética , Masculino , Microscopía Electrónica , Mutagénesis , Unión Neuromuscular/ultraestructura , Compuestos de Piridinio/farmacocinética , Compuestos de Amonio Cuaternario/farmacocinética , ARN Interferente Pequeño/genética , Transducción de Señal/fisiología , Sinapsis/ultraestructura , Vesículas Sinápticas/fisiología , Vesículas Sinápticas/ultraestructura , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA