Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4060, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38744819

RESUMEN

Endocytosis requires a coordinated framework of molecular interactions that ultimately lead to the fission of nascent endocytic structures. How cytosolic proteins such as dynamin concentrate at discrete sites that are sparsely distributed across the plasma membrane remains poorly understood. Two dynamin-1 major splice variants differ by the length of their C-terminal proline-rich region (short-tail and long-tail). Using sptPALM in PC12 cells, neurons and MEF cells, we demonstrate that short-tail dynamin-1 isoforms ab and bb display an activity-dependent recruitment to the membrane, promptly followed by their concentration into nanoclusters. These nanoclusters are sensitive to both Calcineurin and dynamin GTPase inhibitors, and are larger, denser, and more numerous than that of long-tail isoform aa. Spatiotemporal modelling confirms that dynamin-1 isoforms perform distinct search patterns and undergo dimensional reduction to generate endocytic nanoclusters, with short-tail isoforms more robustly exploiting lateral trapping in the generation of nanoclusters compared to the long-tail isoform.


Asunto(s)
Dinamina I , Endocitosis , Isoformas de Proteínas , Animales , Dinamina I/metabolismo , Dinamina I/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/genética , Células PC12 , Ratas , Neuronas/metabolismo , Ratones , Membrana Celular/metabolismo , Calcineurina/metabolismo
2.
Nat Commun ; 15(1): 2217, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38472171

RESUMEN

Neurotransmitter release relies on the regulated fusion of synaptic vesicles (SVs) that are tightly packed within the presynaptic bouton of neurons. The mechanism by which SVs are clustered at the presynapse, while preserving their ability to dynamically recycle to support neuronal communication, remains unknown. Synapsin 2a (Syn2a) tetramerization has been suggested as a potential clustering mechanism. Here, we used Dual-pulse sub-diffractional Tracking of Internalised Molecules (DsdTIM) to simultaneously track single SVs from the recycling and the reserve pools, in live hippocampal neurons. The reserve pool displays a lower presynaptic mobility compared to the recycling pool and is also present in the axons. Triple knockout of Synapsin 1-3 genes (SynTKO) increased the mobility of reserve pool SVs. Re-expression of wild-type Syn2a (Syn2aWT), but not the tetramerization-deficient mutant K337Q (Syn2aK337Q), fully rescued these effects. Single-particle tracking revealed that Syn2aK337QmEos3.1 exhibited altered activity-dependent presynaptic translocation and nanoclustering. Therefore, Syn2a tetramerization controls its own presynaptic nanoclustering and thereby contributes to the dynamic immobilisation of the SV reserve pool.


Asunto(s)
Sinapsinas , Vesículas Sinápticas , Vesículas Sinápticas/fisiología , Sinapsinas/genética , Sinapsis , Transmisión Sináptica/fisiología , Neuronas/fisiología , Terminales Presinápticos
3.
EMBO J ; 43(4): 533-567, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38316990

RESUMEN

The phospholipid and free fatty acid (FFA) composition of neuronal membranes plays a crucial role in learning and memory, but the mechanisms through which neuronal activity affects the brain's lipid landscape remain largely unexplored. The levels of saturated FFAs, particularly of myristic acid (C14:0), strongly increase during neuronal stimulation and memory acquisition, suggesting the involvement of phospholipase A1 (PLA1) activity in synaptic plasticity. Here, we show that genetic ablation of the PLA1 isoform DDHD2 in mice dramatically reduces saturated FFA responses to memory acquisition across the brain. Furthermore, DDHD2 loss also decreases memory performance in reward-based learning and spatial memory models prior to the development of neuromuscular deficits that mirror human spastic paraplegia. Via pulldown-mass spectrometry analyses, we find that DDHD2 binds to the key synaptic protein STXBP1. Using STXBP1/2 knockout neurosecretory cells and a haploinsufficient STXBP1+/- mouse model of human early infantile encephalopathy associated with intellectual disability and motor dysfunction, we show that STXBP1 controls targeting of DDHD2 to the plasma membrane and generation of saturated FFAs in the brain. These findings suggest key roles for DDHD2 and STXBP1 in lipid metabolism and in the processes of synaptic plasticity, learning, and memory.


Asunto(s)
Ácidos Grasos no Esterificados , Memoria a Largo Plazo , Proteínas Munc18 , Fosfolipasas , Animales , Ratones , Encéfalo/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Memoria/fisiología , Proteínas Munc18/genética , Fosfolipasas/genética
4.
Nat Commun ; 14(1): 7277, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37949856

RESUMEN

Neuronal communication relies on the release of neurotransmitters from various populations of synaptic vesicles. Despite displaying vastly different release probabilities and mobilities, the reserve and recycling pool of vesicles co-exist within a single cluster suggesting that small synaptic biomolecular condensates could regulate their nanoscale distribution. Here, we performed a large-scale activity-dependent phosphoproteome analysis of hippocampal neurons in vitro and identified Tau as a highly phosphorylated and disordered candidate protein. Single-molecule super-resolution microscopy revealed that Tau undergoes liquid-liquid phase separation to generate presynaptic nanoclusters whose density and number are regulated by activity. This activity-dependent diffusion process allows Tau to translocate into the presynapse where it forms biomolecular condensates, to selectively control the mobility of recycling vesicles. Tau, therefore, forms presynaptic nano-biomolecular condensates that regulate the nanoscale organization of synaptic vesicles in an activity-dependent manner.


Asunto(s)
Condensados Biomoleculares , Vesículas Sinápticas , Vesículas Sinápticas/metabolismo , Terminales Presinápticos/metabolismo , Sinapsis/fisiología , Neuronas/metabolismo
5.
bioRxiv ; 2023 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-37503261

RESUMEN

Emerging variants of concern of SARS-CoV-2 can significantly reduce the prophylactic and therapeutic efficacy of vaccines and neutralizing antibodies due to mutations in the viral genome. Targeting cell host factors required for infection provides a complementary strategy to overcome this problem since the host genome is less susceptible to variation during the life span of infection. The enzymatic activities of the endosomal PIKfyve phosphoinositide kinase and the serine protease TMPRSS2 are essential to meditate infection in two complementary viral entry pathways. Simultaneous inhibition in cultured cells of their enzymatic activities with the small molecule inhibitors apilimod dimesylate and nafamostat mesylate synergistically prevent viral entry and infection of native SARS-CoV-2 and vesicular stomatitis virus (VSV)-SARS-CoV-2 chimeras expressing the SARS-CoV-2 surface spike (S) protein and of variants of concern. We now report prophylactic prevention of lung infection in mice intranasally infected with SARS-CoV-2 beta by combined intranasal delivery of very low doses of apilimod dimesylate and nafamostat mesylate, in a formulation that is stable for over 3 months at room temperature. Administration of these drugs up to 6 hours post infection did not inhibit infection of the lungs but substantially reduced death of infected airway epithelial cells. The efficiency and simplicity of formulation of the drug combination suggests its suitability as prophylactic or therapeutic treatment against SARS-CoV-2 infection in households, point of care facilities, and under conditions where refrigeration would not be readily available.

7.
Nat Commun ; 14(1): 3353, 2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37291117

RESUMEN

Single-molecule localization microscopy techniques are emerging as vital tools to unravel the nanoscale world of living cells by understanding the spatiotemporal organization of protein clusters at the nanometer scale. Current analyses define spatial nanoclusters based on detections but neglect important temporal information such as cluster lifetime and recurrence in "hotspots" on the plasma membrane. Spatial indexing is widely used in video games to detect interactions between moving geometric objects. Here, we use the R-tree spatial indexing algorithm to determine the overlap of the bounding boxes of individual molecular trajectories to establish membership in nanoclusters. Extending the spatial indexing into the time dimension allows the resolution of spatial nanoclusters into multiple spatiotemporal clusters. Using spatiotemporal indexing, we found that syntaxin1a and Munc18-1 molecules transiently cluster in hotspots, offering insights into the dynamics of neuroexocytosis. Nanoscale spatiotemporal indexing clustering (NASTIC) has been implemented as a free and open-source Python graphic user interface.


Asunto(s)
Algoritmos , Proteínas , Membrana Celular/metabolismo , Proteínas/metabolismo , Análisis Espacio-Temporal
8.
EMBO J ; 42(13): e112095, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37226896

RESUMEN

The unique nerve terminal targeting of botulinum neurotoxin type A (BoNT/A) is due to its capacity to bind two receptors on the neuronal plasma membrane: polysialoganglioside (PSG) and synaptic vesicle glycoprotein 2 (SV2). Whether and how PSGs and SV2 may coordinate other proteins for BoNT/A recruitment and internalization remains unknown. Here, we demonstrate that the targeted endocytosis of BoNT/A into synaptic vesicles (SVs) requires a tripartite surface nanocluster. Live-cell super-resolution imaging and electron microscopy of catalytically inactivated BoNT/A wildtype and receptor-binding-deficient mutants in cultured hippocampal neurons demonstrated that BoNT/A must bind coincidentally to a PSG and SV2 to target synaptic vesicles. We reveal that BoNT/A simultaneously interacts with a preassembled PSG-synaptotagmin-1 (Syt1) complex and SV2 on the neuronal plasma membrane, facilitating Syt1-SV2 nanoclustering that controls endocytic sorting of the toxin into synaptic vesicles. Syt1 CRISPRi knockdown suppressed BoNT/A- and BoNT/E-induced neurointoxication as quantified by SNAP-25 cleavage, suggesting that this tripartite nanocluster may be a unifying entry point for selected botulinum neurotoxins that hijack this for synaptic vesicle targeting.


Asunto(s)
Toxinas Botulínicas Tipo A , Toxinas Botulínicas Tipo A/metabolismo , Membrana Celular/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Vesículas Sinápticas/metabolismo , Animales , Ratas
9.
J Virol ; 97(4): e0014423, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37039676

RESUMEN

2019 coronavirus disease (COVID-19) is a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition to respiratory illness, COVID-19 patients exhibit neurological symptoms lasting from weeks to months (long COVID). It is unclear whether these neurological manifestations are due to an infection of brain cells. We found that a small fraction of human induced pluripotent stem cell (iPSC)-derived neurons, but not astrocytes, were naturally susceptible to SARS-CoV-2. Based on the inhibitory effect of blocking antibodies, the infection seemed to depend on the receptor angiotensin-converting enzyme 2 (ACE2), despite very low levels of its expression in neurons. The presence of double-stranded RNA in the cytoplasm (the hallmark of viral replication), abundant synthesis of viral late genes localized throughout infected cells, and an increase in the level of viral RNA in the culture medium (viral release) within the first 48 h of infection suggested that the infection was productive. Productive entry of SARS-CoV-2 requires the fusion of the viral and cellular membranes, which results in the delivery of the viral genome into the cytoplasm of the target cell. The fusion is triggered by proteolytic cleavage of the viral surface spike protein, which can occur at the plasma membrane or from endosomes or lysosomes. We found that SARS-CoV-2 infection of human neurons was insensitive to nafamostat and camostat, which inhibit cellular serine proteases, including transmembrane serine protease 2 (TMPRSS2). Inhibition of cathepsin L also did not significantly block infection. In contrast, the neuronal infection was blocked by apilimod, an inhibitor of phosphatidyl-inositol 5 kinase (PIK5K), which regulates early to late endosome maturation. IMPORTANCE COVID-19 is a disease caused by the coronavirus SARS-CoV-2. Millions of patients display neurological symptoms, including headache, impairment of memory, seizures, and encephalopathy, as well as anatomical abnormalities, such as changes in brain morphology. SARS-CoV-2 infection of the human brain has been documented, but it is unclear whether the observed neurological symptoms are linked to direct brain infection. The mechanism of virus entry into neurons has also not been characterized. Here, we investigated SARS-CoV-2 infection by using a human iPSC-derived neural cell model and found that a small fraction of cortical-like neurons was naturally susceptible to infection. The productive infection was ACE2 dependent and TMPRSS2 independent. We also found that the virus used the late endosomal and lysosomal pathway for cell entry and that the infection could be blocked by apilimod, an inhibitor of cellular PIK5K.


Asunto(s)
COVID-19 , Células Madre Pluripotentes Inducidas , SARS-CoV-2 , Humanos , Enzima Convertidora de Angiotensina 2 , COVID-19/fisiopatología , Endosomas/metabolismo , Endosomas/virología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Neuronas/virología , Síndrome Post Agudo de COVID-19/fisiopatología , Síndrome Post Agudo de COVID-19/virología , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus/efectos de los fármacos , Fosfotransferasas/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Astrocitos/virología , Células Cultivadas
10.
PLoS Biol ; 20(8): e3001728, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35913989

RESUMEN

Children typically experience more mild symptoms of Coronavirus Disease 2019 (COVID-19) when compared to adults. There is a strong body of evidence that children are also less susceptible to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection with the ancestral viral isolate. However, the emergence of SARS-CoV-2 variants of concern (VOCs) has been associated with an increased number of pediatric infections. Whether this is the result of widespread adult vaccination or fundamental changes in the biology of SARS-CoV-2 remain to be determined. Here, we use primary nasal epithelial cells (NECs) from children and adults, differentiated at an air-liquid interface to show that the ancestral SARS-CoV-2 replicates to significantly lower titers in the NECs of children compared to those of adults. This was associated with a heightened antiviral response to SARS-CoV-2 in the NECs of children. Importantly, the Delta variant also replicated to significantly lower titers in the NECs of children. This trend was markedly less pronounced in the case of Omicron. It is also striking to note that, at least in terms of viral RNA, Omicron replicated better in pediatric NECs compared to both Delta and the ancestral virus. Taken together, these data show that the nasal epithelium of children supports lower infection and replication of ancestral SARS-CoV-2, although this may be changing as the virus evolves.


Asunto(s)
COVID-19 , SARS-CoV-2 , Adulto , Niño , Células Epiteliales , Humanos , SARS-CoV-2/genética
11.
Methods Mol Biol ; 2233: 265-286, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33222141

RESUMEN

The fusion of synaptic vesicles with the plasma membrane underpins neurotransmission. A number of presynaptic proteins play a critical role in overcoming the energy barrier inherent to the fusion of the negatively charged vesicular and plasma membranes. Emerging concepts suggest that this process is hierarchical and dependent on rapid and transient reorganization of proteins in and out of small nanoclusters located in the active zones of nerve terminals. Examining the nanoscale organization of presynaptic molecules requires super-resolution microscopy to overcome the limits of conventional light microscopy. In this chapter, we describe three super-resolution techniques that allow for the examination of the nanoscale organization of proteins within live hippocampal nerve terminals. We used (1) single-particle tracking photoactivated localization microscopy (sptPALM) to resolve the mobility and clustering of syntaxin1A (STX1A), (2) universal Point Accumulation Imaging in Nanoscale Topography (uPAINT) to study the mobility of a pool of vesicular-associated membrane protein 2 (VAMP2) transiting on the plasma membrane, and (3) subdiffractional Tracking of Internalized Molecules (sdTIM) to track VAMP2-positive recycling synaptic vesicles in conjunction with Cholera Toxin subunit B (CTB), which has recently been shown to be trafficked retrogradely from the presynapse to the cell body via signaling endosomes.


Asunto(s)
Exocitosis/genética , Microscopía/métodos , Imagen Individual de Molécula/métodos , Vesículas Sinápticas/genética , Animales , Endocitosis/genética , Hipocampo/ultraestructura , Humanos , Ratones , Neuronas/ultraestructura , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Sinapsis/genética , Sinapsis/ultraestructura , Transmisión Sináptica/genética , Vesículas Sinápticas/ultraestructura
12.
Proc Natl Acad Sci U S A ; 117(48): 30476-30487, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33214152

RESUMEN

None of the current superresolution microscopy techniques can reliably image the changes in endogenous protein nanoclustering dynamics associated with specific conformations in live cells. Single-domain nanobodies have been invaluable tools to isolate defined conformational states of proteins, and we reasoned that expressing these nanobodies coupled to single-molecule imaging-amenable tags could allow superresolution analysis of endogenous proteins in discrete conformational states. Here, we used anti-GFP nanobodies tagged with photoconvertible mEos expressed as intrabodies, as a proof-of-concept to perform single-particle tracking on a range of GFP proteins expressed in live cells, neurons, and small organisms. We next expressed highly specialized nanobodies that target conformation-specific endogenous ß2-adrenoreceptor (ß2-AR) in neurosecretory cells, unveiling real-time mobility behaviors of activated and inactivated endogenous conformers during agonist treatment in living cells. We showed that activated ß2-AR (Nb80) is highly immobile and organized in nanoclusters. The Gαs-GPCR complex detected with Nb37 displayed higher mobility with surprisingly similar nanoclustering dynamics to that of Nb80. Activated conformers are highly sensitive to dynamin inhibition, suggesting selective targeting for endocytosis. Inactivated ß2-AR (Nb60) molecules are also largely immobile but relatively less sensitive to endocytic blockade. Expression of single-domain nanobodies therefore provides a unique opportunity to capture highly transient changes in the dynamic nanoscale organization of endogenous proteins.


Asunto(s)
Modelos Moleculares , Conformación Proteica , Receptores Adrenérgicos beta 2/química , Imagen Individual de Molécula , Anticuerpos de Dominio Único/química , Animales , Línea Celular , Endocitosis , Técnica del Anticuerpo Fluorescente , Expresión Génica , Genes Reporteros , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Unión Proteica , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Proteínas Recombinantes de Fusión , Imagen Individual de Molécula/métodos , Anticuerpos de Dominio Único/metabolismo , Pez Cebra
13.
Science ; 370(6518): 856-860, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33082293

RESUMEN

The causative agent of coronavirus disease 2019 (COVID-19) is the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). For many viruses, tissue tropism is determined by the availability of virus receptors and entry cofactors on the surface of host cells. In this study, we found that neuropilin-1 (NRP1), known to bind furin-cleaved substrates, significantly potentiates SARS-CoV-2 infectivity, an effect blocked by a monoclonal blocking antibody against NRP1. A SARS-CoV-2 mutant with an altered furin cleavage site did not depend on NRP1 for infectivity. Pathological analysis of olfactory epithelium obtained from human COVID-19 autopsies revealed that SARS-CoV-2 infected NRP1-positive cells facing the nasal cavity. Our data provide insight into SARS-CoV-2 cell infectivity and define a potential target for antiviral intervention.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/virología , Neuropilina-1/metabolismo , Neumonía Viral/virología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus , Enzima Convertidora de Angiotensina 2 , Animales , Anticuerpos Monoclonales/inmunología , Betacoronavirus/genética , COVID-19 , Células CACO-2 , Femenino , Células HEK293 , Interacciones Microbiota-Huesped , Humanos , Pulmón/metabolismo , Masculino , Nanopartículas del Metal , Ratones , Ratones Endogámicos C57BL , Mutación , Neuropilina-1/química , Neuropilina-1/genética , Neuropilina-1/inmunología , Neuropilina-2/metabolismo , Mucosa Olfatoria/metabolismo , Mucosa Olfatoria/virología , Pandemias , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Unión Proteica , Dominios Proteicos , Mucosa Respiratoria/metabolismo , SARS-CoV-2 , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Glicoproteína de la Espiga del Coronavirus/química
14.
J Cell Biol ; 219(5)2020 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-32182623

RESUMEN

Most mammalian neurons have a narrow axon, which constrains the passage of large cargoes such as autophagosomes that can be larger than the axon diameter. Radial axonal expansion must therefore occur to ensure efficient axonal trafficking. In this study, we reveal that the speed of various large cargoes undergoing axonal transport is significantly slower than that of small ones and that the transit of diverse-sized cargoes causes an acute, albeit transient, axonal radial expansion, which is immediately restored by constitutive axonal contractility. Using live super-resolution microscopy, we demonstrate that actomyosin-II controls axonal radial contractility and local expansion, and that NM-II filaments associate with periodic F-actin rings via their head domains. Pharmacological inhibition of NM-II activity significantly increases axon diameter by detaching the NM-II from F-actin and impacts the trafficking speed, directionality, and overall efficiency of long-range retrograde trafficking. Consequently, prolonged NM-II inactivation leads to disruption of periodic actin rings and formation of focal axonal swellings, a hallmark of axonal degeneration.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Actomiosina/genética , Autofagosomas/ultraestructura , Axones/ultraestructura , Neuronas/metabolismo , Citoesqueleto de Actina/genética , Actinas/genética , Actinas/metabolismo , Actomiosina/metabolismo , Animales , Autofagosomas/genética , Transporte Axonal/genética , Axones/metabolismo , Movimiento Celular/genética , Proteínas Contráctiles/genética , Conos de Crecimiento/metabolismo , Microtúbulos/genética , Microtúbulos/ultraestructura , Contracción Muscular/genética , Neuronas/ultraestructura , Transporte de Proteínas/genética , Ratas
15.
J Neurochem ; 153(3): 300-333, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31745996

RESUMEN

Despite the human brain being made of nearly 60% fat, the vast majority of studies on the mechanisms of neuronal communication which underpin cognition, memory and learning, primarily focus on proteins and/or (epi)genetic mechanisms. Phospholipids are the main component of all cellular membranes and function as substrates for numerous phospholipid-modifying enzymes, including phospholipases, which release free fatty acids (FFAs) and other lipid metabolites that can alter the intrinsic properties of the membranes, recruit and activate critical proteins, and act as lipid signalling molecules. Here, we will review brain specific phospholipases, their roles in membrane remodelling, neuronal function, learning and memory, as well as their disease implications. In particular, we will highlight key roles of unsaturated FFAs, particularly arachidonic acid, in neurotransmitter release, neuroinflammation and memory. In light of recent findings, we will also discuss the emerging role of phospholipase A1 and the creation of saturated FFAs in the brain.


Asunto(s)
Memoria/fisiología , Neuronas/enzimología , Fosfolipasas/fisiología , Animales , Encéfalo/enzimología , Humanos , Aprendizaje/fisiología , Fosfolípidos/fisiología
16.
Nat Commun ; 10(1): 127, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30631079

RESUMEN

COPI is a key mediator of protein trafficking within the secretory pathway. COPI is recruited to the membrane primarily through binding to Arf GTPases, upon which it undergoes assembly to form coated transport intermediates responsible for trafficking numerous proteins, including Golgi-resident enzymes. Here, we identify GORAB, the protein mutated in the skin and bone disorder gerodermia osteodysplastica, as a component of the COPI machinery. GORAB forms stable domains at the trans-Golgi that, via interactions with the COPI-binding protein Scyl1, promote COPI recruitment to these domains. Pathogenic GORAB mutations perturb Scyl1 binding or GORAB assembly into domains, indicating the importance of these interactions. Loss of GORAB causes impairment of COPI-mediated retrieval of trans-Golgi enzymes, resulting in a deficit in glycosylation of secretory cargo proteins. Our results therefore identify GORAB as a COPI scaffolding factor, and support the view that defective protein glycosylation is a major disease mechanism in gerodermia osteodysplastica.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteína Coat de Complejo I/metabolismo , Enzimas/metabolismo , Aparato de Golgi/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Enfermedades Óseas/congénito , Enfermedades Óseas/genética , Enfermedades Óseas/metabolismo , Proteínas Portadoras/genética , Células Cultivadas , Proteína Coat de Complejo I/genética , Proteínas de Unión al ADN , Enanismo/genética , Enanismo/metabolismo , Glicosilación , Proteínas de la Matriz de Golgi , Células HEK293 , Células HeLa , Humanos , Mutación , Unión Proteica , Transporte de Proteínas , Interferencia de ARN , Enfermedades Cutáneas Genéticas/genética , Enfermedades Cutáneas Genéticas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Prog Neuropsychopharmacol Biol Psychiatry ; 84(Pt B): 362-381, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28870634

RESUMEN

Dendritic spines are small actin-rich protrusions from neuronal dendrites that form the postsynaptic part of most excitatory synapses. Changes in the shape and size of dendritic spines correlate with the functional changes in excitatory synapses and are heavily dependent on the remodeling of the underlying actin cytoskeleton. Recent evidence implicates synapses at dendritic spines as important substrates of pathogenesis in neuropsychiatric disorders, including autism spectrum disorder (ASD). Although synaptic perturbations are not the only alterations relevant for these diseases, understanding the molecular underpinnings of the spine and synapse pathology may provide insight into their etiologies and could reveal new drug targets. In this review, we will discuss recent findings of defective actin regulation in dendritic spines associated with ASD.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Trastorno del Espectro Autista/patología , Espinas Dendríticas/metabolismo , Sinapsis/metabolismo , Animales , Trastorno del Espectro Autista/metabolismo , Humanos
18.
Nat Protoc ; 12(12): 2590-2622, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29189775

RESUMEN

Our understanding of endocytic pathway dynamics is restricted by the diffraction limit of light microscopy. Although super-resolution techniques can overcome this issue, highly crowded cellular environments, such as nerve terminals, can also dramatically limit the tracking of multiple endocytic vesicles such as synaptic vesicles (SVs), which in turn restricts the analytical dissection of their discrete diffusional and transport states. We recently introduced a pulse-chase technique for subdiffractional tracking of internalized molecules (sdTIM) that allows the visualization of fluorescently tagged molecules trapped in individual signaling endosomes and SVs in presynapses or axons with 30- to 50-nm localization precision. We originally developed this approach for tracking single molecules of botulinum neurotoxin type A, which undergoes activity-dependent internalization and retrograde transport in autophagosomes. This method was then adapted to localize the signaling endosomes containing cholera toxin subunit-B that undergo retrograde transport in axons and to track SVs in the crowded environment of hippocampal presynapses. We describe (i) the construction of a custom-made microfluidic device that enables control over neuronal orientation; (ii) the 3D printing of a perfusion system for sdTIM experiments performed on glass-bottom dishes; (iii) the dissection, culturing and transfection of hippocampal neurons in microfluidic devices; and (iv) guidance on how to perform the pulse-chase experiments and data analysis. In addition, we describe the use of single-molecule-tracking analytical tools to reveal the average and the heterogeneous single-molecule mobility behaviors. We also discuss alternative reagents and equipment that can, in principle, be used for sdTIM experiments and describe how to adapt sdTIM to image nanocluster formation and/or tubulation in early endosomes during sorting events. The procedures described in this protocol take ∼1 week.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Endocitosis , Dispositivos Laboratorio en un Chip , Neuronas/citología , Imagen Óptica/métodos , Animales , Técnicas de Cultivo de Célula/instrumentación , Supervivencia Celular , Células Cultivadas , Diseño de Equipo , Hipocampo/citología , Imagen Óptica/instrumentación , Perfusión/instrumentación , Perfusión/métodos , Impresión Tridimensional , Ratas , Ratas Sprague-Dawley , Transfección/instrumentación , Transfección/métodos
19.
J Cell Biol ; 215(2): 277-292, 2016 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-27810917

RESUMEN

Our understanding of endocytic pathway dynamics is severely restricted by the diffraction limit of light microscopy. To address this, we implemented a novel technique based on the subdiffractional tracking of internalized molecules (sdTIM). This allowed us to image anti-green fluorescent protein Atto647N-tagged nanobodies trapped in synaptic vesicles (SVs) from live hippocampal nerve terminals expressing vesicle-associated membrane protein 2 (VAMP2)-pHluorin with 36-nm localization precision. Our results showed that, once internalized, VAMP2-pHluorin/Atto647N-tagged nanobodies exhibited a markedly lower mobility than on the plasma membrane, an effect that was reversed upon restimulation in presynapses but not in neighboring axons. Using Bayesian model selection applied to hidden Markov modeling, we found that SVs oscillated between diffusive states or a combination of diffusive and transport states with opposite directionality. Importantly, SVs exhibiting diffusive motion were relatively less likely to switch to the transport motion. These results highlight the potential of the sdTIM technique to provide new insights into the dynamics of endocytic pathways in a wide variety of cellular settings.


Asunto(s)
Endocitosis , Movimiento (Física) , Fenómenos Ópticos , Vesículas Sinápticas/metabolismo , Animales , Axones/metabolismo , Teorema de Bayes , Membrana Celular/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/citología , Imagenología Tridimensional , Cadenas de Markov , Neuronas/metabolismo , Ratas Sprague-Dawley , Anticuerpos de Dominio Único/metabolismo , Procesos Estocásticos , Sinapsis/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
20.
Sci Rep ; 6: 35969, 2016 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-27786289

RESUMEN

Reticulons (RTNs) are a large family of membrane associated proteins with various functions. NOGO-A/RTN4A has a well-known function in limiting neurite outgrowth and restricting the plasticity of the mammalian central nervous system. On the other hand, Reticulon 4 proteins were shown to be involved in forming and maintaining endoplasmic reticulum (ER) tubules. Using comparative transcriptome analysis and qPCR, we show here that NOGO-B/RTN4B and NOGO-A/RTN4A are simultaneously expressed in cultured epithelial, fibroblast and neuronal cells. Electron tomography combined with immunolabelling reveal that both isoforms localize preferably to curved membranes on ER tubules and sheet edges. Morphological analysis of cells with manipulated levels of NOGO-B/RTN4B revealed that it is required for maintenance of normal ER shape; over-expression changes the sheet/tubule balance strongly towards tubules and causes the deformation of the cell shape while depletion of the protein induces formation of large peripheral ER sheets.


Asunto(s)
Retículo Endoplásmico/metabolismo , Proteínas Nogo/genética , Animales , Línea Celular , Forma de la Célula , Células Cultivadas , Retículo Endoplásmico/ultraestructura , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Perfilación de la Expresión Génica , Humanos , Ratones , Microscopía Inmunoelectrónica , Células 3T3 NIH , Neuronas/metabolismo , Neuronas/ultraestructura , Proteínas Nogo/antagonistas & inhibidores , Proteínas Nogo/metabolismo , Isoformas de Proteínas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...