Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 241
Filtrar
1.
Annu Rev Cell Dev Biol ; 34: 495-521, 2018 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-30044649

RESUMO

After an injury in the adult mammalian central nervous system (CNS), lesioned axons fail to regenerate. This failure to regenerate contrasts with axons' remarkable potential to grow during embryonic development and after an injury in the peripheral nervous system (PNS). Several intracellular mechanisms-including cytoskeletal dynamics, axonal transport and trafficking, signaling and transcription of regenerative programs, and epigenetic modifications-control axon regeneration. In this review, we describe how manipulation of intrinsic mechanisms elicits a regenerative response in different organisms and how strategies are implemented to form the basis of a future regenerative treatment after CNS injury.


Assuntos
Axônios/metabolismo , Sistema Nervoso Central/crescimento & desenvolvimento , Regeneração Nervosa/genética , Sistema Nervoso Periférico/crescimento & desenvolvimento , Animais , Transporte Axonal/genética , Axônios/fisiologia , Humanos , Mamíferos
2.
J Biol Chem ; 300(4): 107137, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38447793

RESUMO

Experimental studies in flies, mice, and humans suggest a significant role of impaired axonal transport in the pathogenesis of Alzheimer's disease (AD). The mechanisms underlying these impairments in axonal transport, however, remain poorly understood. Here we report that the Swedish familial AD mutation causes a standstill of the amyloid precursor protein (APP) in the axons at the expense of its reduced anterograde transport. The standstill reflects the perturbed directionality of the axonal transport of APP, which spends significantly more time traveling in the retrograde direction. This ineffective movement is accompanied by an enhanced association of dynactin-1 with APP, which suggests that reduced anterograde transport of APP is the result of enhanced activation of the retrograde molecular motor dynein by dynactin-1. The impact of the Swedish mutation on axonal transport is not limited to the APP vesicles since it also reverses the directionality of a subset of early endosomes, which become enlarged and aberrantly accumulate in distal locations. In addition, it also reduces the trafficking of lysosomes due to their less effective retrograde movement. Altogether, our experiments suggest a pivotal involvement of retrograde molecular motors and transport in the mechanisms underlying impaired axonal transport in AD and reveal significantly more widespread derangement of axonal transport pathways in the pathogenesis of AD.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Transporte Axonal , Animais , Humanos , Camundongos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Transporte Axonal/genética , Axônios/metabolismo , Axônios/patologia , Complexo Dinactina/metabolismo , Complexo Dinactina/genética , Dineínas/metabolismo , Endossomos/metabolismo , Endossomos/genética , Lisossomos/metabolismo , Mutação , Variação Genética
3.
EMBO J ; 40(20): e107158, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34515347

RESUMO

Nucleolin is a multifunctional RNA Binding Protein (RBP) with diverse subcellular localizations, including the nucleolus in all eukaryotic cells, the plasma membrane in tumor cells, and the axon in neurons. Here we show that the glycine arginine rich (GAR) domain of nucleolin drives subcellular localization via protein-protein interactions with a kinesin light chain. In addition, GAR sequences mediate plasma membrane interactions of nucleolin. Both these modalities are in addition to the already reported involvement of the GAR domain in liquid-liquid phase separation in the nucleolus. Nucleolin transport to axons requires the GAR domain, and heterozygous GAR deletion mice reveal reduced axonal localization of nucleolin cargo mRNAs and enhanced sensory neuron growth. Thus, the GAR domain governs axonal transport of a growth controlling RNA-RBP complex in neurons, and is a versatile localization determinant for different subcellular compartments. Localization determination by GAR domains may explain why GAR mutants in diverse RBPs are associated with neurodegenerative disease.


Assuntos
Nucléolo Celular/metabolismo , Gânglios Espinais/metabolismo , Cinesinas/metabolismo , Neurônios/metabolismo , Fosfoproteínas/química , Proteínas de Ligação a RNA/química , Nervo Isquiático/metabolismo , Sequência de Aminoácidos , Animais , Transporte Axonal/genética , Linhagem Celular Tumoral , Nucléolo Celular/ultraestrutura , Gânglios Espinais/citologia , Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Cinesinas/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mutação , Neurônios/citologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Cultura Primária de Células , Domínios Proteicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Nervo Isquiático/citologia , Nucleolina
4.
J Cell Sci ; 136(5)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36655764

RESUMO

Neuronal function depends on axonal transport by kinesin superfamily proteins (KIFs). KIF1A is the molecular motor that transports synaptic vesicle precursors, synaptic vesicles, dense core vesicles and active zone precursors. KIF1A is regulated by an autoinhibitory mechanism; many studies, as well as the crystal structure of KIF1A paralogs, support a model whereby autoinhibited KIF1A is monomeric in solution, whereas activated KIF1A is dimeric on microtubules. KIF1A-associated neurological disorder (KAND) is a broad-spectrum neuropathy that is caused by mutations in KIF1A. More than 100 point mutations have been identified in KAND. In vitro assays show that most mutations are loss-of-function mutations that disrupt the motor activity of KIF1A, whereas some mutations disrupt its autoinhibition and abnormally hyperactivate KIF1A. Studies on disease model worms suggests that both loss-of-function and gain-of-function mutations cause KAND by affecting the axonal transport and localization of synaptic vesicles. In this Review, we discuss how the analysis of these mutations by molecular genetics, single-molecule assays and force measurements have helped to reveal the physiological significance of KIF1A function and regulation, and what physical parameters of KIF1A are fundamental to axonal transport.


Assuntos
Transporte Axonal , Doenças do Sistema Nervoso , Humanos , Transporte Axonal/genética , Transporte Axonal/fisiologia , Cinesinas/genética , Cinesinas/metabolismo , Microtúbulos/metabolismo , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Neurônios/metabolismo , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo
5.
Proc Natl Acad Sci U S A ; 119(32): e2113795119, 2022 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-35917346

RESUMO

KIF1A is a kinesin superfamily motor protein that transports synaptic vesicle precursors in axons. Cargo binding stimulates the dimerization of KIF1A molecules to induce processive movement along microtubules. Mutations in human Kif1a lead to a group of neurodegenerative diseases called KIF1A-associated neuronal disorder (KAND). KAND mutations are mostly de novo and autosomal dominant; however, it is unknown if the function of wild-type KIF1A motors is inhibited by heterodimerization with mutated KIF1A. Here, we have established Caenorhabditis elegans models for KAND using CRISPR-Cas9 technology and analyzed the effects of human KIF1A mutation on axonal transport. In our C. elegans models, both heterozygotes and homozygotes exhibited reduced axonal transport. Suppressor screening using the disease model identified a mutation that recovers the motor activity of mutated human KIF1A. In addition, we developed in vitro assays to analyze the motility of heterodimeric motors composed of wild-type and mutant KIF1A. We find that mutant KIF1A significantly impaired the motility of heterodimeric motors. Our data provide insight into the molecular mechanism underlying the dominant nature of de novo KAND mutations.


Assuntos
Transporte Axonal , Caenorhabditis elegans , Cinesinas , Doenças Neurodegenerativas , Vesículas Sinápticas , Animais , Transporte Axonal/genética , Caenorhabditis elegans/genética , Modelos Animais de Doenças , Genes Dominantes , Humanos , Cinesinas/genética , Atividade Motora/genética , Mutação , Doenças Neurodegenerativas/genética , Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo
6.
Development ; 148(24)2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34940839

RESUMO

It is more than 25 years since the discovery that kinesin 1 is phosphorylated by several protein kinases. However, fundamental questions still remain as to how specific protein kinase(s) contribute to particular motor functions under physiological conditions. Because, within an whole organism, kinase cascades display considerable crosstalk and play multiple roles in cell homeostasis, deciphering which kinase(s) is/are involved in a particular process has been challenging. Previously, we found that GSK3ß plays a role in motor function. Here, we report that a particular site on kinesin 1 motor domain (KHC), S314, is phosphorylated by GSK3ß in vivo. The GSK3ß-phosphomimetic-KHCS314D stalled kinesin 1 motility without dissociating from microtubules, indicating that constitutive GSK3ß phosphorylation of the motor domain acts as a STOP. In contrast, uncoordinated mitochondrial motility was observed in CRISPR/Cas9-GSK3ß non-phosphorylatable-KHCS314A Drosophila larval axons, owing to decreased kinesin 1 attachment to microtubules and/or membranes, and reduced ATPase activity. Together, we propose that GSK3ß phosphorylation fine-tunes kinesin 1 movement in vivo via differential phosphorylation, unraveling the complex in vivo regulatory mechanisms that exist during axonal motility of cargos attached to multiple kinesin 1 and dynein motors.


Assuntos
Movimento Celular/genética , Proteínas de Drosophila/genética , Glicogênio Sintase Quinase 3 beta/genética , Cinesinas/genética , Microtúbulos/genética , Adenosina Trifosfatases/genética , Animais , Transporte Axonal/genética , Axônios/metabolismo , Sistemas CRISPR-Cas/genética , Movimento Celular/fisiologia , Drosophila melanogaster/genética , Dineínas/genética , Larva/genética , Neurônios/metabolismo , Fosforilação/genética , Domínios Proteicos/genética
7.
PLoS Genet ; 17(11): e1009940, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34843479

RESUMO

The UNC-104/KIF1A motor is crucial for axonal transport of synaptic vesicles, but how the UNC-104/KIF1A motor is activated in vivo is not fully understood. Here, we identified point mutations located in the motor domain or the inhibitory CC1 domain, which resulted in gain-of-function alleles of unc-104 that exhibit hyperactive axonal transport and abnormal accumulation of synaptic vesicles. In contrast to the cell body localization of wild type motor, the mutant motors accumulate on neuronal processes. Once on the neuronal process, the mutant motors display dynamic movement similarly to wild type motors. The gain-of-function mutation on the motor domain leads to an active dimeric conformation, releasing the inhibitory CC1 region from the motor domain. Genetically engineered mutations in the motor domain or CC1 of UNC-104, which disrupt the autoinhibitory interface, also led to the gain of function and hyperactivation of axonal transport. Thus, the CC1/motor domain-mediated autoinhibition is crucial for UNC-104/KIF1A-mediated axonal transport in vivo.


Assuntos
Transporte Axonal/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Vesículas Sinápticas/genética , Animais , Caenorhabditis elegans/genética , Mutação com Ganho de Função/genética , Engenharia Genética , Cinesinas/genética , Domínios Proteicos
8.
PLoS Genet ; 17(1): e1009287, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33465062

RESUMO

Huntington's disease is a progressive autosomal dominant neurodegenerative disorder caused by the expansion of a polyglutamine tract at the N-terminus of a large cytoplasmic protein. The Drosophila huntingtin (htt) gene is widely expressed during all developmental stages from embryos to adults. However, Drosophila htt mutant individuals are viable with no obvious developmental defects. We asked if such defects could be detected in htt mutants in a background that had been genetically sensitized to reveal cryptic developmental functions. Amyloid precursor protein (APP) is linked to Alzheimer's disease. Appl is the Drosophila APP ortholog and Appl signaling modulates axon outgrowth in the mushroom bodies (MBs), the learning and memory center in the fly, in part by recruiting Abl tyrosine kinase. Here, we find that htt mutations suppress axon outgrowth defects of αß neurons in Appl mutant MB by derepressing the activity of Abl. We show that Abl is required in MB αß neurons for their axon outgrowth. Importantly, both Abl overexpression and lack of expression produce similar phenotypes in the MBs, indicating the necessity of tightly regulating Abl activity. We find that Htt behaves genetically as a repressor of Abl activity, and consistent with this, in vivo FRET-based measurements reveal a significant increase in Abl kinase activity in the MBs when Htt levels are reduced. Thus, Appl and Htt have essential but opposing roles in MB development, promoting and suppressing Abl kinase activity, respectively, to maintain the appropriate intermediate level necessary for axon growth.


Assuntos
Aciltransferases/genética , Axônios/metabolismo , Proteínas de Drosophila/genética , Proteína Huntingtina/genética , Doença de Huntington/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/genética , Animais , Transporte Axonal/genética , Axônios/patologia , Drosophila melanogaster/genética , Desenvolvimento Embrionário/genética , Humanos , Doença de Huntington/patologia , Aprendizagem/fisiologia , Memória/fisiologia , Corpos Pedunculados/crescimento & desenvolvimento , Corpos Pedunculados/patologia , Mutação/genética , Degeneração Neural/genética , Degeneração Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , Transdução de Sinais/genética
9.
Hum Mol Genet ; 30(16): 1535-1542, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34002226

RESUMO

Parkinson's disease (PD) is a neurodegenerative disease with movement disorders including resting tremor, rigidity, bradykinesia and postural instability. Recent studies have identified a new PD associated gene, TMEM230 (transmembrane protein 230). However, the pathological roles of TMEM230 and its variants are not fully understood. TMEM230 gene encodes two protein isoforms. Isoform2 is the major protein form (~95%) in human. In this study, we overexpress isoform2 TMEM230 variants (WT or PD-linked *184Wext*5 mutant) or knockdown endogenous protein in cultured SH-5Y5Y cells and mouse primary hippocampus neurons to study their pathological roles. We found that overexpression of WT and mutant TMEM230 or knockdown of endogenous TMEM230-induced neurodegeneration and impaired mitochondria transport at the retrograde direction in axons. Mutant TMEM230 caused more severe neurotoxicity and mitochondrial transport impairment than WT-TMEM230 did. Our results demonstrate that maintaining TMEM230 protein levels is critical for neuron survival and axon transport. These findings suggest that mutant-TMEM230-induced mitochondrial transport impairment could be the early event leading to neurite injury and neurodegeneration in PD development.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Animais , Transporte Axonal/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mutação , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/genética
10.
Hum Mol Genet ; 30(1): 5-20, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33395696

RESUMO

FEZ1-mediated axonal transport plays important roles in central nervous system development but its involvement in the peripheral nervous system is not well-characterized. FEZ1 is deleted in Jacobsen syndrome (JS), an 11q terminal deletion developmental disorder. JS patients display impaired psychomotor skills, including gross and fine motor delay, suggesting that FEZ1 deletion may be responsible for these phenotypes, given its association with the development of motor-related circuits. Supporting this hypothesis, our data show that FEZ1 is selectively expressed in the rat brain and spinal cord. Its levels progressively increase over the developmental course of human motor neurons (MN) derived from embryonic stem cells. Deletion of FEZ1 strongly impaired axon and dendrite development, and significantly delayed the transport of synaptic proteins into developing neurites. Concurring with these observations, Drosophila unc-76 mutants showed severe locomotion impairments, accompanied by a strong reduction of synaptic boutons at neuromuscular junctions. These abnormalities were ameliorated by pharmacological activation of UNC-51/ATG1, a FEZ1-activating kinase, with rapamycin and metformin. Collectively, the results highlight a role for FEZ1 in MN development and implicate its deletion as an underlying cause of motor impairments in JS patients.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas do Citoesqueleto/genética , Proteínas de Drosophila/genética , Transtornos Neurológicos da Marcha/genética , Síndrome da Deleção Distal 11q de Jacobsen/genética , Proteínas do Tecido Nervoso/genética , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Transporte Axonal/genética , Encéfalo/metabolismo , Encéfalo/patologia , Transtornos Neurológicos da Marcha/fisiopatologia , Humanos , Síndrome da Deleção Distal 11q de Jacobsen/fisiopatologia , Locomoção/genética , Locomoção/fisiologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Neurogênese/genética , Ratos
11.
Ann Neurol ; 91(5): 716-729, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35178738

RESUMO

OBJECTIVE: The objective of this study is to develop a novel method for monitoring the integrity of motor neurons in vivo by quantifying net retrograde axonal transport. METHODS: The method uses single photon emission computed tomography to quantify retrograde transport to spinal cord of tetanus toxin fragment C (125 I-TTC) following intramuscular injection. We characterized the transport profiles in 3 transgenic mouse models carrying amyotrophic lateral sclerosis (ALS)-associated genes, aging mice, and SOD1G93A transgenic mice following CRISPR/Cas9 gene editing. Lastly, we studied the effect of prior immunization of tetanus toxoid on the transport profile of TTC. RESULTS: This technique defines a quantitative profile of net retrograde axonal transport of TTC in living mice. The profile is distinctly abnormal in transgenic SOD1G93A mice as young as 65 days (presymptomatic) and worsens with disease progression. Moreover, this method detects a distinct therapeutic benefit of gene editing in transgenic SOD1G93A mice well before other clinical parameters (eg, grip strength) show improvement. Symptomatic transgenic PFN1C71G/C71G ALS mice display gross reductions in net retrograde axonal transport, which is also disturbed in asymptomatic mice harboring a human C9ORF72 transgene with an expanded GGGGCC repeat motif. In wild-type mice, net retrograde axonal transport declines with aging. Lastly, prior immunization with tetanus toxoid does not preclude use of this assay. INTERPRETATION: This assay of net retrograde axonal transport has broad potential clinical applications and should be particularly valuable as a physiological biomarker that permits early detection of benefit from potential therapies for motor neuron diseases. ANN NEUROL 2022;91:716-729.


Assuntos
Esclerose Lateral Amiotrófica , Transporte Axonal , Esclerose Lateral Amiotrófica/diagnóstico por imagem , Esclerose Lateral Amiotrófica/genética , Animais , Transporte Axonal/genética , Biomarcadores , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Profilinas , Medula Espinal/diagnóstico por imagem , Medula Espinal/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1 , Toxoide Tetânico
12.
PLoS Biol ; 18(12): e3000621, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33351792

RESUMO

Neurons extend long axons that require maintenance and are susceptible to degeneration. Long-term integrity of axons depends on intrinsic mechanisms including axonal transport and extrinsic support from adjacent glial cells. The mechanisms of support provided by myelinating oligodendrocytes to underlying axons are only partly understood. Oligodendrocytes release extracellular vesicles (EVs) with properties of exosomes, which upon delivery to neurons improve neuronal viability in vitro. Here, we show that oligodendroglial exosome secretion is impaired in 2 mouse mutants exhibiting secondary axonal degeneration due to oligodendrocyte-specific gene defects. Wild-type oligodendroglial exosomes support neurons by improving the metabolic state and promoting axonal transport in nutrient-deprived neurons. Mutant oligodendrocytes release fewer exosomes, which share a common signature of underrepresented proteins. Notably, mutant exosomes lack the ability to support nutrient-deprived neurons and to promote axonal transport. Together, these findings indicate that glia-to-neuron exosome transfer promotes neuronal long-term maintenance by facilitating axonal transport, providing a novel mechanistic link between myelin diseases and secondary loss of axonal integrity.


Assuntos
Transporte Axonal/fisiologia , Neurônios/metabolismo , Oligodendroglia/metabolismo , Animais , Transporte Axonal/genética , Axônios/fisiologia , Exossomos/metabolismo , Exossomos/fisiologia , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/fisiologia , Feminino , Células HEK293 , Humanos , Manutenção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/metabolismo , Neuroglia , Neurônios/fisiologia , Oligodendroglia/fisiologia , Transdução de Sinais/fisiologia
13.
Semin Cell Dev Biol ; 99: 133-150, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31542222

RESUMO

Because of the extremely polarized morphology, the proper functioning of neurons largely relies on the efficient cargo transport along the axon. Axonal transport defects have been reported in multiple neurodegenerative diseases as an early pathological feature. The discovery of mutations in human genes involved in the transport machinery provide a direct causative relationship between axonal transport defects and neurodegeneration. Here, we summarize the current genetic findings related to axonal transport in neurodegenerative diseases, and we discuss the relationship between axonal transport defects and other pathological changes observed in neurodegeneration. In addition, we summarize the therapeutic approaches targeting the axonal transport machinery in studies of neurodegenerative diseases. Finally, we review the technical advances in tracking axonal transport both in vivo and in vitro.


Assuntos
Transporte Axonal/efeitos dos fármacos , Transporte Axonal/genética , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/genética , Animais , Humanos , Mutação , Doenças Neurodegenerativas/metabolismo
14.
Proc Natl Acad Sci U S A ; 116(37): 18429-18434, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31455732

RESUMO

KIF1A is a kinesin family motor involved in the axonal transport of synaptic vesicle precursors (SVPs) along microtubules (MTs). In humans, more than 10 point mutations in KIF1A are associated with the motor neuron disease hereditary spastic paraplegia (SPG). However, not all of these mutations appear to inhibit the motility of the KIF1A motor, and thus a cogent molecular explanation for how KIF1A mutations lead to neuropathy is not available. In this study, we established in vitro motility assays with purified full-length human KIF1A and found that KIF1A mutations associated with the hereditary SPG lead to hyperactivation of KIF1A motility. Introduction of the corresponding mutations into the Caenorhabditis elegans KIF1A homolog unc-104 revealed abnormal accumulation of SVPs at the tips of axons and increased anterograde axonal transport of SVPs. Our data reveal that hyperactivation of kinesin motor activity, rather than its loss of function, is a cause of motor neuron disease in humans.


Assuntos
Transporte Axonal/genética , Predisposição Genética para Doença/genética , Cinesinas/genética , Cinesinas/metabolismo , Mutação , Vesículas Sinápticas/metabolismo , Animais , Axônios/metabolismo , Caenorhabditis elegans/genética , Humanos , Doença dos Neurônios Motores/genética , Paraplegia Espástica Hereditária/genética
15.
J Neurosci ; 40(3): 542-556, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31754011

RESUMO

Synapse formation, maturation, and turnover require a finely regulated transport system that delivers selected cargos to specific synapses. However, the supporting mechanisms of this process are not fully understood. The present study unravels a new molecular system for vesicle-based axonal transport of proteins in male and female flies (Drosophila melanogaster). Here, we identify the gene CG14579 as the transcription unit corresponding to the regulatory mutations known as central complex broad (ccb). These mutations were previously isolated for their morphological phenotype in R-neurons of the ellipsoid body, a component of the central complex. Mutant axons from R-neurons fail to cross the midline, which is indicative of an aberrant composition of the growth cone. However, the molecular mechanism remained to be deciphered. In this manuscript, we show that CCB is involved in axonal trafficking of FasII and synaptobrevin, but not syntaxin. These results suggest that axonal transport of certain proteins is required for the correct pathfinding of R-neurons. We further investigated the molecular network supporting the CCB system and found that CCB colocalizes and coimmunoprecipitates with Rab11. Epistasis studies indicated that Rab11 is positioned downstream of CCB within this axonal transport system. Interestingly, ccb also interacts with actin and the actin nucleator spire The data revealed that this interaction plays a key role in the development of axonal connections within the ellipsoid body. We propose that the CCB/Rab11/SPIRE system regulates axonal trafficking of synaptic proteins required for proper connectivity and synaptic function.SIGNIFICANCE STATEMENT Proper function of the nervous system requires the establishment of mature, functional synapses. Differential protein composition in the synapse enables optimal performance of cognitive tasks. Therefore, it is critical to have a finely regulated transport system to deliver selected synaptic proteins to synapses. Remarkably, impairments in cytoskeleton-based protein-transport systems often underlie cognitive deficits, such as those associated with aging and neurodegenerative diseases. This study reveals that CCB is part of a novel transport system that delivers certain synaptic proteins via the actin cytoskeleton within the Rab11-related domain of slow recycling endosomes.


Assuntos
Actinas/fisiologia , Transporte Axonal/genética , Transporte Axonal/fisiologia , Proteínas de Drosophila/fisiologia , Proteínas de Membrana/fisiologia , Sinapses/fisiologia , Animais , Animais Geneticamente Modificados , Axônios/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster , Retículo Endoplasmático/metabolismo , Feminino , Cones de Crescimento/fisiologia , Masculino , Proteínas de Membrana/genética , Mutação/genética , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/fisiologia
16.
J Biol Chem ; 295(20): 6926-6935, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32249210

RESUMO

Mouse hepatitis virus (MHV; murine coronavirus) causes meningoencephalitis, myelitis, and optic neuritis followed by axonal loss and demyelination. This murine virus is used as a common model to study acute and chronic virus-induced demyelination in the central nervous system. Studies with recombinant MHV strains that differ in the gene encoding the spike protein have demonstrated that the spike has a role in MHV pathogenesis and retrograde axonal transport. Fusion peptides (FPs) in the spike protein play a key role in MHV pathogenesis. In a previous study of the effect of deleting a single proline residue in the FP of a demyelinating MHV strain, we found that two central, consecutive prolines are important for cell-cell fusion and pathogenesis. The dihedral fluctuation of the FP was shown to be repressed whenever two consecutive prolines were present, in contrast to the presence of a single proline in the chain. Using this proline-deleted MHV strain, here we investigated whether intracranial injection of this strain can induce optic neuritis by retrograde axonal transport from the brain to the retina through the optic nerve. We observed that the proline-deleted recombinant MHV strain is restricted to the optic nerve, is unable to translocate to the retina, and causes only minimal demyelination and no neuronal death. We conclude that an intact proline dyad in the FP of the recombinant demyelinating MHV strain plays a crucial role in translocation of the virus through axons and subsequent neurodegeneration.


Assuntos
Transporte Axonal/genética , Vírus da Hepatite Murina/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Animais , Transporte Axonal/fisiologia , Axônios/metabolismo , Axônios/virologia , Encéfalo/metabolismo , Infecções por Coronavirus/patologia , Doenças Desmielinizantes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vírus da Hepatite Murina/metabolismo , Nervo Óptico/metabolismo , Nervo Óptico/virologia , Peptídeos/metabolismo , Prolina/metabolismo , Deleção de Sequência/genética , Glicoproteína da Espícula de Coronavírus/genética , Proteínas do Envelope Viral/metabolismo
17.
Int J Mol Sci ; 22(18)2021 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-34575995

RESUMO

Amyotrophic Lateral Sclerosis (ALS), neurodegenerative motor neuron disorder is characterized as multisystem disease with important contribution of genetic factors. The etiopahogenesis of ALS is not fully elucidate, but the dominant theory at present relates to RNA processing, as well as protein aggregation and miss-folding, oxidative stress, glutamate excitotoxicity, inflammation and epigenetic dysregulation. Additionally, as mitochondria plays a leading role in cellular homeostasis maintenance, a rising amount of evidence indicates mitochondrial dysfunction as a substantial contributor to disease onset and progression. The aim of this review is to summarize most relevant findings that link genetic factors in ALS pathogenesis with different mechanisms with mitochondrial involvement (respiratory chain, OXPHOS control, calcium buffering, axonal transport, inflammation, mitophagy, etc.). We highlight the importance of a widening perspective for better understanding overlapping pathophysiological pathways in ALS and neurodegeneration in general. Finally, current and potentially novel therapies, especially gene specific therapies, targeting mitochondrial dysfunction are discussed briefly.


Assuntos
Esclerose Lateral Amiotrófica , Mitocôndrias , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/terapia , Animais , Transporte Axonal/genética , Cálcio/metabolismo , Transporte de Elétrons/genética , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mitofagia/genética , Fosforilação Oxidativa , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo
18.
J Neurosci ; 39(40): 7976-7991, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31363064

RESUMO

Alzheimer's disease (AD) is associated with the cleavage of the amyloid precursor protein (APP) to produce the toxic amyloid-ß (Aß) peptide. Accumulation of Aß, together with the concomitant inflammatory response, ultimately leads to neuronal death and cognitive decline. Despite AD progression being underpinned by both neuronal and immunological components, therapeutic strategies based on dual targeting of these systems remains unexplored. Here, we report that inactivation of the p110δ isoform of phosphoinositide 3-kinase (PI3K) reduces anterograde axonal trafficking of APP in hippocampal neurons and dampens secretion of the inflammatory cytokine tumor necrosis factor-alpha by microglial cells in the familial AD APPswe/PS1ΔE9 (APP/PS1) mouse model. Moreover, APP/PS1 mice with kinase-inactive PI3Kδ (δD910A) had reduced Aß peptides levels and plaques in the brain and an abrogated inflammatory response compared with APP/PS1 littermates. Mechanistic investigations reveal that PI3Kδ inhibition decreases the axonal transport of APP by eliciting the formation of highly elongated tubular-shaped APP-containing carriers, reducing the levels of secreted Aß peptide. Importantly, APP/PS1/δD910A mice exhibited no spatial learning or memory deficits. Our data highlight inhibition of PI3Kδ as a new approach to protect against AD pathology due to its dual action of dampening microglial-dependent neuroinflammation and reducing plaque burden by inhibition of neuronal APP trafficking and processing.SIGNIFICANCE STATEMENT During Alzheimer's disease (AD), the accumulation of the toxic amyloid-ß (Aß) peptide in plaques is associated with a chronic excessive inflammatory response. Uncovering new drug targets that simultaneously reduce both Aß plaque load and neuroinflammation holds therapeutic promise. Using a combination of genetic and pharmacological approaches, we found that the p110δ isoform of phosphoinositide 3-kinase (PI3K) is involved in anterograde trafficking of the amyloid precursor protein in neurons and in the secretion of tumor necrosis factor-alpha from microglial cells. Genetic inactivation of PI3Kδ reduces Aß plaque deposition and abrogates the inflammatory response, resulting in a complete rescue of the life span and spatial memory performance. We conclude that inhibiting PI3Kδ represents a novel therapeutic approach to ameliorate AD pathology by dampening plaque accumulation and microglial-dependent neuroinflammation.


Assuntos
Doença de Alzheimer/prevenção & controle , Precursor de Proteína beta-Amiloide/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/genética , Disfunção Cognitiva/genética , Disfunção Cognitiva/prevenção & controle , Encefalite/genética , Encefalite/prevenção & controle , Placa Amiloide/genética , Placa Amiloide/prevenção & controle , Fator de Necrose Tumoral alfa/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Transporte Axonal/genética , Citocinas/metabolismo , Feminino , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/metabolismo , Mutação Puntual , Cultura Primária de Células , Memória Espacial
19.
J Virol ; 93(7)2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30651360

RESUMO

Following its entry into cells, pseudorabies virus (PRV) utilizes microtubules to deliver its nucleocapsid to the nucleus. Previous studies have shown that PRV VP1/2 is an effector of dynein-mediated capsid transport. However, the mechanism of PRV for recruiting microtubule motor proteins for successful neuroinvasion and neurovirulence is not well understood. Here, we provide evidence that PRV pUL21 is an inner tegument protein. We tested its interaction with the cytoplasmic light chains using a bimolecular fluorescence complementation (BiFC) assay and observed that PRV pUL21 interacts with Roadblock-1. This interaction was confirmed by coimmunoprecipitation (co-IP) assays. We also determined the efficiency of retrograde and anterograde axonal transport of PRV strains in explanted neurons using a microfluidic chamber system and investigated pUL21's contribution to PRV neuroinvasion in vivo Further data showed that the carboxyl terminus of pUL21 is essential for its interaction with Roadblock-1, and this domain contributes to PRV retrograde axonal transport in vitro and in vivo Our findings suggest that the carboxyl terminus of pUL21 contributes to PRV neuroinvasion.IMPORTANCE Herpesviruses are a group of DNA viruses that infect both humans and animals. Alphaherpesviruses are distinguished by their ability to establish latent infection in peripheral neurons. After entering neurons, the herpesvirus capsid interacts with cellular motor proteins and undergoes retrograde transport on axon microtubules. This elaborate process is vital to the herpesvirus lifecycle, but the underlying mechanism remains poorly understood. Here, we determined that pUL21 is an inner tegument protein of pseudorabies virus (PRV) and that it interacts with the cytoplasmic dynein light chain Roadblock-1. We also observed that pUL21 promotes retrograde transport of PRV in neuronal cells. Furthermore, our findings confirm that pUL21 contributes to PRV neuroinvasion in vivo Importantly, the carboxyl terminus of pUL21 is responsible for interaction with Roadblock-1, and this domain contributes to PRV neuroinvasion. This study offers fresh insights into alphaherpesvirus neuroinvasion and the interaction between virus and host during PRV infection.


Assuntos
Proteínas do Capsídeo/genética , Herpesvirus Suídeo 1/genética , Herpesvirus Suídeo 1/patogenicidade , Neurônios/virologia , Transporte Axonal/genética , Axônios/virologia , Linhagem Celular , Linhagem Celular Tumoral , Dineínas/genética , Células HEK293 , Células HeLa , Humanos , Microtúbulos/genética , Microtúbulos/virologia , Nucleocapsídeo/genética , Replicação Viral/genética
20.
Mol Cell Proteomics ; 17(11): 2091-2106, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30038033

RESUMO

mRNA translation in axons enables neurons to introduce new proteins at sites distant from their cell body. mRNA-protein interactions drive this post-transcriptional regulation, yet knowledge of RNA binding proteins (RBP) in axons is limited. Here we used proteomics to identify RBPs interacting with the axonal localizing motifs of Nrn1, Hmgb1, Actb, and Gap43 mRNAs, revealing many novel RBPs in axons. Interestingly, no RBP is shared between all four RNA motifs, suggesting graded and overlapping specificities of RBP-mRNA pairings. A systematic assessment of axonal mRNAs interacting with hnRNP H1, hnRNP F, and hnRNP K, proteins that bound with high specificity to Nrn1 and Hmgb1, revealed that axonal mRNAs segregate into axon growth-associated RNA regulons based on hnRNP interactions. Axotomy increases axonal transport of hnRNPs H1, F, and K, depletion of these hnRNPs decreases axon growth and reduces axonal mRNA levels and axonal protein synthesis. Thus, subcellular hnRNP-interacting RNA regulons support neuronal growth and regeneration.


Assuntos
Axônios/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Motivos de Nucleotídeos/genética , RNA Mensageiro/genética , Regulon/genética , Regiões 5' não Traduzidas/genética , Animais , Transporte Axonal/genética , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Masculino , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Ligação Proteica , Biossíntese de Proteínas , Transporte de RNA/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA