Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Cell ; 185(18): 3390-3407.e18, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36055200

RESUMEN

Chemical synapses between axons and dendrites mediate neuronal intercellular communication. Here, we describe a synapse between axons and primary cilia: the axo-ciliary synapse. Using enhanced focused ion beam-scanning electron microscopy on samples with optimally preserved ultrastructure, we discovered synapses between brainstem serotonergic axons and the primary cilia of hippocampal CA1 pyramidal neurons. Functionally, these cilia are enriched in a ciliary-restricted serotonin receptor, the 5-hydroxytryptamine receptor 6 (5-HTR6). Using a cilia-targeted serotonin sensor, we show that opto- and chemogenetic stimulation of serotonergic axons releases serotonin onto cilia. Ciliary 5-HTR6 stimulation activates a non-canonical Gαq/11-RhoA pathway, which modulates nuclear actin and increases histone acetylation and chromatin accessibility. Ablation of this pathway reduces chromatin accessibility in CA1 pyramidal neurons. As a signaling apparatus with proximity to the nucleus, axo-ciliary synapses short circuit neurotransmission to alter the postsynaptic neuron's epigenetic state.


Asunto(s)
Axones/fisiología , Cromatina/química , Cilios , Sinapsis , Núcleo Celular/metabolismo , Cromatina/metabolismo , Cilios/metabolismo , Hipocampo/citología , Hipocampo/fisiología , Serotonina/metabolismo , Transducción de Señal , Sinapsis/fisiología
2.
Cell ; 150(3): 495-507, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863004

RESUMEN

Coated pits assemble by growth of a clathrin lattice, which is linked by adaptors to the underlying membrane. How does this process start? We used live-cell TIRF imaging with single-molecule EGFP sensitivity and high temporal resolution to detect arrival of the clathrin triskelions and AP2 adaptors that initiate coat assembly. Unbiased object identification and trajectory tracking, together with a statistical model, yield the arrival times and numbers of individual proteins, as well as experimentally confirmed estimates of the extent of substitution of endogenous by expressed, fluorescently tagged proteins. Pits initiate by coordinated arrival of clathrin and AP2, which is usually detected as two sequential steps, each of one triskelion with two adaptors. PI-4,5-P2 is essential for initiation. The accessory proteins FCHo1/2 are not; instead, they are required for sustained growth. This objective picture of coated pit initiation also shows that methods outlined here will be broadly useful for studies of dynamic assemblies in living cells.


Asunto(s)
Clatrina/metabolismo , Invaginaciones Cubiertas de la Membrana Celular/metabolismo , Proteínas de la Membrana/metabolismo , Complejo 2 de Proteína Adaptadora/metabolismo , Animales , Línea Celular , Membrana Celular/metabolismo , Chlorocebus aethiops , Proteínas/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(38): e2209514119, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36048924

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cell entry starts with membrane attachment and ends with spike (S) protein-catalyzed membrane fusion depending on two cleavage steps, namely, one usually by furin in producing cells and the second by TMPRSS2 on target cells. Endosomal cathepsins can carry out both. Using real-time three-dimensional single-virion tracking, we show that fusion and genome penetration require virion exposure to an acidic milieu of pH 6.2 to 6.8, even when furin and TMPRSS2 cleavages have occurred. We detect the sequential steps of S1-fragment dissociation, fusion, and content release from the cell surface in TMPRRS2-overexpressing cells only when exposed to acidic pH. We define a key role of an acidic environment for successful infection, found in endosomal compartments and at the surface of TMPRSS2-expressing cells in the acidic milieu of the nasal cavity.


Asunto(s)
COVID-19 , Cavidad Nasal , SARS-CoV-2 , Serina Endopeptidasas , Internalización del Virus , COVID-19/virología , Furina/genética , Furina/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Cavidad Nasal/química , Cavidad Nasal/virología , SARS-CoV-2/fisiología , Serina Endopeptidasas/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(40): e2210990119, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36122200

RESUMEN

Variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge currently available coronavirus disease 2019 vaccines and monoclonal antibody therapies through epitope change on the receptor binding domain of the viral spike glycoprotein. Hence, there is a specific urgent need for alternative antivirals that target processes less likely to be affected by mutation, such as the membrane fusion step of viral entry into the host cell. One such antiviral class includes peptide inhibitors, which block formation of the so-called heptad repeat 1 and 2 (HR1HR2) six-helix bundle of the SARS-CoV-2 spike (S) protein and thus interfere with viral membrane fusion. We performed structural studies of the HR1HR2 bundle, revealing an extended, well-folded N-terminal region of HR2 that interacts with the HR1 triple helix. Based on this structure, we designed an extended HR2 peptide that achieves single-digit nanomolar inhibition of SARS-CoV-2 in cell-based and virus-based assays without the need for modifications such as lipidation or chemical stapling. The peptide also strongly inhibits all major SARS-CoV-2 variants to date. This extended peptide is ∼100-fold more potent than all previously published short, unmodified HR2 peptides, and it has a very long inhibition lifetime after washout in virus infection assays, suggesting that it targets a prehairpin intermediate of the SARS-CoV-2 S protein. Together, these results suggest that regions outside the HR2 helical region may offer new opportunities for potent peptide-derived therapeutics for SARS-CoV-2 and its variants, and even more distantly related viruses, and provide further support for the prehairpin intermediate of the S protein.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Glicoproteína de la Espiga del Coronavirus , Antivirales/química , Antivirales/farmacología , Humanos , Péptidos/química , Péptidos/farmacología , SARS-CoV-2/efectos de los fármacos
5.
Nature ; 552(7685): 410-414, 2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29236694

RESUMEN

Vesicular carriers transport proteins and lipids from one organelle to another, recognizing specific identifiers for the donor and acceptor membranes. Two important identifiers are phosphoinositides and GTP-bound GTPases, which provide well-defined but mutable labels. Phosphatidylinositol and its phosphorylated derivatives are present on the cytosolic faces of most cellular membranes. Reversible phosphorylation of its headgroup produces seven distinct phosphoinositides. In endocytic traffic, phosphatidylinositol-4,5-biphosphate marks the plasma membrane, and phosphatidylinositol-3-phosphate and phosphatidylinositol-4-phosphate mark distinct endosomal compartments. It is unknown what sequence of changes in lipid content confers on the vesicles their distinct identity at each intermediate step. Here we describe 'coincidence-detecting' sensors that selectively report the phosphoinositide composition of clathrin-associated structures, and the use of these sensors to follow the dynamics of phosphoinositide conversion during endocytosis. The membrane of an assembling coated pit, in equilibrium with the surrounding plasma membrane, contains phosphatidylinositol-4,5-biphosphate and a smaller amount of phosphatidylinositol-4-phosphate. Closure of the vesicle interrupts free exchange with the plasma membrane. A substantial burst of phosphatidylinositol-4-phosphate immediately after budding coincides with a burst of phosphatidylinositol-3-phosphate, distinct from any later encounter with the phosphatidylinositol-3-phosphate pool in early endosomes; phosphatidylinositol-3,4-biphosphate and the GTPase Rab5 then appear and remain as the uncoating vesicles mature into Rab5-positive endocytic intermediates. Our observations show that a cascade of molecular conversions, made possible by the separation of a vesicle from its parent membrane, can label membrane-traffic intermediates and determine their destinations.


Asunto(s)
Vesículas Cubiertas por Clatrina/química , Vesículas Cubiertas por Clatrina/metabolismo , Clatrina/metabolismo , Invaginaciones Cubiertas de la Membrana Celular/metabolismo , Endocitosis , Endosomas/metabolismo , Fosfatidilinositoles/metabolismo , Animales , Auxilinas/metabolismo , Células COS , Línea Celular , Membrana Celular/química , Membrana Celular/metabolismo , Chlorocebus aethiops , Invaginaciones Cubiertas de la Membrana Celular/química , Endosomas/química , Humanos , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilinositoles/análisis , Fosfatidilinositoles/química , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Fosfotransferasas/metabolismo , Proteínas de Unión al GTP rab5/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(34): 20803-20813, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32764148

RESUMEN

Virus entry is a multistep process. It initiates when the virus attaches to the host cell and ends when the viral contents reach the cytosol. Genetically unrelated viruses can subvert analogous subcellular mechanisms and use similar trafficking pathways for successful entry. Antiviral strategies targeting early steps of infection are therefore appealing, particularly when the probability for successful interference through a common step is highest. We describe here potent inhibitory effects on content release and infection by chimeric vesicular stomatitis virus (VSV) containing the envelope proteins of Zaire ebolavirus (VSV-ZEBOV) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (VSV-SARS-CoV-2) elicited by Apilimod and Vacuolin-1, small-molecule inhibitors of the main endosomal phosphatidylinositol-3-phosphate/phosphatidylinositol 5-kinase, PIKfyve. We also describe potent inhibition of SARS-CoV-2 strain 2019-nCoV/USA-WA1/2020 by Apilimod. These results define tools for studying the intracellular trafficking of pathogens elicited by inhibition of PIKfyve kinase and suggest the potential for targeting this kinase in developing small-molecule antivirals against SARS-CoV-2.


Asunto(s)
Betacoronavirus/efectos de los fármacos , Ebolavirus/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas , Triazinas/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Betacoronavirus/fisiología , COVID-19 , Células Cultivadas , Infecciones por Coronavirus , Ebolavirus/fisiología , Edición Génica , Humanos , Hidrazonas , Pandemias , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Neumonía Viral , Pirimidinas , SARS-CoV-2 , Proteínas del Envoltorio Viral/genética
7.
Proc Natl Acad Sci U S A ; 117(50): 32105-32113, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33239446

RESUMEN

Cholesterol 25-hydroxylase (CH25H) is an interferon (IFN)-stimulated gene that shows broad antiviral activities against a wide range of enveloped viruses. Here, using an IFN-stimulated gene screen against vesicular stomatitis virus (VSV)-SARS-CoV and VSV-SARS-CoV-2 chimeric viruses, we identified CH25H and its enzymatic product 25-hydroxycholesterol (25HC) as potent inhibitors of SARS-CoV-2 replication. Internalized 25HC accumulates in the late endosomes and potentially restricts SARS-CoV-2 spike protein catalyzed membrane fusion via blockade of cholesterol export. Our results highlight one of the possible antiviral mechanisms of 25HC and provide the molecular basis for its therapeutic development.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Endosomas/genética , Hidroxicolesteroles/farmacología , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Antivirales/farmacología , COVID-19/metabolismo , COVID-19/patología , COVID-19/virología , Endosomas/metabolismo , Humanos , Interferones/metabolismo , Fusión de Membrana/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/genética , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
8.
J Virol ; 95(21): e0097521, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34406858

RESUMEN

Repurposing FDA-approved inhibitors able to prevent infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) could provide a rapid path to establish new therapeutic options to mitigate the effects of coronavirus disease 2019 (COVID-19). Proteolytic cleavages of the spike (S) protein of SARS-CoV-2, mediated by the host cell proteases cathepsin and TMPRSS2, alone or in combination, are key early activation steps required for efficient infection. The PIKfyve kinase inhibitor apilimod interferes with late endosomal viral traffic and through an ill-defined mechanism prevents in vitro infection through late endosomes mediated by cathepsin. Similarly, inhibition of TMPRSS2 protease activity by camostat mesylate or nafamostat mesylate prevents infection mediated by the TMPRSS2-dependent and cathepsin-independent pathway. Here, we combined the use of apilimod with camostat mesylate or nafamostat mesylate and found an unexpected ∼5- to 10-fold increase in their effectiveness to prevent SARS-CoV-2 infection in different cell types. Comparable synergism was observed using both a chimeric vesicular stomatitis virus (VSV) containing S of SARS-CoV-2 (VSV-SARS-CoV-2) and SARS-CoV-2. The substantial ∼5-fold or higher decrease of the half-maximal effective concentrations (EC50s) suggests a plausible treatment strategy based on the combined use of these inhibitors. IMPORTANCE Infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causing the coronavirus disease 2019 (COVID-2019) global pandemic. There are ongoing efforts to uncover effective antiviral agents that could mitigate the severity of the disease by controlling the ensuing viral replication. Promising candidates include small molecules that inhibit the enzymatic activities of host proteins, thus preventing SARS-CoV-2 entry and infection. They include apilimod, an inhibitor of PIKfyve kinase, and camostat mesylate and nafamostat mesylate, inhibitors of TMPRSS2 protease. Our research is significant for having uncovered an unexpected synergism in the effective inhibitory activity of apilimod used together with camostat mesylate or nafamostat mesylate.


Asunto(s)
Antivirales/farmacología , Benzamidinas/farmacología , Ésteres/farmacología , Guanidinas/farmacología , Hidrazonas/farmacología , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Pirimidinas/farmacología , SARS-CoV-2/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Sinergismo Farmacológico , Humanos , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de Proteasas/farmacología , SARS-CoV-2/fisiología , Células Vero , Internalización del Virus , Tratamiento Farmacológico de COVID-19
9.
EMBO J ; 35(21): 2270-2284, 2016 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-27670760

RESUMEN

The large GTPase dynamin is the first protein shown to catalyze membrane fission. Dynamin and its related proteins are essential to many cell functions, from endocytosis to organelle division and fusion, and it plays a critical role in many physiological functions such as synaptic transmission and muscle contraction. Research of the past three decades has focused on understanding how dynamin works. In this review, we present the basis for an emerging consensus on how dynamin functions. Three properties of dynamin are strongly supported by experimental data: first, dynamin oligomerizes into a helical polymer; second, dynamin oligomer constricts in the presence of GTP; and third, dynamin catalyzes membrane fission upon GTP hydrolysis. We present the two current models for fission, essentially diverging in how GTP energy is spent. We further discuss how future research might solve the remaining open questions presently under discussion.


Asunto(s)
Membrana Celular/fisiología , Dinaminas/fisiología , Animales , Guanosina Trifosfato/fisiología , Humanos
10.
Traffic ; 17(1): 40-52, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26481905

RESUMEN

The sphingosine 1-phosphate receptor 1 (S1PR1) is one of five G protein-coupled receptors activated by the lipid sphingosine 1-phosphate (S1P). Stimulation of S1PR1 by binding S1P or the synthetic agonist FTY720P results in rapid desensitization, associated in part with depletion of receptor from the cell surface. We report here combining spinning disc confocal fluorescence microscopy and flow cytometry to show that rapid internalization of activated S1PR1 relies on a functional clathrin-mediated endocytic pathway. Uptake of activated S1PR1 was strongly inhibited in cells disrupted in their clathrin-mediated endocytosis by depleting clathrin or AP-2 or by treating cells with dynasore-OH. The uptake of activated S1P1R was strongly inhibited in cells lacking both ß-arrestin 1 and ß-arrestin 2, indicating that activated S1PR1 follows the canonical route of endocytosis for G-protein coupled receptor's (GPCR)'s.


Asunto(s)
Clatrina/metabolismo , Endocitosis , Receptores de Lisoesfingolípidos/metabolismo , Animales , Arrestinas/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Organofosfatos/farmacología , Receptores de Lisoesfingolípidos/agonistas , Esfingosina/análogos & derivados , Esfingosina/farmacología , Receptores de Esfingosina-1-Fosfato , beta-Arrestina 1 , Arrestina beta 2 , beta-Arrestinas
12.
J Virol ; 90(9): 4494-4510, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26912630

RESUMEN

UNLABELLED: Virus entry into cells is a multistep process that often requires the subversion of subcellular machineries. A more complete understanding of these steps is necessary to develop new antiviral strategies. While studying the potential role of the actin network and one of its master regulators, the small GTPase Cdc42, during Junin virus (JUNV) entry, we serendipitously uncovered the small molecule ZCL278, reported to inhibit Cdc42 function as an entry inhibitor for JUNV and for vesicular stomatitis virus, lymphocytic choriomeningitis virus, and dengue virus but not for the nonenveloped poliovirus. Although ZCL278 did not interfere with JUNV attachment to the cell surface or virus particle internalization into host cells, it prevented the release of JUNV ribonucleoprotein cores into the cytosol and decreased pH-mediated viral fusion with host membranes. We also identified SVG-A astroglial cell-derived cells to be highly permissive for JUNV infection and generated new cell lines expressing fluorescently tagged Rab5c or Rab7a or lacking Cdc42 using clustered regularly interspaced short palindromic repeat (CRISPR)-caspase 9 (Cas9) gene-editing strategies. Aided by these tools, we uncovered that perturbations in the actin cytoskeleton or Cdc42 activity minimally affect JUNV entry, suggesting that the inhibitory effect of ZCL278 is not mediated by ZCL278 interfering with the activity of Cdc42. Instead, ZCL278 appears to redistribute viral particles from endosomal to lysosomal compartments. ZCL278 also inhibited JUNV replication in a mouse model, and no toxicity was detected. Together, our data suggest the unexpected antiviral activity of ZCL278 and highlight its potential for use in the development of valuable new tools to study the intracellular trafficking of pathogens. IMPORTANCE: The Junin virus is responsible for outbreaks of Argentine hemorrhagic fever in South America, where 5 million people are at risk. Limited options are currently available to treat infections by Junin virus or other viruses of the Arenaviridae, making the identification of additional tools, including small-molecule inhibitors, of great importance. How Junin virus enters cells is not yet fully understood. Here we describe new cell culture models in which the cells are susceptible to Junin virus infection and to which we applied CRISPR-Cas9 genome engineering strategies to help characterize early steps during virus entry. We also uncovered ZCL278 to be a new antiviral small molecule that potently inhibits the cellular entry of the Junin virus and other enveloped viruses. Moreover, we show that ZCL278 also functions in vivo, thereby preventing Junin virus replication in a mouse model, opening the possibility for the discovery of ZCL278 derivatives of therapeutic potential.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Descubrimiento de Drogas , Tiourea/análogos & derivados , Internalización del Virus/efectos de los fármacos , Actinas/metabolismo , Animales , Línea Celular , Células Cultivadas , Clatrina/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/virología , Técnicas de Inactivación de Genes , Fiebre Hemorrágica Americana/genética , Fiebre Hemorrágica Americana/metabolismo , Fiebre Hemorrágica Americana/virología , Humanos , Virus Junin/efectos de los fármacos , Virus Junin/fisiología , Ratones , Unión Proteica , Transporte de Proteínas , Proteolisis , Ribonucleoproteínas/metabolismo , Tiourea/farmacología , Carga Viral , Proteínas Virales/metabolismo , Acoplamiento Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo
13.
Haematologica ; 100(4): 439-51, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25552701

RESUMEN

Clathrin-dependent endocytosis is an essential cellular process shared by all cell types. Despite this, precisely how endocytosis is regulated in a cell-type-specific manner and how this key pathway functions physiologically or pathophysiologically remain largely unknown. PICALM, which encodes the clathrin adaptor protein PICALM, was originally identified as a component of the CALM/AF10 leukemia oncogene. Here we show, by employing a series of conditional Picalm knockout mice, that PICALM critically regulates transferrin uptake in erythroid cells by functioning as a cell-type-specific regulator of transferrin receptor endocytosis. While transferrin receptor is essential for the development of all hematopoietic lineages, Picalm was dispensable for myeloid and B-lymphoid development. Furthermore, global Picalm inactivation in adult mice did not cause gross defects in mouse fitness, except for anemia and a coat color change. Freeze-etch electron microscopy of primary erythroblasts and live-cell imaging of murine embryonic fibroblasts revealed that Picalm function is required for efficient clathrin coat maturation. We showed that the PICALM PIP2 binding domain is necessary for transferrin receptor endocytosis in erythroblasts and absolutely essential for erythroid development from mouse hematopoietic stem/progenitor cells in an erythroid culture system. We further showed that Picalm deletion entirely abrogated the disease phenotype in a Jak2(V617F) knock-in murine model of polycythemia vera. Our findings provide new insights into the regulation of cell-type-specific transferrin receptor endocytosis in vivo. They also suggest a new strategy to block cellular uptake of transferrin-bound iron, with therapeutic potential for disorders characterized by inappropriate red blood cell production, such as polycythemia vera.


Asunto(s)
Hematopoyesis/genética , Proteínas de Ensamble de Clatrina Monoméricas/genética , Policitemia Vera/genética , Anemia Hipocrómica/genética , Animales , Diferenciación Celular , Vesículas Cubiertas por Clatrina/metabolismo , Vesículas Cubiertas por Clatrina/ultraestructura , Modelos Animales de Enfermedad , Endocitosis , Eritroblastos/metabolismo , Eritroblastos/ultraestructura , Eritropoyesis/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Inmunofenotipificación , Linfopoyesis/genética , Ratones , Ratones Noqueados , Proteínas de Ensamble de Clatrina Monoméricas/química , Proteínas de Ensamble de Clatrina Monoméricas/deficiencia , Proteínas de Ensamble de Clatrina Monoméricas/metabolismo , Mielopoyesis/genética , Fenotipo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Policitemia Vera/mortalidad , Dominios y Motivos de Interacción de Proteínas , Receptores de Transferrina/metabolismo
14.
bioRxiv ; 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38260258

RESUMEN

The endocytic pathway is both an essential route of molecular uptake in cells and a potential entry point for pathology-inducing cargo. The cell-to-cell spread of cytotoxic aggregates, such as those of α-synuclein (α-syn) in Parkinson's Disease (PD), exemplifies this duality. Here we used a human iPSC-derived induced neuronal model (iNs) prone to death mediated by aggregation in late endosomes and lysosomes of endogenous α-syn, seeded by internalized pre-formed fibrils of α-syn (PFFs). This PFF-mediated death was not observed with parental iPSCs or other non-neuronal cells. Using live-cell optical microscopy to visualize the read out of biosensors reporting endo-lysosome wounding, we discovered that up to about 10% of late endosomes and lysosomes in iNs exhibited spontaneous constitutive perforations, regardless of the presence of internalized PFFs. This wounding, absent in parental iPSCs and non-neuronal cells, corresponded to partial damage by nanopores in the limiting membranes of a subset of endolysosomes directly observed by volumetric focused ion beam scanning electron microscopy (FIB-SEM) in iNs and in CA1 pyramidal neurons from mouse brain, and not found in iPSCs or in other non-neuronal cells in culture or in mouse liver and skin. We suggest that the compromised limiting membranes in iNs and neurons in general are the primary conduit for cytosolic α-syn to access PFFs entrapped within endo-lysosomal lumens, initiating PFF-mediated α-syn aggregation. Significantly, eradicating the intrinsic endolysosomal perforations in iNs by inhibiting the endosomal Phosphatidylinositol-3-Phosphate/Phosphatidylinositol 5-Kinase (PIKfyve kinase) using Apilimod or Vacuolin-1 markedly reduced PFF-induced α-syn aggregation, despite PFFs continuing to enter the endolysosomal compartment. Crucially, this intervention also diminished iN death associated with PFF incubation. Our results reveal the surprising presence of intrinsically perforated endo-lysosomes in neurons, underscoring their crucial early involvement in the genesis of toxic α-syn aggregates induced by internalized PFFs. This discovery offers a basis for employing PIKfyve kinase inhibition as a potential therapeutic strategy to counteract synucleinopathies.

15.
Dev Cell ; 59(11): 1425-1438.e8, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38574735

RESUMEN

Mammalian Notch signaling occurs when the binding of Delta or Jagged to Notch stimulates the proteolytic release of the Notch intracellular domain (NICD), which enters the nucleus to control target gene expression. To determine the temporal dynamics of events associated with Notch signaling under native conditions, we fluorescently tagged Notch and Delta at their endogenous genomic loci and visualized them upon pairing of receiver (Notch) and sender (Delta) cells as a function of time after cell contact. At contact sites, Notch and Delta immediately accumulated at 1:1 stoichiometry in synapses, which resolved by 15-20 min after contact. Synapse formation preceded the entrance of the Notch extracellular domain into the sender cell and accumulation of NICD in the nucleus of the receiver cell, which approached a maximum after ∼45 min and was prevented by chemical and genetic inhibitors of signaling. These findings directly link Notch-Delta synapse dynamics to NICD production with spatiotemporal precision.


Asunto(s)
Núcleo Celular , Receptores Notch , Transducción de Señal , Sinapsis , Humanos , Núcleo Celular/metabolismo , Receptores Notch/metabolismo , Sinapsis/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Dominios Proteicos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética
16.
N Engl J Med ; 362(3): 206-16, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-20089971

RESUMEN

BACKGROUND: Establishing the genetic basis of phenotypes such as skeletal dysplasia in model organisms can provide insights into biologic processes and their role in human disease. METHODS: We screened mutagenized mice and observed a neonatal lethal skeletal dysplasia with an autosomal recessive pattern of inheritance. Through genetic mapping and positional cloning, we identified the causative mutation. RESULTS: Affected mice had a nonsense mutation in the thyroid hormone receptor interactor 11 gene (Trip11), which encodes the Golgi microtubule-associated protein 210 (GMAP-210); the affected mice lacked this protein. Golgi architecture was disturbed in multiple tissues, including cartilage. Skeletal development was severely impaired, with chondrocytes showing swelling and stress in the endoplasmic reticulum, abnormal cellular differentiation, and increased cell death. Golgi-mediated glycosylation events were altered in fibroblasts and chondrocytes lacking GMAP-210, and these chondrocytes had intracellular accumulation of perlecan, an extracellular matrix protein, but not of type II collagen or aggrecan, two other extracellular matrix proteins. The similarities between the skeletal and cellular phenotypes in these mice and those in patients with achondrogenesis type 1A, a neonatal lethal form of skeletal dysplasia in humans, suggested that achondrogenesis type 1A may be caused by GMAP-210 deficiency. Sequence analysis revealed loss-of-function mutations in the 10 unrelated patients with achondrogenesis type 1A whom we studied. CONCLUSIONS: GMAP-210 is required for the efficient glycosylation and cellular transport of multiple proteins. The identification of a mutation affecting GMAP-210 in mice, and then in humans, as the cause of a lethal skeletal dysplasia underscores the value of screening for abnormal phenotypes in model organisms and identifying the causative mutations.


Asunto(s)
Condrocitos/citología , Codón sin Sentido , Proteínas Nucleares/genética , Osteocondrodisplasias/genética , Animales , Diferenciación Celular , Proliferación Celular , Proteínas del Citoesqueleto , Retículo Endoplásmico/ultraestructura , Genes Recesivos , Glicosilación , Aparato de Golgi/ultraestructura , Humanos , Ratones , Ratones Mutantes , Proteínas Nucleares/deficiencia , Fenotipo , Polimorfismo de Nucleótido Simple , Procesamiento Proteico-Postraduccional/fisiología , Análisis de Secuencia de ADN
17.
J Virol ; 86(9): 5330-40, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22357278

RESUMEN

Viral pathogens usurp cell surface receptors to access clathrin endocytic structures, yet the mechanisms of virus incorporation into these structures remain incompletely understood. Here we used fluorescence microscopy to directly visualize the association of single canine parvovirus (CPV) capsids with cellular transferrin receptors (TfR) on the surfaces of live feline cells and to monitor how these CPV-TfR complexes access endocytic structures. We found that most capsids associated with fewer than five TfRs and that ∼25% of TfR-bound capsids laterally diffused into assembling clathrin-coated pits less than 30 s after attachment. Capsids that did not encounter a coated pit dissociated from the cell surface with a half-life of ∼30 s. Together, our results show how CPV exploits the natural mechanism of TfR endocytosis to engage the clathrin endocytic pathway and reveal that the low affinity of capsids for feline TfRs limits the residence time of capsids on the cell surface and thus the efficiency of virus internalization.


Asunto(s)
Clatrina/metabolismo , Invaginaciones Cubiertas de la Membrana Celular/metabolismo , Parvovirus Canino/metabolismo , Receptores de Transferrina/química , Animales , Cápside/metabolismo , Gatos , Línea Celular , Membrana Celular/metabolismo , Difusión , Perros , Endocitosis , Cinética , Unión Proteica , Receptores de Transferrina/metabolismo , Receptores Virales/metabolismo , Ensamble de Virus , Internalización del Virus
18.
J Cell Biol ; 222(2)2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36469001

RESUMEN

Volume electron microscopy is an important imaging modality in contemporary cell biology. Identification of intracellular structures is a laborious process limiting the effective use of this potentially powerful tool. We resolved this bottleneck with automated segmentation of intracellular substructures in electron microscopy (ASEM), a new pipeline to train a convolutional neural network to detect structures of a wide range in size and complexity. We obtained dedicated models for each structure based on a small number of sparsely annotated ground truth images from only one or two cells. Model generalization was improved with a rapid, computationally effective strategy to refine a trained model by including a few additional annotations. We identified mitochondria, Golgi apparatus, endoplasmic reticulum, nuclear pore complexes, caveolae, clathrin-coated pits, and vesicles imaged by focused ion beam scanning electron microscopy. We uncovered a wide range of membrane-nuclear pore diameters within a single cell and derived morphological metrics from clathrin-coated pits and vesicles, consistent with the classical constant-growth assembly model.


Asunto(s)
Procesamiento de Imagen Asistido por Computador , Microscopía Electrónica , Redes Neurales de la Computación , Clatrina , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/ultraestructura , Microscopía Electrónica/métodos , Mitocondrias/ultraestructura , Poro Nuclear/ultraestructura , Caveolas/ultraestructura , Biología Celular
19.
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.

20.
Sci Signal ; 16(796): eadg6474, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37527352

RESUMEN

Notch signaling relies on ligand-induced proteolysis of the transmembrane receptor Notch to liberate a nuclear effector that drives cell fate decisions. Upon ligand binding, sequential cleavage of Notch by the transmembrane protease ADAM10 and the intracellular protease γ-secretase releases the Notch intracellular domain (NICD), which translocates to the nucleus and forms a complex that induces target gene transcription. To map the location and timing of the individual steps required for the proteolysis and movement of Notch from the plasma membrane to the nucleus, we used proximity labeling with quantitative, multiplexed mass spectrometry to monitor the interaction partners of endogenous NOTCH2 after ligand stimulation in the presence of a γ-secretase inhibitor and as a function of time after inhibitor removal. Our studies showed that γ-secretase-mediated cleavage of NOTCH2 occurred in an intracellular compartment and that formation of nuclear complexes and recruitment of chromatin-modifying enzymes occurred within 45 min of inhibitor washout. These findings provide a detailed spatiotemporal map tracking the path of Notch from the plasma membrane to the nucleus and identify signaling events that are potential targets for modulating Notch activity.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide , Receptores Notch , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Ligandos , Receptores Notch/genética , Receptores Notch/metabolismo , Membrana Celular/metabolismo , Transducción de Señal , Receptor Notch1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA