Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 152(3): 479-91, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23374344

RESUMO

Fast axonal transport (FAT) requires consistent energy over long distances to fuel the molecular motors that transport vesicles. We demonstrate that glycolysis provides ATP for the FAT of vesicles. Although inhibiting ATP production from mitochondria did not affect vesicles motility, pharmacological or genetic inhibition of the glycolytic enzyme GAPDH reduced transport in cultured neurons and in Drosophila larvae. GAPDH localizes on vesicles via a huntingtin-dependent mechanism and is transported on fast-moving vesicles within axons. Purified motile vesicles showed GAPDH enzymatic activity and produced ATP. Finally, we show that vesicular GAPDH is necessary and sufficient to provide on-board energy for fast vesicular transport. Although detaching GAPDH from vesicles reduced transport, targeting GAPDH to vesicles was sufficient to promote FAT in GAPDH deficient neurons. This specifically localized glycolytic machinery may supply constant energy, independent of mitochondria, for the processive movement of vesicles over long distances in axons.


Assuntos
Transporte Axonal , Drosophila melanogaster/metabolismo , Glicólise , Neurônios/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Axônios/metabolismo , Encéfalo/citologia , Células Cultivadas , Drosophila melanogaster/crescimento & desenvolvimento , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Camundongos , Mitocôndrias/metabolismo , Ratos
2.
Mol Psychiatry ; 27(3): 1805-1815, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35165396

RESUMO

Sensorimotor information processing underlies normal cognitive and behavioral traits and has classically been evaluated through prepulse inhibition (PPI) of a startle reflex. PPI is a behavioral dimension deregulated in several neurological and psychiatric disorders, yet the mechanisms underlying the cross-diagnostic nature of PPI deficits across these conditions remain to be understood. To identify circuitry mechanisms for PPI, we performed circuitry recording over the prefrontal cortex and striatum, two brain regions previously implicated in PPI, using wild-type (WT) mice compared to Disc1-locus-impairment (LI) mice, a model representing neuropsychiatric conditions. We demonstrated that the corticostriatal projection regulates neurophysiological responses during the PPI testing in WT, whereas these circuitry responses were disrupted in Disc1-LI mice. Because our biochemical analyses revealed attenuated brain-derived neurotrophic factor (Bdnf) transport along the corticostriatal circuit in Disc1-LI mice, we investigated the potential role of Bdnf in this circuitry for regulation of PPI. Virus-mediated delivery of Bdnf into the striatum rescued PPI deficits in Disc1-LI mice. Pharmacologically augmenting Bdnf transport by chronic lithium administration, partly via phosphorylation of Huntingtin (Htt) serine-421 and its integration into the motor machinery, restored striatal Bdnf levels and rescued PPI deficits in Disc1-LI mice. Furthermore, reducing the cortical Bdnf expression negated this rescuing effect of lithium, confirming the key role of Bdnf in lithium-mediated PPI rescuing. Collectively, the data suggest that striatal Bdnf supply, collaboratively regulated by Htt and Disc1 along the corticostriatal circuit, is involved in sensorimotor gating, highlighting the utility of dimensional approach in investigating pathophysiological mechanisms across neuropsychiatric disorders.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Corpo Estriado , Proteínas do Tecido Nervoso , Córtex Pré-Frontal , Inibição Pré-Pulso , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corpo Estriado/metabolismo , Humanos , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Córtex Pré-Frontal/metabolismo , Inibição Pré-Pulso/fisiologia , Reflexo de Sobressalto/fisiologia , Filtro Sensorial/fisiologia
3.
Small ; 15(47): e1902796, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31583817

RESUMO

Single-particle tracking with quantum dots (QDs) constitutes a powerful tool to track the nanoscopic dynamics of individual cell membrane components unveiling their membrane diffusion characteristics. Here, the nano-resolved population dynamics of QDs is exploited to reconstruct the topography and structural changes of the cell membrane surface with high temporal and spatial resolution. For this proof-of-concept study, bright, small, and stable biofunctional QD nanoconstructs are utilized recognizing the endogenous neuronal cannabinoid receptor 1, a highly expressed and fast-diffusing membrane protein, together with a commercial point-localization microscope. Rapid QD diffusion on the axonal plasma membrane of cultured hippocampal neurons allows precise reconstruction of the membrane surface in less than 1 min with a spatial resolution of tens of nanometers. Access of the QD nanoconstructs to the synaptic cleft enables rapid 3D topological reconstruction of the entire presynaptic component. Successful reconstruction of membrane nano-topology and deformation at the second time-scale is also demonstrated for HEK293 cell filopodia and axons. Named "nanoPaint," this super-resolution imaging technique amenable to any endogenous transmembrane target represents a versatile platform to rapidly and accurately reconstruct the cell membrane nano-topography, thereby enabling the study of the rapid dynamic phenomena involved in neuronal membrane plasticity.


Assuntos
Membrana Celular/química , Imageamento Tridimensional , Nanopartículas/química , Nanotecnologia/métodos , Animais , Células HEK293 , Humanos , Proteínas de Membrana/metabolismo , Pontos Quânticos
4.
J Neurosci ; 33(15): 6298-309, 2013 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-23575829

RESUMO

Huntingtin (HTT), the protein mutated in Huntington's disease (HD), controls transport of the neurotrophin, brain-derived neurotrophic factor (BDNF), within corticostriatal neurons. Transport and delivery of BDNF to the striatum are reduced in disease, which contributes to striatal neuron degeneration. BDNF released by cortical neurons activates TrkB receptors at striatal dendrites to promote striatum survival. However, it remains to be determined whether transport of TrkB, the BDNF receptor, depends on HTT and whether such transport is altered in mutant situation. Here we show that TrkB binds to and colocalizes with HTT and dynein. Silencing HTT reduces vesicular transport of TrkB in striatal neurons. In HD, the polyQ expansion in HTT alters the binding of TrkB-containing vesicles to microtubules and reduces transport. Using a combination of microfluidic devices that isolate dendrites from cell bodies and BDNF coupled to quantum dots, we selectively analyzed TrkB retrograde transport in response to BDNF stimulation at dendrite terminals. We show that the retrograde transport of TrkB vesicles within striatal dendrites and the BDNF/TrkB-induced signaling through ERK phosphorylation and c-fos induction are decreased in neurons from an HD mouse model. Together, our findings demonstrate that HTT is a crucial regulator of TrkB trafficking. Transport defects in HD are not restricted to BDNF transport in cortical neurons but also affect trafficking of its ligand-bound receptor in the striatal neurons. This transport alteration may further impair BDNF-TrkB survival signaling within the corticostriatal connection that is most affected in HD.


Assuntos
Corpo Estriado/metabolismo , Dendritos/metabolismo , Doença de Huntington/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Receptor trkB/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Linhagem Celular , Modelos Animais de Doenças , Dineínas/metabolismo , Proteína Huntingtina , Doença de Huntington/genética , Camundongos , Microtúbulos/metabolismo , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Cultura Primária de Células , Transporte Proteico , Ratos , Transdução de Sinais/genética , Vesículas Transportadoras/genética , Vesículas Transportadoras/metabolismo
5.
Sci Adv ; 10(22): eado0077, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38809980

RESUMO

While our understanding of the nanoscale architecture of anterograde synaptic transmission is rapidly expanding, the qualitative and quantitative molecular principles underlying distinct mechanisms of retrograde synaptic communication remain elusive. We show that a particular form of tonic cannabinoid signaling is essential for setting target cell-dependent synaptic variability. It does not require the activity of the two major endocannabinoid-producing enzymes. Instead, by developing a workflow for physiological, anatomical, and molecular measurements at the same unitary synapse, we demonstrate that the nanoscale stoichiometric ratio of type 1 cannabinoid receptors (CB1Rs) to the release machinery is sufficient to predict synapse-specific release probability. Accordingly, selective decrease of extrasynaptic CB1Rs does not affect synaptic transmission, whereas in vivo exposure to the phytocannabinoid Δ9-tetrahydrocannabinol disrupts the intrasynaptic nanoscale stoichiometry and reduces synaptic variability. These findings imply that synapses leverage the nanoscale stoichiometry of presynaptic receptor coupling to the release machinery to establish synaptic strength in a target cell-dependent manner.


Assuntos
Receptor CB1 de Canabinoide , Transdução de Sinais , Sinapses , Transmissão Sináptica , Animais , Transmissão Sináptica/efeitos dos fármacos , Receptor CB1 de Canabinoide/metabolismo , Sinapses/metabolismo , Terminações Pré-Sinápticas/metabolismo , Camundongos , Endocanabinoides/metabolismo , Endocanabinoides/farmacologia , Dronabinol/farmacologia
6.
J Cell Biol ; 222(12)2023 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-37902728

RESUMO

In response to repulsive cues, axonal growth cones can quickly retract. This requires the prompt activity of contractile actomyosin, which is formed by the non-muscle myosin II (NMII) bound to actin filaments. NMII is a molecular motor that provides the necessary mechanical force at the expense of ATP. Here, we report that this process is energetically coupled to glycolysis and is independent of cellular ATP levels. Induction of axonal retraction requires simultaneous generation of ATP by glycolysis, as shown by chemical inhibition and genetic knock-down of GAPDH. Co-immunoprecipitation and proximal-ligation assay showed that actomyosin associates with ATP-generating glycolytic enzymes and that this association is strongly enhanced during retraction. Using microfluidics, we confirmed that the energetic coupling between glycolysis and actomyosin necessary for axonal retraction is localized to the growth cone and near axonal shaft. These results indicate a tight coupling between on-demand energy production by glycolysis and energy consumption by actomyosin contraction suggesting a function of glycolysis in axonal guidance.


Assuntos
Actomiosina , Axônios , Glicólise , Citoesqueleto de Actina , Actomiosina/metabolismo , Trifosfato de Adenosina , Axônios/fisiologia
7.
Neurobiol Dis ; 45(2): 786-95, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22127389

RESUMO

Mecp2 deficiency or overexpression causes a wide spectrum of neurological diseases in humans among which Rett Syndrome is the prototype. Pathogenic mechanisms are thought to involve transcriptional deregulation of target genes such as Bdnf together with defects in the general transcriptional program of affected cells. Here we found that two master genes, Huntingtin (Htt) and huntingtin-associated protein (Hap1), involved in the control of Bdnf axonal transport, are altered in the brain of Mecp2-deficient mice. We also revealed an in vivo defect of Bdnf transport throughout the cortico striatal pathway of Mecp2-deficient animals. We found that the velocity of Bdnf-containing vesicles is reduced in vitro in the Mecp2-deficient axons and this deficit can be rescued by the re-expression of Mecp2. The defect in axonal transport is not restricted to Bdnf since transport of the amyloid precursor protein (App) that is Htt and Hap1-dependent is also altered. Finally, treating Mecp2-deficient mice with cysteamine, a molecule increasing the secretion of Bdnf vesicles, improved the lifespan and reduced motor defects, suggesting a new therapeutic strategy for Rett syndrome.


Assuntos
Transporte Axonal/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína 2 de Ligação a Metil-CpG/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Transdução de Sinais , Animais , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/genética , Modelos Animais de Doenças , Imunofluorescência , Perfilação da Expressão Gênica , Proteína Huntingtina , Imuno-Histoquímica , Masculino , Proteína 2 de Ligação a Metil-CpG/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Transporte Proteico/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Transfecção
8.
EMBO J ; 27(15): 2124-34, 2008 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-18615096

RESUMO

The transport of vesicles in neurons is a highly regulated process, with vesicles moving either anterogradely or retrogradely depending on the nature of the molecular motors, kinesins and dynein, respectively, which propel vesicles along microtubules (MTs). However, the mechanisms that determine the directionality of transport remain unclear. Huntingtin, the protein mutated in Huntington's disease, is a positive regulatory factor for vesicular transport. Huntingtin is phosphorylated at serine 421 by the kinase Akt but the role of this modification is unknown. Here, we demonstrate that phosphorylation of wild-type huntingtin at S421 is crucial to control the direction of vesicles in neurons. When phosphorylated, huntingtin recruits kinesin-1 to the dynactin complex on vesicles and MTs. Using brain-derived neurotrophic factor as a marker of vesicular transport, we demonstrate that huntingtin phosphorylation promotes anterograde transport. Conversely, when huntingtin is not phosphorylated, kinesin-1 detaches and vesicles are more likely to undergo retrograde transport. This also applies to other vesicles suggesting an essential role for huntingtin in the control of vesicular directionality in neurons.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Animais , Transporte Biológico Ativo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Células Cultivadas , Vesículas Citoplasmáticas/metabolismo , Complexo Dinactina , Humanos , Proteína Huntingtina , Cinesinas/fisiologia , Camundongos , Proteínas Associadas aos Microtúbulos/fisiologia , Microtúbulos/metabolismo , Fosforilação , Ratos , Proteínas de Transporte Vesicular/fisiologia
9.
Hum Mol Genet ; 17(24): 3837-46, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18772195

RESUMO

Huntingtin (htt), the protein mutated in Huntington's disease, is a positive regulatory factor for vesicular transport whose function is lost in disease. Here, we demonstrate that phosphorylation of htt at serine 421 (S421) restores its function in axonal transport. Using a strategy involving RNA (ribonucleic acid) interference and re-expression of various constructs, we show that polyQ (polyglutamine)-htt is unable to promote transport of brain-derived neurotrophic factor (BDNF)-containing vesicles, but polyQ-htt constitutively phosphorylated at S421 is as effective as the wild-type (wt) as concerns transport of these vesicles. The S421 phosphorylated polyQ-htt displays the wt function of inducing BDNF release. Phosphorylation restores the interaction between htt and the p150(Glued) subunit of dynactin and their association with microtubules in vitro and in cells. We also show that the IGF-1 (insulin growth factor type I)/Akt pathway by promoting htt phosphorylation compensates for the transport defect. This is the first description of a mechanism that restores the htt function altered in disease.


Assuntos
Transporte Axonal/genética , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Serina/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Linhagem Celular , Células Cultivadas , Vetores Genéticos/genética , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/fisiopatologia , Camundongos , Neurônios/fisiologia , Peptídeos/metabolismo , Fosforilação , Transporte Proteico/genética , Ratos
10.
Front Cell Neurosci ; 14: 180, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32754013

RESUMO

By limiting protein exchange between the soma and the axon, the axon initial segment (AIS) enables the segregation of specific proteins and hence the differentiation of the somatodendritic compartment and the axonal compartment. Electron microscopy and super-resolution fluorescence imaging have provided important insights in the ultrastructure of the AIS. Yet, the full extent of its filtering properties is not fully delineated. In particular, it is unclear whether and how the AIS opposes the free exchange of soluble proteins. Here we describe a robust framework to combine whole-cell photobleaching and retrospective high-resolution imaging in developing neurons. With this assay, we found that cytoplasmic soluble proteins that are not excluded from the axon upon expression over tens of hours exhibit a strong mobility reduction at the AIS - i.e., are indeed compartmentalized - when monitored over tens of minutes. This form of compartmentalization is developmentally regulated, requires intact F-actin and may be correlated with the composition and ultrastructure of the submembranous spectrin cytoskeleton. Using computational modeling, we provide evidence that both neuronal morphology and the AIS regulate this compartmentalization but act on distinct time scales, with the AIS having a more pronounced effect on fast exchanges. Our results thus suggest that the rate of protein accumulation in the soma may impact to what extent and over which timescales freely moving molecules can be segregated from the axon. This in turn has important implications for our understanding of compartment-specific signaling in neurons.

11.
J Neurosci ; 28(39): 9723-31, 2008 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-18815258

RESUMO

Gene expression changes are a hallmark of the neuropathology of Huntington's disease (HD), but the exact molecular mechanisms of this effect remain uncertain. Here, we report that in vitro models of disease comprised of primary striatal neurons expressing N-terminal fragments of mutant huntingtin (via lentiviral gene delivery) faithfully reproduce the gene expression changes seen in human HD. Neither viral infection nor unrelated (enhanced green fluorescent protein) transgene expression had a major effect on resultant RNA profiles. Expression of a wild-type fragment of huntingtin [htt171-18Q] also caused only a small number of RNA changes. The disease-related signal in htt171-82Q versus htt171-18Q comparisons was far greater, resulting in the differential detection of 20% of all mRNA probe sets. Transcriptomic effects of mutated htt171 are time- and polyglutamine-length dependent and occur in parallel with other manifestations of polyglutamine toxicity over 4-8 weeks. Specific RNA changes in htt171-82Q-expressing striatal cells accurately recapitulated those observed in human HD caudate and included decreases in PENK (proenkephalin), RGS4 (regulator of G-protein signaling 4), dopamine D(1) receptor (DRD1), DRD2, CNR1 (cannabinoid CB(1) receptor), and DARPP-32 (dopamine- and cAMP-regulated phosphoprotein-32; also known as PPP1R1B) mRNAs. HD-related transcriptomic changes were also observed in primary neurons expressing a longer fragment of mutant huntingtin (htt853-82Q). The gene expression changes observed in cultured striatal neurons are not secondary to abnormalities of neuronal firing or glutamatergic, dopaminergic, or brain-derived neurotrophic factor signaling, thereby demonstrating that HD-induced dysregulation of the striatal transcriptome might be attributed to intrinsic effects of mutant huntingtin.


Assuntos
Regulação da Expressão Gênica/fisiologia , Doença de Huntington/genética , Neurônios/fisiologia , Animais , Corpo Estriado/patologia , Modelos Animais de Doenças , Embrião de Mamíferos , Encefalinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lentivirus/fisiologia , Análise em Microsséries , Mutação , Neurônios/efeitos dos fármacos , Peptídeos/farmacologia , Fosfoproteínas/metabolismo , Precursores de Proteínas/metabolismo , Proteínas RGS/metabolismo , Ratos , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Transfecção/métodos
12.
Mol Biol Cell ; 17(4): 1652-63, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16452635

RESUMO

Alterations of mitochondrial function may play a central role in neuronal death in Huntington's disease (HD). However, the molecular mechanisms underlying such functional deficits of mitochondria are not elucidated yet. We herein showed that the expression of two important constituents of mitochondrial complex II, the 30-kDa iron-sulfur (Ip) subunit and the 70-kDa FAD (Fp) subunit, was preferentially decreased in the striatum of HD patients compared with controls. We also examined several mitochondrial proteins in striatal neurons that were infected with lentiviral vectors coding for the N-terminus part of huntingtin (Htt) with either a pathological (Htt171-82Q) or physiological (Htt171-19Q) polyglutamine tract. Compared with Htt171-19Q, expression of Htt171-82Q preferentially decreased the levels of Ip and Fp subunits and affected the dehydrogenase activity of the complex. The Htt171-82Q-induced preferential loss of complex II was not associated with a decrease in mRNA levels, suggesting the involvement of a posttranscriptional mechanism. Importantly, the overexpression of either Ip or Fp subunit restored complex II levels and blocked mitochondrial dysfunction and striatal cell death induced by Htt171-82Q in striatal neurons. The present results strongly suggest that complex II defects in HD may be instrumental in striatal cell death.


Assuntos
Apoptose , Corpo Estriado/enzimologia , Complexo II de Transporte de Elétrons/metabolismo , Doença de Huntington/enzimologia , Doença de Huntington/genética , Proteínas Ferro-Enxofre/metabolismo , Mitocôndrias/enzimologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Subunidades Proteicas/metabolismo , Succinato Desidrogenase/metabolismo , Corpo Estriado/citologia , Regulação para Baixo , Complexo II de Transporte de Elétrons/genética , Vetores Genéticos/genética , Humanos , Proteína Huntingtina , Lentivirus/genética , Mitocôndrias/genética , Proteínas Mitocondriais/metabolismo , Mutagênese , Mutação , Neurônios/enzimologia , Peptídeos/genética , Subunidades Proteicas/genética , Transfecção
13.
Nat Commun ; 9(1): 4272, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30323233

RESUMO

Impaired hippocampal synaptic plasticity contributes to cognitive impairment in Huntington's disease (HD). However, the molecular basis of such synaptic plasticity defects is not fully understood. Combining live-cell nanoparticle tracking and super-resolution imaging, we show that AMPAR surface diffusion, a key player in synaptic plasticity, is disturbed in various rodent models of HD. We demonstrate that defects in the brain-derived neurotrophic factor (BDNF)-tyrosine receptor kinase B (TrkB) signaling pathway contribute to the deregulated AMPAR trafficking by reducing the interaction between transmembrane AMPA receptor regulatory proteins (TARPs) and the PDZ-domain scaffold protein PSD95. The disturbed AMPAR surface diffusion is rescued by the antidepressant drug tianeptine via the BDNF signaling pathway. Tianeptine also restores the impaired LTP and hippocampus-dependent memory in different HD mouse models. These findings unravel a mechanism underlying hippocampal synaptic and memory dysfunction in HD, and highlight AMPAR surface diffusion as a promising therapeutic target.


Assuntos
Hipocampo/fisiopatologia , Doença de Huntington/fisiopatologia , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Receptores de AMPA/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Difusão , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Potenciação de Longa Duração/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurogênese/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Receptor trkB/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Tiazepinas/farmacologia
15.
F1000Res ; 6: 724, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28663786

RESUMO

Nucleoside triphosphate (NTP)s, like ATP (adenosine 5'-triphosphate) and GTP (guanosine 5'-triphosphate), have long been considered sufficiently concentrated and diffusible to fuel all cellular ATPases (adenosine triphosphatases) and GTPases (guanosine triphosphatases) in an energetically healthy cell without becoming limiting for function. However, increasing evidence for the importance of local ATP and GTP pools, synthesised in close proximity to ATP- or GTP-consuming reactions, has fundamentally challenged our view of energy metabolism. It has become evident that cellular energy metabolism occurs in many specialised 'microcompartments', where energy in the form of NTPs is transferred preferentially from NTP-generating modules directly to NTP-consuming modules. Such energy channeling occurs when diffusion through the cytosol is limited, where these modules are physically close and, in particular, if the NTP-consuming reaction has a very high turnover, i.e. is very processive. Here, we summarise the evidence for these conclusions and describe new insights into the physiological importance and molecular mechanisms of energy channeling gained from recent studies. In particular, we describe the role of glycolytic enzymes for axonal vesicle transport and nucleoside diphosphate kinases for the functions of dynamins and dynamin-related GTPases.

16.
Nat Commun ; 7: 13233, 2016 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-27775035

RESUMO

The glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) facilitates fast axonal transport in neurons. However, given that GAPDH does not produce ATP, it is unclear whether glycolysis per se is sufficient to propel vesicles. Although many proteins regulating transport have been identified, the molecular composition of transported vesicles in neurons has yet to be fully elucidated. Here we selectively enrich motile vesicles and perform quantitative proteomic analysis. In addition to the expected molecular motors and vesicular proteins, we find an enrichment of all the glycolytic enzymes. Using biochemical approaches and super-resolution microscopy, we observe that most glycolytic enzymes are selectively associated with vesicles and facilitate transport of vesicles in neurons. Finally, we provide evidence that mouse brain vesicles produce ATP from ADP and glucose, and display movement in a reconstituted in vitro transport assay of native vesicles. We conclude that transport of vesicles along microtubules can be autonomous.


Assuntos
Encéfalo/metabolismo , Metabolismo Energético , Glicólise , Neurônios/metabolismo , Vesículas Transportadoras/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Camundongos , Camundongos Transgênicos , Microtúbulos/metabolismo , Neurônios/citologia , Proteoma/metabolismo , Proteômica/métodos , Ratos
17.
J Neurosci ; 22(9): 3473-83, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11978824

RESUMO

A new strategy based on lentiviral-mediated delivery of mutant huntingtin (htt) was used to create a genetic model of Huntington's disease (HD) in rats and to assess the relative contribution of polyglutamine (CAG) repeat size, htt expression levels, and protein length on the onset and specificity of the pathology. Lentiviral vectors coding for the first 171, 853, and 1520 amino acids of wild-type (19 CAG) or mutant htt (44, 66, and 82 CAG) driven by either the phosphoglycerate kinase 1 (PGK) or the cytomegalovirus (CMV) promoters were injected in rat striatum. A progressive pathology characterized by sequential appearance of ubiquitinated htt aggregates, loss of dopamine- and cAMP-regulated phosphoprotein of 32 kDa staining, and cell death was observed over 6 months with mutant htt. Earlier onset and more severe pathology occurred with shorter fragments, longer CAG repeats, and higher expression levels. Interestingly, the aggregates were predominantly located in the nucleus of PGK-htt171-injected rats, whereas they were present in both the nucleus and processes of CMV-htt171-injected animals expressing lower transgene levels. Finally, a selective sparing of interneurons was observed in animals injected with vectors expressing mutant htt. These data demonstrate that lentiviral-mediated expression of mutant htt provides a robust in vivo genetic model for selective neural degeneration that will facilitate future studies on the pathogenesis of cell death and experimental therapeutics for HD.


Assuntos
Corpo Estriado/patologia , Doença de Huntington/patologia , Lentivirus , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Peptídeos/genética , Animais , Western Blotting , Linhagem Celular , Núcleo Celular/patologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Fosfoproteína 32 Regulada por cAMP e Dopamina , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Corpos de Inclusão/patologia , Lentivirus/genética , Microinjeções , Mutação , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/toxicidade , Proteínas Nucleares/biossíntese , Proteínas Nucleares/toxicidade , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Distribuição Tecidual , Expansão das Repetições de Trinucleotídeos
18.
ACS Nano ; 9(11): 11479-89, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26505527

RESUMO

Long-term inspection of biological phenomena requires probes of elevated intra- and extracellular stability and target biospecificity. The high fluorescence and photostability of quantum dot (QD) nanoparticles contributed to foster their promise as bioimaging tools that could overcome limitations associated with traditional fluorophores. However, QDs' potential as a bioimaging platform relies upon a precise control over the surface chemistry modifications of these nano-objects. Here, a zwitterion-vinylimidazole block copolymer ligand was synthesized, which regroups all anchoring groups in one compact terminal block, while the rest of the chain is endowed with antifouling and bioconjugation moieties. By further application of an oriented bioconjugation approach with whole IgG antibodies, QD nanobioconjugates were obtained that display outstanding intra- and extracellular stability as well as biorecognition capacity. Imaging the internalization and intracellular dynamics of a transmembrane cell receptor, the CB1 brain cannabinoid receptor, both in HEK293 cells and in neurons, illustrates the breadth of potential applications of these nanoprobes.


Assuntos
Betaína/análogos & derivados , Diagnóstico por Imagem/métodos , Imidazóis/química , Polímeros/química , Pontos Quânticos/química , Betaína/síntese química , Betaína/química , Coloides , Fluoresceína/química , Células HEK293 , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Imidazóis/síntese química , Cinética , Polimerização , Polímeros/síntese química , Receptor CB1 de Canabinoide/metabolismo , Propriedades de Superfície
19.
Methods Mol Biol ; 277: 199-213, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15201458

RESUMO

This chapter describes the potential use of viral-mediated gene transfer in the central nervous system as a new strategy in developing animal models of neurodegenerative diseases. To illustrate the approach, procedures for the production of lentiviral vectors encoding polyQ proteins are provided, as well as methods for the determination of viral titers, in vitro infection, and basic protocols for in vivo studies in rodents.


Assuntos
Técnicas de Transferência de Genes , Doenças Genéticas Inatas/genética , Lentivirus/genética , Repetições de Trinucleotídeos/genética , Animais , Linhagem Celular , Feminino , Humanos , Imuno-Histoquímica , Gravidez , Ratos , Ratos Sprague-Dawley
20.
PLoS One ; 9(6): e99341, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24926995

RESUMO

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder resulting from polyglutamine expansion in the huntingtin (HTT) protein and for which there is no cure. Although suppression of both wild type and mutant HTT expression by RNA interference is a promising therapeutic strategy, a selective silencing of mutant HTT represents the safest approach preserving WT HTT expression and functions. We developed small hairpin RNAs (shRNAs) targeting single nucleotide polymorphisms (SNP) present in the HTT gene to selectively target the disease HTT isoform. Most of these shRNAs silenced, efficiently and selectively, mutant HTT in vitro. Lentiviral-mediated infection with the shRNAs led to selective degradation of mutant HTT mRNA and prevented the apparition of neuropathology in HD rat's striatum expressing mutant HTT containing the various SNPs. In transgenic BACHD mice, the mutant HTT allele was also silenced by this approach, further demonstrating the potential for allele-specific silencing. Finally, the allele-specific silencing of mutant HTT in human embryonic stem cells was accompanied by functional recovery of the vesicular transport of BDNF along microtubules. These findings provide evidence of the therapeutic potential of allele-specific RNA interference for HD.


Assuntos
Encéfalo/citologia , Terapia Genética/métodos , Doença de Huntington/terapia , Proteínas Mutantes/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , RNA Interferente Pequeno/genética , Animais , Encéfalo/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células HEK293 , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Técnicas In Vitro , Masculino , Camundongos , Proteínas Mutantes/genética , Polimorfismo de Nucleotídeo Único , Isoformas de RNA/metabolismo , Estabilidade de RNA , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA