Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 442
Filtrar
1.
Methods Mol Biol ; 2828: 79-85, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39147972

RESUMEN

Bacteria can propel themselves by rotating a flagellum or a flagellar bundle. To image this thin structure in motile bacteria, the flagella can be vitally stained with fluorophores. This chapter describes a flagellar staining protocol with the additional possibility of visualizing the cell body. It offers the opportunity to track conformational changes of flagella and simultaneously track the positions of the cell bodies. The additional use of a filter increases the number of motile cells and improves the signal-to-noise ratio of images. The flagellar staining requires a prior introduction of a surface-exposed cysteine, which is not covered in this chapter.


Asunto(s)
Bacterias , Flagelos , Colorantes Fluorescentes , Coloración y Etiquetado , Flagelos/metabolismo , Flagelos/ultraestructura , Colorantes Fluorescentes/química , Coloración y Etiquetado/métodos , Bacterias/metabolismo , Microscopía Fluorescente/métodos
2.
Nat Commun ; 15(1): 3456, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658528

RESUMEN

Intraflagellar transport (IFT) orchestrates entry of proteins into primary cilia. At the ciliary base, assembled IFT trains, driven by kinesin-2 motors, can transport cargo proteins into the cilium, across the crowded transition zone. How trains assemble at the base and how proteins associate with them is far from understood. Here, we use single-molecule imaging in the cilia of C. elegans chemosensory neurons to directly visualize the entry of kinesin-2 motors, kinesin-II and OSM-3, as well as anterograde cargo proteins, IFT dynein and tubulin. Single-particle tracking shows that IFT components associate with trains sequentially, both in time and space. Super-resolution maps of IFT components in wild-type and mutant worms reveal ciliary ultrastructure and show that kinesin-II is essential for axonemal organization. Finally, imaging cilia lacking kinesin-II and/or transition zone function uncovers the interplay of kinesin-II and OSM-3 in driving efficient transport of IFT trains across the transition zone.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Cilios , Cinesinas , Caenorhabditis elegans/metabolismo , Animales , Cilios/metabolismo , Cilios/ultraestructura , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Cinesinas/metabolismo , Cinesinas/genética , Flagelos/metabolismo , Flagelos/ultraestructura , Tubulina (Proteína)/metabolismo , Axonema/metabolismo , Axonema/ultraestructura , Dineínas/metabolismo , Transporte Biológico , Imagen Individual de Molécula , Transporte de Proteínas
3.
Nature ; 623(7985): 193-201, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37880360

RESUMEN

Voltage-sensing domains control the activation of voltage-gated ion channels, with a few exceptions1. One such exception is the sperm-specific Na+/H+ exchanger SLC9C1, which is the only known transporter to be regulated by voltage-sensing domains2-5. After hyperpolarization of sperm flagella, SLC9C1 becomes active, causing pH alkalinization and CatSper Ca2+ channel activation, which drives chemotaxis2,6. SLC9C1 activation is further regulated by cAMP2,7, which is produced by soluble adenyl cyclase (sAC). SLC9C1 is therefore an essential component of the pH-sAC-cAMP signalling pathway in metazoa8,9, required for sperm motility and fertilization4. Despite its importance, the molecular basis of SLC9C1 voltage activation is unclear. Here we report cryo-electron microscopy (cryo-EM) structures of sea urchin SLC9C1 in detergent and nanodiscs. We show that the voltage-sensing domains are positioned in an unusual configuration, sandwiching each side of the SLC9C1 homodimer. The S4 segment is very long, 90 Å in length, and connects the voltage-sensing domains to the cytoplasmic cyclic-nucleotide-binding domains. The S4 segment is in the up configuration-the inactive state of SLC9C1. Consistently, although a negatively charged cavity is accessible for Na+ to bind to the ion-transporting domains of SLC9C1, an intracellular helix connected to S4 restricts their movement. On the basis of the differences in the cryo-EM structure of SLC9C1 in the presence of cAMP, we propose that, upon hyperpolarization, the S4 segment moves down, removing this constriction and enabling Na+/H+ exchange.


Asunto(s)
Microscopía por Crioelectrón , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Activación del Canal Iónico , Erizos de Mar , Intercambiadores de Sodio-Hidrógeno , Animales , Masculino , Adenilil Ciclasas/metabolismo , AMP Cíclico/metabolismo , Flagelos/química , Flagelos/metabolismo , Flagelos/ultraestructura , Concentración de Iones de Hidrógeno , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/química , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/ultraestructura , Potenciales de la Membrana , Multimerización de Proteína , Erizos de Mar/química , Erizos de Mar/metabolismo , Erizos de Mar/ultraestructura , Intercambiadores de Sodio-Hidrógeno/química , Intercambiadores de Sodio-Hidrógeno/metabolismo , Intercambiadores de Sodio-Hidrógeno/ultraestructura , Motilidad Espermática , Espermatozoides/química , Espermatozoides/metabolismo , Espermatozoides/ultraestructura
4.
Science ; 377(6605): 543-548, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35901159

RESUMEN

The cilium is an antenna-like organelle that performs numerous cellular functions, including motility, sensing, and signaling. The base of the cilium contains a selective barrier that regulates the entry of large intraflagellar transport (IFT) trains, which carry cargo proteins required for ciliary assembly and maintenance. However, the native architecture of the ciliary base and the process of IFT train assembly remain unresolved. In this work, we used in situ cryo-electron tomography to reveal native structures of the transition zone region and assembling IFT trains at the ciliary base in Chlamydomonas. We combined this direct cellular visualization with ultrastructure expansion microscopy to describe the front-to-back stepwise assembly of IFT trains: IFT-B forms the backbone, onto which bind IFT-A, dynein-1b, and finally kinesin-2 before entry into the cilium.


Asunto(s)
Chlamydomonas , Cilios , Flagelos , Chlamydomonas/metabolismo , Cilios/metabolismo , Microscopía por Crioelectrón/métodos , Dineínas/metabolismo , Tomografía con Microscopio Electrónico/métodos , Flagelos/metabolismo , Flagelos/ultraestructura , Cinesinas/metabolismo , Transporte de Proteínas , Transducción de Señal
5.
J Mol Biol ; 433(21): 167188, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34454944

RESUMEN

Type III protein secretion is widespread in Gram-negative pathogens. It comprises the injectisome with a surface-exposed needle and an inner membrane translocase. The translocase contains the SctRSTU export channel enveloped by the export gate subunit SctV that binds chaperone/exported clients and forms a putative ante-chamber. We probed the assembly, function, structure and dynamics of SctV from enteropathogenic E. coli (EPEC). In both EPEC and E. coli lab strains, SctV forms peripheral oligomeric clusters that are detergent-extracted as homo-nonamers. Membrane-embedded SctV9 is necessary and sufficient to act as a receptor for different chaperone/exported protein pairs with distinct C-domain binding sites that are essential for secretion. Negative staining electron microscopy revealed that peptidisc-reconstituted His-SctV9 forms a tripartite particle of ∼22 nm with a N-terminal domain connected by a short linker to a C-domain ring structure with a ∼5 nm-wide inner opening. The isolated C-domain ring was resolved with cryo-EM at 3.1 Å and structurally compared to other SctV homologues. Its four sub-domains undergo a three-stage "pinching" motion. Hydrogen-deuterium exchange mass spectrometry revealed this to involve dynamic and rigid hinges and a hyper-flexible sub-domain that flips out of the ring periphery and binds chaperones on and between adjacent protomers. These motions are coincident with local conformational changes at the pore surface and ring entry mouth that may also be modulated by the ATPase inner stalk. We propose that the intrinsic dynamics of the SctV protomer are modulated by chaperones and the ATPase and could affect allosterically the other subunits of the nonameric ring during secretion.


Asunto(s)
Adenosina Trifosfatasas/química , Escherichia coli Enteropatógena/ultraestructura , Proteínas de Escherichia coli/química , Flagelos/ultraestructura , Canales de Translocación SEC/química , Sistemas de Secreción Tipo III/ultraestructura , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Regulación Alostérica , Sitios de Unión , Clonación Molecular , Microscopía por Crioelectrón , Medición de Intercambio de Deuterio , Escherichia coli Enteropatógena/genética , Escherichia coli Enteropatógena/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Flagelos/genética , Flagelos/metabolismo , Expresión Génica , Regulación Bacteriana de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Cinética , Espectrometría de Masas , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canales de Translocación SEC/genética , Canales de Translocación SEC/metabolismo , Especificidad por Sustrato , Sistemas de Secreción Tipo III/genética , Sistemas de Secreción Tipo III/metabolismo
6.
PLoS Genet ; 17(3): e1009388, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33661892

RESUMEN

Ciliary beating requires the coordinated activity of numerous axonemal complexes. The protein composition and role of radial spokes (RS), nexin links (N-DRC) and dyneins (ODAs and IDAs) is well established. However, how information is transmitted from the central apparatus to the RS and across other ciliary structures remains unclear. Here, we identify a complex comprising the evolutionarily conserved proteins Ccdc96 and Ccdc113, positioned parallel to N-DRC and forming a connection between RS3, dynein g, and N-DRC. Although Ccdc96 and Ccdc113 can be transported to cilia independently, their stable docking and function requires the presence of both proteins. Deletion of either CCDC113 or CCDC96 alters cilia beating frequency, amplitude and waveform. We propose that the Ccdc113/Ccdc96 complex transmits signals from RS3 and N-DRC to dynein g and thus regulates its activity and the ciliary beat pattern.


Asunto(s)
Proteínas Portadoras/metabolismo , Cilios/fisiología , Dineínas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas de Plantas/metabolismo , Axonema/metabolismo , Proteínas Portadoras/química , Chlamydomonas/fisiología , Cilios/ultraestructura , Flagelos/fisiología , Flagelos/ultraestructura , Técnica del Anticuerpo Fluorescente , Proteínas Asociadas a Microtúbulos/química , Complejos Multiproteicos/ultraestructura , Conformación Proteica , Transporte de Proteínas , Relación Estructura-Actividad , Tetrahymena thermophila/fisiología
7.
Parasitol Res ; 120(3): 1131-1135, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33511472

RESUMEN

Giardia comprises one genus with several morphologically distinct species described in mammals (including humans, marsupials, rodents), birds, and amphibians. This group of protists provokes diarrhoea diseases in humans and animals worldwide. Transmission of the parasite occurs through the faecal-oral route. Regarding the presence of Giardia in invertebrates, some works have shown that flies can transmit Giardia cysts by contact and transport between contaminated faeces and food. In this way, flies would eventually transmit this parasite. To date, Giardia's presence in the gut of other invertebrates has not been described in the literature. Here we show by first time, using scanning electron microscopy, the presence of Giardia-like trophozoites in the gut of termite Heterotermes tenuis. Two groups of Giardia were found based exclusively on the size and the flange shape of the protozoa: one presented eight flagella, a ventral disc, size, and shape very similar to Giardia intestinalis. In contrast, other cells were smaller and showed some differences in the external morphology. We cannot exclude the possibility that they correspond to the same species and that these differences result from protozoan heterogeneity.


Asunto(s)
Giardia/aislamiento & purificación , Giardiasis/parasitología , Isópteros/parasitología , Animales , Brasil , Heces/parasitología , Flagelos/ultraestructura , Giardia/clasificación , Giardia/ultraestructura , Giardiasis/transmisión , Microscopía Electrónica de Rastreo , Orgánulos/ultraestructura , Trofozoítos/citología
9.
Nat Commun ; 11(1): 5520, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139725

RESUMEN

Axonemal dynein ATPases direct ciliary and flagellar beating via adenosine triphosphate (ATP) hydrolysis. The modulatory effect of adenosine monophosphate (AMP) and adenosine diphosphate (ADP) on flagellar beating is not fully understood. Here, we describe a deficiency of cilia and flagella associated protein 45 (CFAP45) in humans and mice that presents a motile ciliopathy featuring situs inversus totalis and asthenospermia. CFAP45-deficient cilia and flagella show normal morphology and axonemal ultrastructure. Proteomic profiling links CFAP45 to an axonemal module including dynein ATPases and adenylate kinase as well as CFAP52, whose mutations cause a similar ciliopathy. CFAP45 binds AMP in vitro, consistent with structural modelling that identifies an AMP-binding interface between CFAP45 and AK8. Microtubule sliding of dyskinetic sperm from Cfap45-/- mice is rescued with the addition of either AMP or ADP with ATP, compared to ATP alone. We propose that CFAP45 supports mammalian ciliary and flagellar beating via an adenine nucleotide homeostasis module.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Astenozoospermia/genética , Proteínas del Citoesqueleto/deficiencia , Situs Inversus/genética , Adolescente , Adulto , Animales , Astenozoospermia/patología , Axonema/ultraestructura , Sistemas CRISPR-Cas/genética , Cilios/metabolismo , Cilios/ultraestructura , Proteínas del Citoesqueleto/genética , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Epidídimo/patología , Femenino , Flagelos/metabolismo , Flagelos/ultraestructura , Humanos , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Planarias/citología , Planarias/genética , Planarias/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/patología , Situs Inversus/diagnóstico por imagen , Situs Inversus/patología , Motilidad Espermática/genética , Tomografía Computarizada por Rayos X , Secuenciación del Exoma
10.
Exp Parasitol ; 219: 108009, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33007296

RESUMEN

Cell-cell interaction and active migration (and invasion) of parasites into skin host-cell(s) are key steps for successful infection by Leishmania. Chemotaxis constitutes a primordial chapter of Leishmania-host cell interaction, potentially modulated by neuropeptides released into the skin due, for example, to the noxious stimuli represented by the insect bite. Herein we have evaluated in vitro the effect of sensory (Substance P, SP) and autonomic (Vasoactive Intestinal Peptide, VIP, and Neuropeptide Y, NPY) neuropeptides on parasite taxis, and investigated the potential modulatory effect of SP on Leishmania (Viannia) braziliensis-macrophage interaction. We demonstrated that VIP (10-10 M) and NPY (10-9 M) are chemorepellent to the parasites, while SP (10-8 M) produces a chemoattractant response. SP did not affect macrophage viability but seems to impair parasite-macrophage interaction as it decreased promastigote adherence to macrophages. As this effect is blocked by ([D-Pro 2, D-Trp7,9]-Substance P (10-6 M), the observed action may be mediated by neurokinin-1 (NK1) transmembrane receptors. VIP and NPY repellent chemotactic effect is impaired by their corresponding receptor antagonists. Additionally, they suggest that SP may be a key molecule to guide promastigote migration towards, and interaction, with dendritic cells and macrophage host cells.


Asunto(s)
Leishmania braziliensis/metabolismo , Neuropéptido Y/metabolismo , Sustancia P/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Animales , Quimiotaxis , Flagelos/ultraestructura , Leishmania braziliensis/fisiología , Leishmania braziliensis/ultraestructura , Macrófagos , Ratones
11.
J Fish Biol ; 97(5): 1491-1506, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32869341

RESUMEN

The aim of this study was to analyse spermatogenesis in the African butterflyfish, Pantodon buchholzi, using transmission electron microscopy and scanning electron microscopy. P. buchholzi is the most basal teleost that exhibits insemination and produces a highly complex introsperm with the most elongate midpiece known in teleost fishes. Their early stages (spermatogonia and spermatocytes) do not differ greatly from those of other fishes, with the exception of Golgi apparatus degradation appearing as spindle-shaped bodies (SSBs). In round, early spermatids, the development of the flagellum begins after the migration of the centriolar complex towards the nucleus. Later, the elongation of the midpiece coincides with the displacement of the mitochondria and their fusion to produce nine mitochondrial derivatives (MDs). In these spermatids, the nucleus is situated laterally to the midpiece, with condensing chromatin in the centre of the nucleus. Within the midpiece, the flagellum is located within a cytoplasmic canal and is surrounded by a cytoplasmic sleeve containing fibres, MDs and a great amount of cytoplasm located on one side. During the next phase, nuclear rotation, the highly condensed chromatin is displaced to a position above the centriolar apparatus, whereas chromatin-free nucleoplasm is transferred to the cytoplasm. Later, this nucleoplasm, still surrounded by the nuclear membrane, is eliminated into the cyst lumen as the nucleoplasmic packet. Within the highly elongate spermatids, other excess organelles (SSBs, endoplasmic reticulum and mitochondria) are eliminated as residual bodies (RBs). Fully developed spermatozoa, which contain conical-shaped nuclei, eventually coalesce to form unencapsulated sperm packets (spermatozeugmata) that are surrounded by RBs at the level of the extremely elongate midpieces. Later, RBs are removed at the periphery of the cyst by means of phagocytosis by Sertoli cells.


Asunto(s)
Peces/fisiología , Espermatogénesis/fisiología , Espermatozoides/ultraestructura , Animales , Núcleo Celular/ultraestructura , Citoplasma/ultraestructura , Flagelos/ultraestructura , Masculino , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura , Espermátides/ultraestructura , Espermatocitos/ultraestructura , Espermatogonias/ultraestructura
12.
J Cell Biol ; 219(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32496561

RESUMEN

Cilia and flagella are microtubule-based cellular projections with important sensory and motility functions. Their absence or malfunction is associated with a growing number of human diseases collectively referred to as ciliopathies. However, the fundamental mechanisms underpinning cilia biogenesis and functions remain only partly understood. Here, we show that depleting LUZP1 or its interacting protein, EPLIN, increases the levels of MyosinVa at the centrosome and primary cilia formation. We further show that LUZP1 localizes to both actin filaments and the centrosome/basal body. Like EPLIN, LUZP1 is an actin-stabilizing protein that regulates actin dynamics, at least in part, by mobilizing ARP2 to the centrosomes. Both LUZP1 and EPLIN interact with known ciliogenesis and cilia-length regulators and as such represent novel players in actin-dependent centrosome to basal body conversion. Ciliogenesis deregulation caused by LUZP1 or EPLIN loss may thus contribute to the pathology of their associated disease states.


Asunto(s)
Actinas/genética , Cilios/metabolismo , Proteínas del Citoesqueleto/genética , Células Epiteliales/metabolismo , Cadenas Pesadas de Miosina/genética , Miosina Tipo V/genética , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/ultraestructura , Proteína 2 Relacionada con la Actina/química , Proteína 2 Relacionada con la Actina/genética , Proteína 2 Relacionada con la Actina/metabolismo , Actinas/química , Actinas/metabolismo , Animales , Cuerpos Basales/metabolismo , Cuerpos Basales/ultraestructura , Línea Celular Tumoral , Centrosoma/metabolismo , Centrosoma/ultraestructura , Cilios/ultraestructura , Ciliopatías/genética , Ciliopatías/metabolismo , Ciliopatías/patología , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Células Epiteliales/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Flagelos/metabolismo , Flagelos/ultraestructura , Expresión Génica , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Cadenas Pesadas de Miosina/química , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo V/química , Miosina Tipo V/metabolismo , Estabilidad Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
J Biol Chem ; 295(24): 8331-8347, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32354742

RESUMEN

Introduced about a century ago, suramin remains a frontline drug for the management of early-stage East African trypanosomiasis (sleeping sickness). Cellular entry into the causative agent, the protozoan parasite Trypanosoma brucei, occurs through receptor-mediated endocytosis involving the parasite's invariant surface glycoprotein 75 (ISG75), followed by transport into the cytosol via a lysosomal transporter. The molecular basis of the trypanocidal activity of suramin remains unclear, but some evidence suggests broad, but specific, impacts on trypanosome metabolism (i.e. polypharmacology). Here we observed that suramin is rapidly accumulated in trypanosome cells proportionally to ISG75 abundance. Although we found little evidence that suramin disrupts glycolytic or glycosomal pathways, we noted increased mitochondrial ATP production, but a net decrease in cellular ATP levels. Metabolomics highlighted additional impacts on mitochondrial metabolism, including partial Krebs' cycle activation and significant accumulation of pyruvate, corroborated by increased expression of mitochondrial enzymes and transporters. Significantly, the vast majority of suramin-induced proteins were normally more abundant in the insect forms compared with the blood stage of the parasite, including several proteins associated with differentiation. We conclude that suramin has multiple and complex effects on trypanosomes, but unexpectedly partially activates mitochondrial ATP-generating activity. We propose that despite apparent compensatory mechanisms in drug-challenged cells, the suramin-induced collapse of cellular ATP ultimately leads to trypanosome cell death.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Mitocondrias/metabolismo , Suramina/farmacología , Trypanosoma brucei brucei/metabolismo , Adenosina Trifosfato/metabolismo , Flagelos/efectos de los fármacos , Flagelos/metabolismo , Flagelos/ultraestructura , Glucólisis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Metaboloma/efectos de los fármacos , Microcuerpos/efectos de los fármacos , Microcuerpos/metabolismo , Microcuerpos/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Modelos Moleculares , Prolina/metabolismo , Proteoma/metabolismo , ATPasas de Translocación de Protón/metabolismo , Proteínas Protozoarias/metabolismo , Ácido Pirúvico/metabolismo
14.
Mol Genet Genomic Med ; 8(7): e1284, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32410354

RESUMEN

BACKGROUND: Acephalic spermatozoa is an extremely rare type of teratozoospermia that is associated with male infertility. Several genes have been reported to be relevant to acephalic spermatozoa. Thus, more genetic pathogenesis needs to be explored. METHODS: Whole-exome sequencing was performed in a patient with acephalic spermatozoa. Then Sanger sequencing was used for validation in the patient and his family. The patient's spermatozoa sample was observed by papanicolaou staining and transmission electron microscopy. Western blot and immunofluorescence were performed to detect the level and localization of related proteins. RESULTS: A novel homozygous frameshift insertion mutation c.545dupT;p.Ala183Serfs*10 in exon 8 of TSGA10 (NM_001349012.1) was identified. Our results showed misarranged mitochondrial sheath and abnormal flagellum in the patient's spermatozoa. TSGA10 failed to be detected in the patient's spermatozoa. However, the expression of SUN5 and PMFBP1 remained unaffected. CONCLUSION: These results suggest that the novel homozygous frameshift insertion mutation of TSGA10 is a cause of acephalic spermatozoa.


Asunto(s)
Proteínas del Citoesqueleto/genética , Mutación con Pérdida de Función , Espermatozoides/ultraestructura , Teratozoospermia/genética , Adulto , Femenino , Flagelos/ultraestructura , Mutación del Sistema de Lectura , Homocigoto , Humanos , Masculino , Mitocondrias/ultraestructura , Linaje , Fenotipo , Teratozoospermia/patología
15.
J Cell Biol ; 219(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32435793

RESUMEN

In the absence of Hedgehog ligand, patched-1 (Ptch1) localizes to cilia and prevents ciliary accumulation and activation of smoothened (Smo). Upon ligand binding, Ptch1 is removed from cilia, and Smo is derepressed and accumulates in cilia where it activates signaling. The mechanisms regulating these dynamic movements are not well understood, but defects in intraflagellar transport components, including Ift27 and the BBSome, cause Smo to accumulate in cilia without pathway activation. We find that in the absence of ligand-induced pathway activation, Smo is ubiquitinated and removed from cilia, and this process is dependent on Ift27 and BBSome components. Activation of Hedgehog signaling decreases Smo ubiquitination and ciliary removal, resulting in its accumulation. Blocking ubiquitination of Smo by an E1 ligase inhibitor or by mutating two lysine residues in intracellular loop three causes Smo to aberrantly accumulate in cilia without pathway activation. These data provide a mechanism to control Smo's ciliary level during Hedgehog signaling by regulating the ubiquitination state of the receptor.


Asunto(s)
Cilios/metabolismo , Flagelos/metabolismo , Proteínas Hedgehog/genética , Procesamiento Proteico-Postraduccional , Transducción de Señal , Receptor Smoothened/genética , Ubiquitina/genética , Animales , Transporte Biológico , Línea Celular Transformada , Cilios/ultraestructura , Embrión de Mamíferos , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Flagelos/ultraestructura , Proteínas Hedgehog/metabolismo , Ratones , Modelos Moleculares , Receptor Patched-1/genética , Receptor Patched-1/metabolismo , Estructura Secundaria de Proteína , Proteínas/genética , Proteínas/metabolismo , Receptor Smoothened/química , Receptor Smoothened/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Ubiquitina/metabolismo , Ubiquitinación , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo
16.
Int J Mol Sci ; 21(8)2020 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-32325779

RESUMEN

In eukaryotic cilia and flagella, various types of axonemal dyneins orchestrate their distinct functions to generate oscillatory bending of axonemes. The force-generating mechanism of dyneins has recently been well elucidated, mainly in cytoplasmic dyneins, thanks to progress in single-molecule measurements, X-ray crystallography, and advanced electron microscopy. These techniques have shed light on several important questions concerning what conformational changes accompany ATP hydrolysis and whether multiple motor domains are coordinated in the movements of dynein. However, due to the lack of a proper expression system for axonemal dyneins, no atomic coordinates of the entire motor domain of axonemal dynein have been reported. Therefore, a substantial amount of knowledge on the molecular architecture of axonemal dynein has been derived from electron microscopic observations on dynein arms in axonemes or on isolated axonemal dynein molecules. This review describes our current knowledge and perspectives of the force-generating mechanism of axonemal dyneins in solo and in ensemble.


Asunto(s)
Adenosina Trifosfato/metabolismo , Dineínas Axonemales/química , Flagelos/metabolismo , Microtúbulos/metabolismo , Animales , Dineínas Axonemales/metabolismo , Dineínas Axonemales/ultraestructura , Axonema/química , Axonema/metabolismo , Cilios/metabolismo , Cristalografía por Rayos X , Dineínas Citoplasmáticas/metabolismo , Flagelos/ultraestructura
17.
J Microbiol ; 58(4): 245-251, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31760612

RESUMEN

The bacterial flagellum is an appendage structure that provides a means for motility to promote survival in fluctuating environments. For the intracellular pathogen Salmonella enterica serovar Typhimurium to survive within macrophages, flagellar gene expression must be tightly regulated, and thus, is controlled at multiple levels, including DNA recombination, transcription, post-transcription, protein synthesis, and assembly within host cells. To understand the contribution of flagella to Salmonella pathogenesis within the host, it is critical to detect flagella production within macrophages via microscopy. In this paper, we describe two methods for detecting bacterial flagella by microscopy both in vitro and in vivo infection models.


Asunto(s)
Flagelos/ultraestructura , Salmonella typhimurium/ultraestructura , Animales , Línea Celular , Macrófagos/microbiología , Ratones , Microscopía Electrónica de Transmisión , Infecciones por Salmonella/microbiología , Salmonella typhimurium/patogenicidad
18.
Proc Natl Acad Sci U S A ; 116(46): 23152-23162, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31659045

RESUMEN

The nexin-dynein regulatory complex (N-DRC) in motile cilia and flagella functions as a linker between neighboring doublet microtubules, acts to stabilize the axonemal core structure, and serves as a central hub for the regulation of ciliary motility. Although the N-DRC has been studied extensively using genetic, biochemical, and structural approaches, the precise arrangement of the 11 (or more) N-DRC subunits remains unknown. Here, using cryo-electron tomography, we have compared the structure of Chlamydomonas wild-type flagella to that of strains with specific DRC subunit deletions or rescued strains with tagged DRC subunits. Our results show that DRC7 is a central linker subunit that helps connect the N-DRC to the outer dynein arms. DRC11 is required for the assembly of DRC8, and DRC8/11 form a subcomplex in the proximal lobe of the linker domain that is required to form stable contacts to the neighboring B-tubule. Gold labeling of tagged subunits determines the precise locations of the previously ambiguous N terminus of DRC4 and C terminus of DRC5. DRC4 is now shown to contribute to the core scaffold of the N-DRC. Our results reveal the overall architecture of N-DRC, with the 3 subunits DRC1/2/4 forming a core complex that serves as the scaffold for the assembly of the "functional subunits," namely DRC3/5-8/11. These findings shed light on N-DRC assembly and its role in regulating flagellar beating.


Asunto(s)
Chlamydomonas/metabolismo , Dineínas/metabolismo , Flagelos/ultraestructura , Proteínas Asociadas a Microtúbulos/metabolismo , Chlamydomonas/genética , Chlamydomonas/ultraestructura , Estructura Cuaternaria de Proteína
19.
Mol Biol Cell ; 30(21): 2659-2680, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31483737

RESUMEN

Ciliary motility depends on both the precise spatial organization of multiple dynein motors within the 96 nm axonemal repeat and the highly coordinated interactions between different dyneins and regulatory complexes located at the base of the radial spokes. Mutations in genes encoding cytoplasmic assembly factors, intraflagellar transport factors, docking proteins, dynein subunits, and associated regulatory proteins can all lead to defects in dynein assembly and ciliary motility. Significant progress has been made in the identification of dynein subunits and extrinsic factors required for preassembly of dynein complexes in the cytoplasm, but less is known about the docking factors that specify the unique binding sites for the different dynein isoforms on the surface of the doublet microtubules. We have used insertional mutagenesis to identify a new locus, IDA8/BOP2, required for targeting the assembly of a subset of inner dynein arms (IDAs) to a specific location in the 96 nm repeat. IDA8 encodes flagellar-associated polypeptide (FAP)57/WDR65, a highly conserved WD repeat, coiled coil domain protein. Using high resolution proteomic and structural approaches, we find that FAP57 forms a discrete complex. Cryo-electron tomography coupled with epitope tagging and gold labeling reveal that FAP57 forms an extended structure that interconnects multiple IDAs and regulatory complexes.


Asunto(s)
Proteínas Algáceas/metabolismo , Axonema/metabolismo , Cilios/metabolismo , Dineínas/metabolismo , Flagelos/metabolismo , Proteómica/métodos , Proteínas Algáceas/genética , Secuencia de Aminoácidos , Axonema/genética , Chlamydomonas reinhardtii/genética , Chlamydomonas reinhardtii/metabolismo , Cilios/genética , Cilios/ultraestructura , Microscopía por Crioelectrón/métodos , Dineínas/genética , Tomografía con Microscopio Electrónico , Flagelos/genética , Flagelos/ultraestructura , Microscopía Fluorescente/métodos , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Mutación , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Grabación de Cinta de Video/métodos
20.
J Cell Sci ; 132(14)2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31217284

RESUMEN

Trypanosoma brucei possesses a motile flagellum that determines cell morphology and the cell division plane. Inheritance of the newly assembled flagellum during the cell cycle is controlled by the Polo-like kinase homolog TbPLK, which also regulates cytokinesis initiation. How TbPLK is targeted to its subcellular locations remains poorly understood. Here we report the trypanosome-specific protein BOH1 that cooperates with TbPLK to regulate flagellum inheritance and cytokinesis initiation in the procyclic form of T. brucei BOH1 localizes to an unusual sub-domain in the flagellum-associated hook complex, bridging the hook complex, the centrin arm and the flagellum attachment zone. Depletion of BOH1 disrupts hook-complex morphology, inhibits centrin-arm elongation and abolishes flagellum attachment zone assembly, leading to flagellum mis-positioning and detachment. Further, BOH1 deficiency impairs the localization of TbPLK and the cytokinesis regulator CIF1 to the cytokinesis initiation site, providing a molecular mechanism for its role in cytokinesis initiation. These findings reveal the roles of BOH1 in maintaining hook-complex morphology and regulating flagellum inheritance, and establish BOH1 as an upstream regulator of the TbPLK-mediated cytokinesis regulatory pathway.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Citocinesis , Flagelos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Protozoarias/metabolismo , Trypanosoma brucei brucei/metabolismo , Flagelos/ultraestructura , Técnicas de Silenciamiento del Gen , Unión Proteica , Dominios Proteicos , Proteínas Protozoarias/química , Trypanosoma brucei brucei/ultraestructura , Quinasa Tipo Polo 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA