Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 887
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(12): 2531-2543.e11, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37295401

RESUMEN

RNA editing is a widespread epigenetic process that can alter the amino acid sequence of proteins, termed "recoding." In cephalopods, most transcripts are recoded, and recoding is hypothesized to be an adaptive strategy to generate phenotypic plasticity. However, how animals use RNA recoding dynamically is largely unexplored. We investigated the function of cephalopod RNA recoding in the microtubule motor proteins kinesin and dynein. We found that squid rapidly employ RNA recoding in response to changes in ocean temperature, and kinesin variants generated in cold seawater displayed enhanced motile properties in single-molecule experiments conducted in the cold. We also identified tissue-specific recoded squid kinesin variants that displayed distinct motile properties. Finally, we showed that cephalopod recoding sites can guide the discovery of functional substitutions in non-cephalopod kinesin and dynein. Thus, RNA recoding is a dynamic mechanism that generates phenotypic plasticity in cephalopods and can inform the characterization of conserved non-cephalopod proteins.


Asunto(s)
Cefalópodos , Dineínas , Animales , Dineínas/genética , Dineínas/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , ARN/metabolismo , Cefalópodos/genética , Cefalópodos/metabolismo , Proteínas/metabolismo , Microtúbulos/metabolismo , Proteínas de Microtúbulos , Miosinas/metabolismo
2.
Cell ; 184(11): 2860-2877.e22, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33964210

RESUMEN

Most human embryos are aneuploid. Aneuploidy frequently arises during the early mitotic divisions of the embryo, but its origin remains elusive. Human zygotes that cluster their nucleoli at the pronuclear interface are thought to be more likely to develop into healthy euploid embryos. Here, we show that the parental genomes cluster with nucleoli in each pronucleus within human and bovine zygotes, and clustering is required for the reliable unification of the parental genomes after fertilization. During migration of intact pronuclei, the parental genomes polarize toward each other in a process driven by centrosomes, dynein, microtubules, and nuclear pore complexes. The maternal and paternal chromosomes eventually cluster at the pronuclear interface, in direct proximity to each other, yet separated. Parental genome clustering ensures the rapid unification of the parental genomes on nuclear envelope breakdown. However, clustering often fails, leading to chromosome segregation errors and micronuclei, incompatible with healthy embryo development.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Aneuploidia , Animales , Bovinos , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Centrosoma/metabolismo , Segregación Cromosómica/fisiología , Cromosomas/metabolismo , Fertilización/genética , Humanos , Masculino , Microtúbulos/metabolismo , Mitosis , Oocitos/metabolismo , Espermatozoides/metabolismo , Cigoto/metabolismo
3.
Annu Rev Cell Dev Biol ; 38: 155-178, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35905769

RESUMEN

Eukaryotic cells across the tree of life organize their subcellular components via intracellular transport mechanisms. In canonical transport, myosin, kinesin, and dynein motor proteins interact with cargos via adaptor proteins and move along filamentous actin or microtubule tracks. In contrast to this canonical mode, hitchhiking is a newly discovered mode of intracellular transport in which a cargo attaches itself to an already-motile cargo rather than directly associating with a motor protein itself. Many cargos including messenger RNAs, protein complexes, and organelles hitchhike on membrane-bound cargos. Hitchhiking-like behaviors have been shown to impact cellular processes including local protein translation, long-distance signaling, and organelle network reorganization. Here, we review instances of cargo hitchhiking in fungal, animal, and plant cells and discuss the potential cellular and evolutionary importance of hitchhiking in these different contexts.


Asunto(s)
Dineínas , Cinesinas , Actinas/metabolismo , Animales , Dineínas/genética , Dineínas/metabolismo , Cinesinas/genética , Microtúbulos/genética , Microtúbulos/metabolismo , Miosinas/genética , Miosinas/metabolismo , Células Vegetales/metabolismo
4.
Cell ; 175(3): 822-834.e18, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30318141

RESUMEN

Mdn1 is an essential AAA (ATPase associated with various activities) protein that removes assembly factors from distinct precursors of the ribosomal 60S subunit. However, Mdn1's large size (∼5,000 amino acid [aa]) and its limited homology to other well-studied proteins have restricted our understanding of its remodeling function. Here, we present structures for S. pombe Mdn1 in the presence of AMPPNP at up to ∼4 Å or ATP plus Rbin-1, a chemical inhibitor, at ∼8 Å resolution. These data reveal that Mdn1's MIDAS domain is tethered to its ring-shaped AAA domain through an ∼20 nm long structured linker and a flexible ∼500 aa Asp/Glu-rich motif. We find that the MIDAS domain, which also binds other ribosome-assembly factors, docks onto the AAA ring in a nucleotide state-specific manner. Together, our findings reveal how conformational changes in the AAA ring can be directly transmitted to the MIDAS domain and thereby drive the targeted release of assembly factors from ribosomal 60S-subunit precursors.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/química , Simulación de Dinámica Molecular , Proteínas de Schizosaccharomyces pombe/química , Schizosaccharomyces/enzimología , Secuencias de Aminoácidos , Animales , Sitios de Unión , Microscopía por Crioelectrón , Biogénesis de Organelos , Unión Proteica , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Células Sf9 , Spodoptera
5.
Cell ; 169(7): 1303-1314.e18, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28602352

RESUMEN

Cytoplasmic dynein-1 binds dynactin and cargo adaptor proteins to form a transport machine capable of long-distance processive movement along microtubules. However, it is unclear why dynein-1 moves poorly on its own or how it is activated by dynactin. Here, we present a cryoelectron microscopy structure of the complete 1.4-megadalton human dynein-1 complex in an inhibited state known as the phi-particle. We reveal the 3D structure of the cargo binding dynein tail and show how self-dimerization of the motor domains locks them in a conformation with low microtubule affinity. Disrupting motor dimerization with structure-based mutagenesis drives dynein-1 into an open form with higher affinity for both microtubules and dynactin. We find the open form is also inhibited for movement and that dynactin relieves this by reorienting the motor domains to interact correctly with microtubules. Our model explains how dynactin binding to the dynein-1 tail directly stimulates its motor activity.


Asunto(s)
Dineínas Citoplasmáticas/química , Complejos Multiproteicos/química , Animales , Microscopía por Crioelectrón , Dineínas Citoplasmáticas/metabolismo , Dineínas Citoplasmáticas/ultraestructura , Dimerización , Complejo Dinactina/química , Complejo Dinactina/metabolismo , Humanos , Ratones , Microtúbulos/química , Microtúbulos/metabolismo , Modelos Moleculares , Proteínas Motoras Moleculares/química , Proteínas Motoras Moleculares/metabolismo , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Células Sf9 , Spodoptera , Porcinos
6.
Cell ; 170(6): 1197-1208.e12, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28886386

RESUMEN

Regulation is central to the functional versatility of cytoplasmic dynein, a motor involved in intracellular transport, cell division, and neurodevelopment. Previous work established that Lis1, a conserved regulator of dynein, binds to its motor domain and induces a tight microtubule-binding state in dynein. The work we present here-a combination of biochemistry, single-molecule assays, and cryoelectron microscopy-led to the surprising discovery that Lis1 has two opposing modes of regulating dynein, being capable of inducing both low and high affinity for the microtubule. We show that these opposing modes depend on the stoichiometry of Lis1 binding to dynein and that this stoichiometry is regulated by the nucleotide state of dynein's AAA3 domain. The low-affinity state requires Lis1 to also bind to dynein at a novel conserved site, mutation of which disrupts Lis1's function in vivo. We propose a new model for the regulation of dynein by Lis1.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Dineínas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/química , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Microscopía por Crioelectrón , Dineínas/química , Humanos , Proteínas Asociadas a Microtúbulos/química , Modelos Moleculares , Proteínas Motoras Moleculares/metabolismo , Dominios Proteicos , Proteínas de Saccharomyces cerevisiae/química , Alineación de Secuencia
7.
EMBO J ; 43(7): 1257-1272, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38454149

RESUMEN

Dynein-2 is a large multiprotein complex that powers retrograde intraflagellar transport (IFT) of cargoes within cilia/flagella, but the molecular mechanism underlying this function is still emerging. Distinctively, dynein-2 contains two identical force-generating heavy chains that interact with two different intermediate chains (WDR34 and WDR60). Here, we dissect regulation of dynein-2 function by WDR34 and WDR60 using an integrative approach including cryo-electron microscopy and CRISPR/Cas9-enabled cell biology. A 3.9 Å resolution structure shows how WDR34 and WDR60 use surprisingly different interactions to engage equivalent sites of the two heavy chains. We show that cilia can assemble in the absence of either WDR34 or WDR60 individually, but not both subunits. Dynein-2-dependent distribution of cargoes depends more strongly on WDR60, because the unique N-terminal extension of WDR60 facilitates dynein-2 targeting to cilia. Strikingly, this N-terminal extension can be transplanted onto WDR34 and retain function, suggesting it acts as a flexible tether to the IFT "trains" that assemble at the ciliary base. We discuss how use of unstructured tethers represents an emerging theme in IFT train interactions.


Asunto(s)
Cilios , Dineínas , Dineínas/metabolismo , Microscopía por Crioelectrón , Transporte Biológico , Cilios/metabolismo , Flagelos/metabolismo
8.
Trends Biochem Sci ; 48(4): 315-316, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36754682

RESUMEN

In a recent study, Chaaban and Carter use cryo-electron microscopy (cryo-EM) and an innovative data-processing pipeline to determine the first high-resolution structure of the dynein-dynactin-BICDR1 complex assembled on microtubules. The structure of the complex reveals novel stoichiometry and provides new mechanistic insight into dynein function and mechanism.


Asunto(s)
Dineínas , Proteínas Asociadas a Microtúbulos , Dineínas/metabolismo , Proteínas Asociadas a Microtúbulos/análisis , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/metabolismo , Microscopía por Crioelectrón , Microtúbulos/química , Microtúbulos/metabolismo , Complejo Dinactina/análisis , Complejo Dinactina/química , Complejo Dinactina/metabolismo
9.
EMBO J ; 42(12): e112466, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37051721

RESUMEN

Axonemal outer dynein arm (ODA) motors generate force for ciliary beating. We analyzed three states of the ODA during the power stroke cycle using in situ cryo-electron tomography, subtomogram averaging, and classification. These states of force generation depict the prepower stroke, postpower stroke, and intermediate state conformations. Comparison of these conformations to published in vitro atomic structures of cytoplasmic dynein, ODA, and the Shulin-ODA complex revealed differences in the orientation and position of the dynein head. Our analysis shows that in the absence of ATP, all dynein linkers interact with the AAA3/AAA4 domains, indicating that interactions with the adjacent microtubule doublet B-tubule direct dynein orientation. For the prepower stroke conformation, there were changes in the tail that is anchored on the A-tubule. We built models starting with available high-resolution structures to generate a best-fitting model structure for the in situ pre- and postpower stroke ODA conformations, thereby showing that ODA in a complex with Shulin adopts a similar conformation as the active prepower stroke ODA in the axoneme.


Asunto(s)
Dineínas , Tomografía con Microscopio Electrónico , Dineínas/metabolismo , Dineínas Axonemales/química , Dineínas Axonemales/metabolismo , Axonema/metabolismo , Cilios/metabolismo , Adenosina Trifosfato , Flagelos/metabolismo
10.
EMBO J ; 42(23): e114473, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37872872

RESUMEN

The microtubule motor dynein mediates polarised trafficking of a wide variety of organelles, vesicles and macromolecules. These functions are dependent on the dynactin complex, which helps recruit cargoes to dynein's tail and activates motor movement. How the dynein-dynactin complex orchestrates trafficking of diverse cargoes is unclear. Here, we identify HEATR5B, an interactor of the adaptor protein-1 (AP1) clathrin adaptor complex, as a novel player in dynein-dynactin function. HEATR5B was recovered in a biochemical screen for proteins whose association with the dynein tail is augmented by dynactin. We show that HEATR5B binds directly to the dynein tail and dynactin and stimulates motility of AP1-associated endosomal membranes in human cells. We also demonstrate that the Drosophila HEATR5B homologue is an essential gene that selectively promotes dynein-based transport of AP1-bound membranes to the Golgi apparatus. As HEATR5B lacks the coiled-coil architecture typical of dynein adaptors, our data point to a non-canonical process orchestrating motor function on a specific cargo. We additionally show that HEATR5B promotes association of AP1 with endosomal membranes independently of dynein. Thus, HEATR5B co-ordinates multiple events in AP1-based trafficking.


Asunto(s)
Dineínas , Proteínas Asociadas a Microtúbulos , Humanos , Dineínas/metabolismo , Complejo Dinactina/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Transporte Biológico/fisiología , Microtúbulos/metabolismo , Endosomas/metabolismo
11.
Development ; 151(14)2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39007638

RESUMEN

Vertebrate motile cilia are classified as (9+2) or (9+0), based on the presence or absence of the central pair apparatus, respectively. Cryogenic electron microscopy analyses of (9+2) cilia have uncovered an elaborate axonemal protein composition. The extent to which these features are conserved in (9+0) cilia remains unclear. CFAP53, a key axonemal filamentous microtubule inner protein (fMIP) and a centriolar satellites component, is essential for motility of (9+0), but not (9+2) cilia. Here, we show that in (9+2) cilia, CFAP53 functions redundantly with a paralogous fMIP, MNS1. MNS1 localises to ciliary axonemes, and combined loss of both proteins in zebrafish and mice caused severe outer dynein arm loss from (9+2) cilia, significantly affecting their motility. Using immunoprecipitation, we demonstrate that, whereas MNS1 can associate with itself and CFAP53, CFAP53 is unable to self-associate. We also show that additional axonemal dynein-interacting proteins, two outer dynein arm docking (ODAD) complex members, show differential localisation between types of motile cilia. Together, our findings clarify how paralogous fMIPs, CFAP53 and MNS1, function in regulating (9+2) versus (9+0) cilia motility, and further emphasise extensive structural diversity among these organelles.


Asunto(s)
Axonema , Cilios , Pez Cebra , Animales , Cilios/metabolismo , Cilios/ultraestructura , Pez Cebra/metabolismo , Ratones , Axonema/metabolismo , Axonema/ultraestructura , Dineínas Axonemales/metabolismo , Dineínas Axonemales/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Dineínas/metabolismo
12.
Proc Natl Acad Sci U S A ; 121(5): e2318522121, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38261620

RESUMEN

Axonemal dynein motors drive ciliary motility and can consist of up to twenty distinct components with a combined mass of ~2 MDa. In mammals, failure of dyneins to assemble within the axonemal superstructure leads to primary ciliary dyskinesia. Syndromic phenotypes include infertility, rhinitis, severe bronchial conditions, and situs inversus. Nineteen specific cytosolic factors (Dynein Axonemal Assembly Factors; DNAAFs) are necessary for axonemal dynein assembly, although the detailed mechanisms involved remain very unclear. Here, we identify the essential assembly factor DNAAF3 as a structural ortholog of S-adenosylmethionine-dependent methyltransferases. We demonstrate that dynein heavy chains, especially those forming the ciliary outer arms, are methylated on key residues within various nucleotide-binding sites and on microtubule-binding domain helices directly involved in the transition to low binding affinity. These variable modifications, which are generally missing in a Chlamydomonas null mutant for the DNAAF3 ortholog PF22 (DAB1), likely impact on motor mechanochemistry fine-tuning the activities of individual dynein complexes.


Asunto(s)
Dineínas Axonemales , Metiltransferasas , Animales , Citosol , Citoesqueleto , Metilación , Mamíferos
13.
EMBO J ; 41(5): e108119, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35099830

RESUMEN

Lysosomes function not only as degradatory compartments but also as dynamic intracellular calcium ion stores. The transient receptor potential mucolipin 1 (TRPML1) channel mediates lysosomal Ca2+ release, thereby participating in multiple cellular functions. The pentameric Ragulator complex, which plays a critical role in the activation of mTORC1, is also involved in lysosomal trafficking and is anchored to lysosomes through its LAMTOR1 subunit. Here, we report that the Ragulator restricts lysosomal trafficking in dendrites of hippocampal neurons via LAMTOR1-mediated tonic inhibition of TRPML1 activity, independently of mTORC1. LAMTOR1 directly interacts with TRPML1 through its N-terminal domain. Eliminating this inhibition in hippocampal neurons by LAMTOR1 deletion or by disrupting LAMTOR1-TRPML1 binding increases TRPML1-mediated Ca2+ release and facilitates dendritic lysosomal trafficking powered by dynein. LAMTOR1 deletion in the hippocampal CA1 region of adult mice results in alterations in synaptic plasticity, and in impaired object-recognition memory and contextual fear conditioning, due to TRPML1 activation. Mechanistically, changes in synaptic plasticity are associated with increased GluA1 dephosphorylation by calcineurin and lysosomal degradation. Thus, LAMTOR1-mediated inhibition of TRPML1 is critical for regulating dendritic lysosomal motility, synaptic plasticity, and learning.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Calcio/metabolismo , Hipocampo/metabolismo , Lisosomas/metabolismo , Neuronas/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Células HeLa , Humanos , Ratones , Plasticidad Neuronal/fisiología
14.
J Cell Sci ; 137(20)2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38804679

RESUMEN

The definitive demonstration of protein localization on primary cilia has been a challenge for cilia biologists. Primary cilia are solitary thread-like projections that have a specialized protein composition, but as the ciliary structure overlays the cell membrane and other cell parts, the identity of ciliary proteins are difficult to ascertain by conventional imaging approaches like immunofluorescence microscopy. Surface scanning electron microscopy combined with immunolabeling (immuno-SEM) bypasses some of these indeterminacies by unambiguously showing protein expression in the context of the three-dimensional ultrastructure of the cilium. Here, we apply immuno-SEM to specifically identify proteins on the primary cilia of mouse and human pancreatic islets, including post-translationally modified tubulin, intraflagellar transport (IFT)88, the small GTPase Arl13b, as well as subunits of axonemal dynein. Key parameters in sample preparation, immunolabeling and imaging acquisition are discussed to facilitate similar studies by others in the cilia research community.


Asunto(s)
Cilios , Islotes Pancreáticos , Cilios/ultraestructura , Cilios/metabolismo , Animales , Humanos , Ratones , Islotes Pancreáticos/ultraestructura , Islotes Pancreáticos/metabolismo , Microscopía Electrónica de Rastreo/métodos
15.
J Cell Sci ; 137(3)2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38329417

RESUMEN

Most of the vesicular transport pathways inside the cell are facilitated by molecular motors that move along cytoskeletal networks. Autophagy is a well-explored catabolic pathway that is initiated by the formation of an isolation membrane known as the phagophore, which expands to form a double-membraned structure that captures its cargo and eventually moves towards the lysosomes for fusion. Molecular motors and cytoskeletal elements have been suggested to participate at different stages of the process as the autophagic vesicles move along cytoskeletal tracks. Dynein and kinesins govern autophagosome trafficking on microtubules through the sequential recruitment of their effector proteins, post-translational modifications and interactions with LC3-interacting regions (LIRs). In contrast, myosins are actin-based motors that participate in various stages of the autophagic flux, as well as in selective autophagy pathways. However, several outstanding questions remain with regard to how the dominance of a particular motor protein over another is controlled, and to the molecular mechanisms that underlie specific disease variants in motor proteins. In this Review, we aim to provide an overview of the role of molecular motors in autophagic flux, as well as highlight their dysregulation in diseases, such as neurodegenerative disorders and pathogenic infections, and ageing.


Asunto(s)
Autofagosomas , Autofagia , Citoesqueleto , Actinas , Dineínas , Cinesinas
16.
J Cell Sci ; 137(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38533689

RESUMEN

Primary cilia are essential eukaryotic organelles required for signalling and secretion. Dynein-2 is a microtubule-motor protein complex and is required for ciliogenesis via its role in facilitating retrograde intraflagellar transport (IFT) from the cilia tip to the cell body. Dynein-2 must be assembled and loaded onto IFT trains for entry into cilia for this process to occur, but how dynein-2 is assembled and how it is recycled back into a cilium remain poorly understood. Here, we identify centrosomal protein of 170 kDa (CEP170) as a dynein-2-interacting protein in mammalian cells. We show that loss of CEP170 perturbs intraflagellar transport and hedgehog signalling, and alters the stability of dynein-2 holoenzyme complex. Together, our data indicate a role for CEP170 in supporting cilia function and dynein-2 assembly.


Asunto(s)
Cilios , Proteínas Asociadas a Microtúbulos , Cilios/metabolismo , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Animales , Dineínas/metabolismo , Dineínas/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Transducción de Señal , Ratones , Flagelos/metabolismo
17.
Proc Natl Acad Sci U S A ; 120(39): e2303376120, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37722034

RESUMEN

In many species, only one oocyte is specified among a group of interconnected germline sister cells. In Drosophila melanogaster, 16 interconnected cells form a germline cyst, where one cell differentiates into an oocyte, while the rest become nurse cells that supply the oocyte with mRNAs, proteins, and organelles through intercellular cytoplasmic bridges named ring canals via microtubule-based transport. In this study, we find that a microtubule polymerase Mini spindles (Msps), the Drosophila homolog of XMAP215, is essential for maintenance of the oocyte specification. mRNA encoding Msps is transported and concentrated in the oocyte by dynein-dependent transport along microtubules. Translated Msps stimulates microtubule polymerization in the oocyte, causing more microtubule plus ends to grow from the oocyte through the ring canals into nurse cells, further enhancing nurse cell-to-oocyte transport by dynein. Knockdown of msps blocks the oocyte growth and causes gradual loss of oocyte determinants. Thus, the Msps-dynein duo creates a positive feedback loop, ensuring oocyte fate maintenance by promoting high microtubule polymerization activity in the oocyte, and enhancing dynein-dependent nurse cell-to-oocyte transport.


Asunto(s)
Dineínas Citoplasmáticas , Drosophila , Animales , Drosophila melanogaster , Microtúbulos , Nucleotidiltransferasas , Oocitos
18.
Proc Natl Acad Sci U S A ; 120(39): e2302801120, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37722038

RESUMEN

Primary cilia are specialized organelles supporting the development and function of cells and organisms. Intraflagellar transport (IFT) is essential for cilia formation, maintenance, and function. In C. elegans ciliated sensory neurons, IFT interacts with signaling molecules to generate distinct morphological and function features and also to maintain the integrity of cilia. Here, we report an IFT-dependent feedback control on the conserved MAPKKK DLK-1 in the ciliated sensory neurons. DLK proteins are widely known to act in synapse formation, axon regeneration, and degeneration, but their roles in other neuronal compartments are understudied. By forward genetic screening for altered expression of the endogenously tagged DLK-1 we identified multiple ift mutants showing increased DLK-1 accumulation in the defective sensory endings. We show that in response to acute IFT disruption, DLK-1 accumulates rapidly and reversibly. The expression levels of the transcription factor CEBP-1, known to act downstream of DLK-1 in the development and maintenance of synapses and axons, are also increased in the ciliated sensory neurons of ift mutants. Interestingly, the regulation of CEBP-1 expression shows sensory neuron-type dependency on DLK-1. Moreover, in the sensory neuron AWC, which has elaborate cilia morphology, up-regulated CEBP-1 represses DLK-1 at the transcription level, thereby dampening DLK-1 accumulation. Last, the IFT-dependent regulatory loop of DLK-1 and CEBP-1 offers neuroprotection in a cilia degeneration model. These findings uncover a surveillance mechanism in which tight control on the DLK-1 signaling protects cilia integrity in a context-specific manner.


Asunto(s)
Proteínas de Caenorhabditis elegans , Cilios , Animales , Cilios/genética , Retroalimentación , Axones , Caenorhabditis elegans/genética , Regeneración Nerviosa , Células Receptoras Sensoriales , Quinasas Quinasa Quinasa PAM , Proteínas de Caenorhabditis elegans/genética
19.
Traffic ; 24(3): 114-130, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35146839

RESUMEN

The cytoskeleton is an essential component of the cell and it is involved in multiple physiological functions, including intracellular organization and transport. It is composed of three main families of proteinaceous filaments; microtubules, actin filaments and intermediate filaments and their accessory proteins. Motor proteins, which comprise the dynein, kinesin and myosin superfamilies, are a remarkable group of accessory proteins that mainly mediate the intracellular transport of cargoes along with the cytoskeleton. Like other cellular structures and pathways, viruses can exploit the cytoskeleton to promote different steps of their life cycle through associations with motor proteins. The complexity of the cytoskeleton and the differences among viruses, however, has led to a wide diversity of interactions, which in most cases remain poorly understood. Unveiling the details of these interactions is necessary not only for a better comprehension of specific infections, but may also reveal new potential drug targets to fight dreadful diseases such as rabies disease and acquired immunodeficiency syndrome (AIDS). In this review, we describe a few examples of the mechanisms that some human viruses, that is, rabies virus, adenovirus, herpes simplex virus, human immunodeficiency virus, influenza A virus and papillomavirus, have developed to hijack dyneins, kinesins and myosins.


Asunto(s)
Proteínas del Citoesqueleto , Virus , Humanos , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Virus/metabolismo , Microtúbulos/metabolismo , Citoesqueleto de Actina/metabolismo , Miosinas/metabolismo , Cinesinas/metabolismo , Dineínas/metabolismo
20.
J Biol Chem ; 300(6): 107323, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677516

RESUMEN

Organelles and vesicular cargoes are transported by teams of kinesin and dynein motors along microtubules. We isolated endocytic organelles from cells at different stages of maturation and reconstituted their motility along microtubules in vitro. We asked how the sets of motors transporting a cargo determine its motility and response to the microtubule-associated protein tau. Here, we find that phagosomes move in both directions along microtubules, but the directional bias changes during maturation. Early phagosomes exhibit retrograde-biased transport while late phagosomes are directionally unbiased. Correspondingly, early and late phagosomes are bound by different numbers and combinations of kinesins-1, -2, -3, and dynein. Tau stabilizes microtubules and directs transport within neurons. While single-molecule studies show that tau differentially regulates the motility of kinesins and dynein in vitro, less is known about its role in modulating the trafficking of endogenous cargoes transported by their native teams of motors. Previous studies showed that tau preferentially inhibits kinesin motors, which biases late phagosome transport towards the microtubule minus-end. Here, we show that tau strongly inhibits long-range, dynein-mediated motility of early phagosomes. Tau reduces forces generated by teams of dynein motors on early phagosomes and accelerates dynein unbinding under load. Thus, cargoes differentially respond to tau, where dynein complexes on early phagosomes are more sensitive to tau inhibition than those on late phagosomes. Mathematical modeling further explains how small changes in the number of kinesins and dynein on cargoes impact the net directionality but also that cargoes with different sets of motors respond differently to tau.


Asunto(s)
Dineínas , Cinesinas , Microtúbulos , Proteínas tau , Cinesinas/metabolismo , Cinesinas/genética , Proteínas tau/metabolismo , Proteínas tau/genética , Dineínas/metabolismo , Dineínas/genética , Animales , Microtúbulos/metabolismo , Fagosomas/metabolismo , Transporte Biológico , Ratones , Humanos , Endocitosis/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA