Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 156(5): 1017-31, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24581499

RESUMEN

The spindle assembly checkpoint (SAC) delays anaphase until all chromosomes are bioriented on the mitotic spindle. Under current models, unattached kinetochores transduce the SAC by catalyzing the intramitotic production of a diffusible inhibitor of APC/C(Cdc20) (the anaphase-promoting complex/cyclosome and its coactivator Cdc20, a large ubiquitin ligase). Here we show that nuclear pore complexes (NPCs) in interphase cells also function as scaffolds for anaphase-inhibitory signaling. This role is mediated by Mad1-Mad2 complexes tethered to the nuclear basket, which activate soluble Mad2 as a binding partner and inhibitor of Cdc20 in the cytoplasm. Displacing Mad1-Mad2 from nuclear pores accelerated anaphase onset, prevented effective correction of merotelic errors, and increased the threshold of kinetochore-dependent signaling needed to halt mitosis in response to spindle poisons. A heterologous Mad1-NPC tether restored Cdc20 inhibitor production and normal M phase control. We conclude that nuclear pores and kinetochores both emit "wait anaphase" signals that preserve genome integrity.


Asunto(s)
Anafase , Proteínas de Ciclo Celular/metabolismo , Puntos de Control de la Fase M del Ciclo Celular , Proteínas Mad2/metabolismo , Poro Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Transporte Activo de Núcleo Celular , Proteínas de Ciclo Celular/genética , Dimerización , Células HCT116 , Células HeLa , Humanos , Interfase , Cinetocoros/metabolismo , Mitosis , Proteínas Nucleares/genética
2.
Proc Natl Acad Sci U S A ; 120(1): e2213846120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36574677

RESUMEN

Piezo1 and 2 are evolutionarily conserved mechanosensory cation channels known to function on the cell surface by responding to external pressure and transducing a mechanically activated Ca2+ current. Here we show that both Piezo1 and 2 also exhibit concentrated intracellular localization at centrosomes. Both Piezo1 and 2 loss-of-function and Piezo1 activation by the small molecule Yoda1 result in supernumerary centrosomes, premature centriole disengagement, multi-polar spindles, and mitotic delay. By using a GFP, Calmodulin and M13 Protein fusion (GCaMP) Ca2+-sensitive reporter, we show that perturbations in Piezo modulate Ca2+ flux at centrosomes. Moreover, the inhibition of Polo-like-kinase 1 eliminates Yoda1-induced centriole disengagement. Because previous studies have implicated force generation by microtubules as essential for maintaining centrosomal integrity, we propose that mechanotransduction by Piezo maintains pericentrosomal Ca2+ within a defined range, possibly through sensing cell intrinsic forces from microtubules.


Asunto(s)
Centrosoma , Mecanotransducción Celular , Centrosoma/metabolismo , Centriolos , Microtúbulos
3.
Proc Natl Acad Sci U S A ; 113(5): 1267-72, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26764383

RESUMEN

Chemotaxis, the directional migration of cells in a chemical gradient, is robust to fluctuations associated with low chemical concentrations and dynamically changing gradients as well as high saturating chemical concentrations. Although a number of reports have identified cellular behavior consistent with a directional memory that could account for behavior in these complex environments, the quantitative and molecular details of such a memory process remain unknown. Using microfluidics to confine cellular motion to a 1D channel and control chemoattractant exposure, we observed directional memory in chemotactic neutrophil-like cells. We modeled this directional memory as a long-lived intracellular asymmetry that decays slower than observed membrane phospholipid signaling. Measurements of intracellular dynamics revealed that moesin at the cell rear is a long-lived element that when inhibited, results in a reduction of memory. Inhibition of ROCK (Rho-associated protein kinase), downstream of RhoA (Ras homolog gene family, member A), stabilized moesin and directional memory while depolymerization of microtubules (MTs) disoriented moesin deposition and also reduced directional memory. Our study reveals that long-lived polarized cytoskeletal structures, specifically moesin, actomyosin, and MTs, provide a directional memory in neutrophil-like cells even as they respond on short time scales to external chemical cues.


Asunto(s)
Polaridad Celular , Quimiotaxis , Citoesqueleto/metabolismo , Memoria Inmunológica , Células HL-60 , Humanos
4.
FASEB J ; 30(3): 1155-70, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26644349

RESUMEN

Group IVA phospholipase A2 [cytosolic phospholipase A2α (cPLA2α)] is a key mediator of inflammation and tumorigenesis. In this study, by using a combination of chemical inhibition and genetic approaches in zebrafish and murine cells, we identify a mechanism by which cPLA2α promotes cell proliferation. We identified 2 cpla2α genes in zebrafish, cpla2αa and cpla2αb, with conserved phospholipase activity. In zebrafish, loss of cpla2α expression or inhibition of cpla2α activity diminished G1 progression through the cell cycle. This phenotype was also seen in both mouse embryonic fibroblasts and mesangial cells. G1 progression was rescued by the addition of arachidonic acid or prostaglandin E2 (PGE2), indicating a phospholipase-dependent mechanism. We further show that PGE2, through PI3K/AKT activation, promoted Forkhead box protein O1 (FOXO1) phosphorylation and FOXO1 nuclear export. This led to up-regulation of cyclin D1 and down-regulation of p27(Kip1), thus promoting G1 progression. Finally, using pharmacologic inhibitors, we show that cPLA2α, rapidly accelerated fibrosarcoma (RAF)/MEK/ERK, and PI3K/AKT signaling pathways cooperatively regulate G1 progression in response to platelet-derived growth factor stimulation. In summary, these data indicate that cPLA2α, through its phospholipase activity, is a critical effector of G1 phase progression through the cell cycle and suggest that pharmacological targeting of this enzyme may have important therapeutic benefits in disease mechanisms that involve excessive cell proliferation, in particular, cancer and proliferative glomerulopathies.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Fase G1/fisiología , Fosfolipasas A2 Grupo IV/metabolismo , Animales , Ácido Araquidónico/farmacología , División Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Dinoprostona/farmacología , Regulación hacia Abajo/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fase G1/efectos de los fármacos , Fosfolipasas A2 Grupo IV/genética , Células HEK293 , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Pez Cebra
5.
Proc Natl Acad Sci U S A ; 111(33): 12085-90, 2014 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-25092318

RESUMEN

Expansions of preexisting polyglutamine (polyQ) tracts in at least nine different proteins cause devastating neurodegenerative diseases. There are many unique features to these pathologies, but there must also be unifying mechanisms underlying polyQ toxicity. Using a polyQ-expanded fragment of huntingtin exon-1 (Htt103Q), the causal protein in Huntington disease, we and others have created tractable models for investigating polyQ toxicity in yeast cells. These models recapitulate key pathological features of human diseases and provide access to an unrivalled genetic toolbox. To identify toxicity modifiers, we performed an unbiased overexpression screen of virtually every protein encoded by the yeast genome. Surprisingly, there was no overlap between our modifiers and those from a conceptually identical screen reported recently, a discrepancy we attribute to an artifact of their overexpression plasmid. The suppressors of Htt103Q toxicity recovered in our screen were strongly enriched for glutamine- and asparagine-rich prion-like proteins. Separated from the rest of the protein, the prion-like sequences of these proteins were themselves potent suppressors of polyQ-expanded huntingtin exon-1 toxicity, in both yeast and human cells. Replacing the glutamines in these sequences with asparagines abolished suppression and converted them to enhancers of toxicity. Replacing asparagines with glutamines created stronger suppressors. The suppressors (but not the enhancers) coaggregated with Htt103Q, forming large foci at the insoluble protein deposit in which proteins were highly immobile. Cells possessing foci had fewer (if any) small diffusible oligomers of Htt103Q. Until such foci were lost, cells were protected from death. We discuss the therapeutic implications of these findings.


Asunto(s)
Exones , Proteínas del Tejido Nervioso/genética , Priones/fisiología , Proteínas Ligadas a GPI/fisiología , Humanos , Proteína Huntingtina , Microscopía Confocal
6.
Am J Hum Genet ; 93(5): 915-25, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24140113

RESUMEN

Intraflagellar transport (IFT) depends on two evolutionarily conserved modules, subcomplexes A (IFT-A) and B (IFT-B), to drive ciliary assembly and maintenance. All six IFT-A components and their motor protein, DYNC2H1, have been linked to human skeletal ciliopathies, including asphyxiating thoracic dystrophy (ATD; also known as Jeune syndrome), Sensenbrenner syndrome, and Mainzer-Saldino syndrome (MZSDS). Conversely, the 14 subunits in the IFT-B module, with the exception of IFT80, have unknown roles in human disease. To identify additional IFT-B components defective in ciliopathies, we independently performed different mutation analyses: candidate-based sequencing of all IFT-B-encoding genes in 1,467 individuals with a nephronophthisis-related ciliopathy or whole-exome resequencing in 63 individuals with ATD. We thereby detected biallelic mutations in the IFT-B-encoding gene IFT172 in 12 families. All affected individuals displayed abnormalities of the thorax and/or long bones, as well as renal, hepatic, or retinal involvement, consistent with the diagnosis of ATD or MZSDS. Additionally, cerebellar aplasia or hypoplasia characteristic of Joubert syndrome was present in 2 out of 12 families. Fibroblasts from affected individuals showed disturbed ciliary composition, suggesting alteration of ciliary transport and signaling. Knockdown of ift172 in zebrafish recapitulated the human phenotype and demonstrated a genetic interaction between ift172 and ift80. In summary, we have identified defects in IFT172 as a cause of complex ATD and MZSDS. Our findings link the group of skeletal ciliopathies to an additional IFT-B component, IFT172, similar to what has been shown for IFT-A.


Asunto(s)
Ataxia Cerebelosa/genética , Síndrome de Ellis-Van Creveld/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Retinitis Pigmentosa/genética , Alelos , Secuencia de Aminoácidos , Animales , Pueblo Asiatico/genética , Huesos/anomalías , Huesos/metabolismo , Huesos/patología , Ataxia Cerebelosa/patología , Craneosinostosis/genética , Craneosinostosis/patología , Dineínas Citoplasmáticas/genética , Dineínas Citoplasmáticas/metabolismo , Dineínas/genética , Dineínas/metabolismo , Displasia Ectodérmica/genética , Displasia Ectodérmica/patología , Síndrome de Ellis-Van Creveld/patología , Epistasis Genética , Femenino , Fibroblastos/patología , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Masculino , Datos de Secuencia Molecular , Mutación , Fenotipo , Retinitis Pigmentosa/patología , Población Blanca/genética , Pez Cebra/genética
7.
Proc Natl Acad Sci U S A ; 110(38): 15497-501, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-24006363

RESUMEN

Homo-oligomerization is found in many biological systems and has been extensively studied in vitro. However, our ability to quantify and understand oligomerization processes in cells is still limited. We used fluorescence correlation spectroscopy and mathematical modeling to measure the dynamics of the tetramers formed by the tumor suppressor protein p53 in single living cells. Previous in vitro studies suggested that in basal conditions all p53 molecules are bound in dimers. We found that in resting cells p53 is present in a mix of oligomeric states with a large cell-to-cell variation. After DNA damage, p53 molecules in all cells rapidly assemble into tetramers before p53 protein levels increase. We developed a model to understand the connection between p53 accumulation and tetramerization. We found that the rapid increase in p53 tetramers requires a combination of active tetramerization and protein stabilization, however tetramerization alone is sufficient to activate p53 transcriptional targets. This suggests triggering tetramerization as a mechanism for activating the p53 pathway in cancer cells. Many other transcription factors homo-oligomerize, and our approach provides a unique way for probing the dynamics and functional consequences of oligomerization.


Asunto(s)
Daño del ADN , Regulación de la Expresión Génica/fisiología , Modelos Biológicos , Polimerizacion , Proteína p53 Supresora de Tumor/metabolismo , Western Blotting , Cicloheximida , Humanos , Células MCF-7 , Espectrometría de Fluorescencia/métodos , Imagen de Lapso de Tiempo
8.
Proc Natl Acad Sci U S A ; 110(52): 21006-11, 2013 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-24324148

RESUMEN

Cells integrate multiple measurement modalities to navigate their environment. Soluble and substrate-bound chemical gradients and physical cues have all been shown to influence cell orientation and migration. Here we investigate the role of asymmetric hydraulic pressure in directional sensing. Cells confined in microchannels identified and chose a path of lower hydraulic resistance in the absence of chemical cues. In a bifurcating channel with asymmetric hydraulic resistances, this choice was preceded by the elaboration of two leading edges with a faster extension rate along the lower resistance channel. Retraction of the "losing" edge appeared to precipitate a final choice of direction. The pressure differences altering leading edge protrusion rates were small, suggesting weak force generation by leading edges. The response to the physical asymmetry was able to override a dynamically generated chemical cue. Motile cells may use this bias as a result of hydraulic resistance, or "barotaxis," in concert with chemotaxis to navigate complex environments.


Asunto(s)
Movimiento Celular/fisiología , Hidrodinámica , Presión , Línea Celular Tumoral , Quimiotaxis/fisiología , Humanos , Técnicas Analíticas Microfluídicas/métodos , Microscopía Fluorescente , Microesferas
9.
J Biol Chem ; 289(33): 22771-22784, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24982423

RESUMEN

Chromosome ends contain nucleoprotein structures known as telomeres. Damage to chromosome ends during interphase elicits a DNA damage response (DDR) resulting in cell cycle arrest. However, little is known regarding the signaling from damaged chromosome ends (designated here as "TIPs") during mitosis. In the present study, we investigated the consequences of DNA damage induced at a single TIP in mitosis. We used laser microirradiation to damage mitotic TIPs or chromosome arms (non-TIPs) in PtK2 kidney epithelial cells. We found that damage to a single TIP, but not a non-TIP, delays anaphase onset. This TIP-specific checkpoint response is accompanied by differential recruitment of DDR proteins. Although phosphorylation of H2AX and the recruitment of several repair factors, such as Ku70-Ku80, occur in a comparable manner at both TIP and non-TIP damage sites, DDR factors such as ataxia telangiectasia mutated (ATM), MDC1, WRN, and FANCD2 are specifically recruited to TIPs but not to non-TIPs. In addition, Nbs1, BRCA1, and ubiquitin accumulate at damaged TIPs more rapidly than at damaged non-TIPs. ATR and 53BP1 are not detected at either TIPs or non-TIPs in mitosis. The observed delay in anaphase onset is dependent on the activity of DDR kinases ATM and Chk1, and the spindle assembly checkpoint kinase Mps1. Cells damaged at a single TIP or non-TIP eventually exit mitosis with unrepaired lesions. Damaged TIPs are segregated into micronuclei at a significantly higher frequency than damaged non-TIPs. Together, these findings reveal a mitosis-specific DDR uniquely associated with chromosome ends.


Asunto(s)
Anafase , Cromosomas de los Mamíferos/metabolismo , Daño del ADN , Células Epiteliales/metabolismo , Riñón/metabolismo , Rayos Láser/efectos adversos , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Células Epiteliales/citología , Exodesoxirribonucleasas/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Histonas/metabolismo , Riñón/citología , Fosforilación , Potoroidae , Proteínas Quinasas/metabolismo
10.
Cell Mol Life Sci ; 71(11): 2165-78, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24104765

RESUMEN

Primary cilia with a diameter of ~200 nm have been implicated in development and disease. Calcium signaling within a primary cilium has never been directly visualized and has therefore remained a speculation. Fluid-shear stress and dopamine receptor type-5 (DR5) agonist are among the few stimuli that require cilia for intracellular calcium signal transduction. However, it is not known if these stimuli initiate calcium signaling within the cilium or if the calcium signal originates in the cytoplasm. Using an integrated single-cell imaging technique, we demonstrate for the first time that calcium signaling triggered by fluid-shear stress initiates in the primary cilium and can be distinguished from the subsequent cytosolic calcium response through the ryanodine receptor. Importantly, this flow-induced calcium signaling depends on the ciliary polycystin-2 calcium channel. While DR5-specific agonist induces calcium signaling mainly in the cilioplasm via ciliary CaV1.2, thrombin specifically induces cytosolic calcium signaling through the IP3 receptor. Furthermore, a non-specific calcium ionophore triggers both ciliary and cytosolic calcium responses. We suggest that cilia not only act as sensory organelles but also function as calcium signaling compartments. Cilium-dependent signaling can spread to the cytoplasm or be contained within the cilioplasm. Our study thus provides the first model to understand signaling within the cilioplasm of a living cell.


Asunto(s)
Señalización del Calcio , Cilios/metabolismo , Células Epiteliales/metabolismo , Mecanotransducción Celular , Canales Catiónicos TRPP/metabolismo , Animales , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Ionóforos de Calcio/farmacología , Cilios/efectos de los fármacos , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Imagen Molecular , Cultivo Primario de Células , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Reología , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Análisis de la Célula Individual , Estrés Mecánico , Porcinos , Canales Catiónicos TRPP/genética , Trombina/farmacología
11.
Bioconjug Chem ; 25(6): 1043-51, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24797834

RESUMEN

Small molecule fluorophores are indispensable tools for modern biomedical imaging techniques. In this report, we present the development of a new class of BODIPY dyes based on an alkoxy-fluoro-boron-dipyrromethene core. These novel fluorescent dyes, which we term MayaFluors, are characterized by good aqueous solubility and favorable in vitro physicochemical properties. MayaFluors are readily accessible in good yields in a one-pot, two-step approach starting from well-established BODIPY dyes, and allow for facile modification with functional groups of relevance to bioconjugate chemistry and bioorthogonal labeling. Biological profiling in living cells demonstrates excellent membrane permeability, low nonspecific binding, and lack of cytotoxicity.


Asunto(s)
Compuestos de Boro/análisis , Colorantes Fluorescentes/análisis , Imagen Molecular/métodos , Compuestos de Boro/síntesis química , Compuestos de Boro/química , Permeabilidad de la Membrana Celular , Supervivencia Celular , Femenino , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/química , Humanos , Células MCF-7 , Estructura Molecular , Solubilidad , Células Tumorales Cultivadas
12.
J Am Soc Nephrol ; 24(1): 100-12, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23274954

RESUMEN

A missense mutation in mouse Nek8, which encodes a ciliary kinase, produces the juvenile cystic kidneys (jck) model of polycystic kidney disease, but the functions of Nek8 are incompletely understood. Here, we generated a Nek8-null allele and found that homozygous mutant mice die at birth and exhibit randomization of left-right asymmetry, cardiac anomalies, and glomerular kidney cysts. The requirement for Nek8 in left-right patterning is conserved, as knockdown of the zebrafish ortholog caused randomized heart looping. Ciliogenesis was intact in Nek8-deficient embryos and cells, but we observed misexpression of left-sided marker genes early in development, suggesting that nodal ciliary signaling was perturbed. We also generated jck/Nek8 compound heterozygotes; these mutants developed less severe cystic disease than jck homozygotes and provided genetic evidence that the jck allele may encode a gain-of-function protein. Notably, NEK8 and polycystin-2 (PC2) proteins interact, and we found that Nek8(-/-) and Pkd2(-/-) embryonic phenotypes are strikingly similar. Nek8-deficient embryos and cells did express PC2 normally, which localized properly to the cilia. However, similar to cells lacking PC2, NEK8-depleted inner medullary collecting duct cells exhibited a defective response to fluid shear, suggesting that NEK8 may play a role in mediating PC2-dependent signaling.


Asunto(s)
Tipificación del Cuerpo , Cilios/fisiología , Enfermedades Renales Poliquísticas/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Femenino , Cardiopatías Congénitas/embriología , Heterocigoto , Homocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Quinasas Relacionadas con NIMA , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Canales Catiónicos TRPP/metabolismo , Pez Cebra
13.
Chromosoma ; 121(5): 509-25, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22782189

RESUMEN

With the goal of creating two genetically identical daughter cells, cell division culminates in the equal segregation of sister chromatids. This phase of cell division is monitored by a cell cycle checkpoint known as the spindle assembly checkpoint (SAC). The SAC actively prevents chromosome segregation while one or more chromosomes, or more accurately kinetochores, remain unattached to the mitotic spindle. Such unattached kinetochores recruit SAC proteins to assemble a diffusible anaphase inhibitor. Kinetochores stop production of this inhibitor once microtubules (MTs) of the mitotic spindle are bound, but productive attachment of all kinetochores is required to satisfy the SAC, initiate anaphase, and exit from mitosis. Although mechanisms of kinetochore signaling and SAC inhibitor assembly and function have received the bulk of attention in the past two decades, recent work has focused on the principles of SAC silencing. Here, we review the mechanisms that silence SAC signaling at the kinetochore, and in particular, how attachment to spindle MTs and biorientation on the mitotic spindle may turn off inhibitor generation. Future challenges in this area are highlighted towards the goal of building a comprehensive molecular model of this process.


Asunto(s)
Cinetocoros/metabolismo , Puntos de Control de la Fase M del Ciclo Celular , Mitosis , Huso Acromático/metabolismo , Animales , Segregación Cromosómica , Humanos , Microtúbulos/genética , Microtúbulos/metabolismo , Huso Acromático/genética
14.
Hum Mol Genet ; 20(2): 354-67, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21041232

RESUMEN

Autosomal-dominant polycystic kidney disease (ADPKD) is the most common hereditary and systemic disorder associated with various cardiovascular complications. It has been implicated with dysfunction in primary cilia. We and others have shown that the immediate function of endothelial cilia is to sense extracellular signal. The long-term function of cilia is hypothesized to regulate cell cycle. Here, we show that ciliary function (polycystins) and structure (polaris) are required for proper cellular division. Cilia mutant cells undergo abnormal cell division with apparent defects in mitotic spindle formation, cellular spindle assembly checkpoint and centrosome amplification. Down-regulation of the chromosomal passenger survivin contributes to these abnormalities, which further result in cell polyploidy. Re-expression of survivin restores a competent spindle assembly checkpoint and reduces polyploidy. Aged animals show a more severe phenotype in cellular division, consistent with progression of cardiovascular complications seen in older ADPKD patients. For the first time, we show that structure and function of mechanosensory cilia are crucial in maintaining proper cellular proliferation. Furthermore, developmental aging plays a crucial role in the progression of these abnormal cellular phenotypes. We propose that abnormal function or structure of primary cilia not only causes failure to transmit extracellular signals, but also is associated with cytokinesis defects in both mice and humans with polycystic kidney disease.


Asunto(s)
Segregación Cromosómica , Regulación hacia Abajo , Células Endoteliales/patología , Proteínas Inhibidoras de la Apoptosis/genética , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/fisiopatología , Poliploidía , Proteínas Represoras/genética , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Proliferación Celular , Centrosoma/metabolismo , Cilios/genética , Cilios/metabolismo , Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica , Inestabilidad Genómica , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Cariotipificación , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Huso Acromático/patología , Survivin , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
EMBO J ; 28(15): 2162-73, 2009 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-19629044

RESUMEN

The idle assembly checkpoint acts to delay chromosome segregation until all duplicated sister chromatids are captured by the mitotic spindle. This pathway ensures that each daughter cell receives a complete copy of the genome. The high fidelity and robustness of this process have made it a subject of intense study in both the experimental and computational realms. A significant number of checkpoint proteins have been identified but how they orchestrate the communication between local spindle attachment and global cytoplasmic signalling to delay segregation is not yet understood. Here, we propose a systems view of the spindle assembly checkpoint to focus attention on the key regulators of the dynamics of this pathway. These regulators in turn have been the subject of detailed cellular measurements and computational modelling to connect molecular function to the dynamics of spindle assembly checkpoint signalling. A review of these efforts reveals the insights provided by such approaches and underscores the need for further interdisciplinary studies to reveal in full the quantitative underpinnings of this cellular control pathway.


Asunto(s)
Ciclo Celular , Segregación Cromosómica , Huso Acromático/fisiología , Proteínas de Ciclo Celular/metabolismo , Modelos Biológicos , Proteínas Nucleares/metabolismo
16.
Hum Mol Genet ; 19(17): 3306-19, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20554582

RESUMEN

Autosomal recessive polycystic kidney disease (ARPKD) is a significant hereditary renal disease occurring in infancy and childhood, which presents with greatly enlarged echogenic kidneys, ultimately leading to renal insufficiency and end-stage renal disease. ARPKD is caused by mutations in a single gene PKHD1, which encodes fibrocystin/polyductin (FPC), a large single transmembrane protein generally known to be on the primary cilium, basal body and plasma membrane. Here, using our newly generated antibody raised against the entire C-terminal intracellular cytoplasmic domain (ICD) of FPC, as well as our previously well-characterized antibody against a peptide of ICD, we report for the first time that at least one isoform of FPC is localized to the centrosome and mitotic spindle of dividing cells in multiple cell lines, including MDCK, mIMCD3, LLC-PK1, HEK293, RCTEC and HFCT cells. Using short-hairpin-mediated RNA interference, we show that the inhibition of FPC function in MDCK and mIMCD3 cells leads to centrosome amplification, chromosome lagging and multipolar spindle formation. Consistent with our in vitro findings, we also observed centrosome amplification in the kidneys from human ARPKD patients. These findings demonstrate a novel function of FPC in centrosome duplication and mitotic spindle assembly during cell division. We propose that mitotic defects due to FPC dysfunction contribute to cystogenesis in ARPKD.


Asunto(s)
Riñón Poliquístico Autosómico Recesivo/metabolismo , Receptores de Superficie Celular/metabolismo , Huso Acromático/metabolismo , Adolescente , Animales , Línea Celular Tumoral , Centrosoma/metabolismo , Niño , Preescolar , Modelos Animales de Enfermedad , Perros , Femenino , Humanos , Lactante , Masculino , Ratones , Riñón Poliquístico Autosómico Recesivo/genética , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Huso Acromático/genética
17.
J Cell Sci ; 123(Pt 10): 1623-33, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20392738

RESUMEN

Error-free chromosome segregation depends on timely activation of the multi-subunit E3 ubiquitin ligase APC/C. Activation of the APC/C initiates chromosome segregation and mitotic exit by targeting critical cell-cycle regulators for destruction. The APC/C is the principle target of the mitotic checkpoint, which prevents segregation while chromosomes are unattached to spindle microtubules. We now report the identification and characterization of APC16, a conserved subunit of the APC/C. APC16 was found in association with tandem-affinity-purified mitotic checkpoint complex protein complexes. APC16 is a bona fide subunit of human APC/C: it is present in APC/C complexes throughout the cell cycle, the phenotype of APC16-depleted cells copies depletion of other APC/C subunits, and APC16 is important for APC/C activity towards mitotic substrates. APC16 sequence homologues can be identified in metazoans, but not fungi, by four conserved primary sequence stretches. We provide evidence that the C. elegans gene K10D2.4 and the D. rerio gene zgc:110659 are functional equivalents of human APC16. Our findings show that APC/C is composed of previously undescribed subunits, and raise the question of why metazoan APC/C is molecularly different from unicellular APC/C.


Asunto(s)
Mitosis , Complejos de Ubiquitina-Proteína Ligasa/genética , Ubiquitina-Proteína Ligasas/genética , Ciclosoma-Complejo Promotor de la Anafase , Animales , Caenorhabditis elegans , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Segregación Cromosómica/genética , Secuencia Conservada/genética , Células HeLa , Humanos , Proteínas Mad2 , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Interferente Pequeño/genética , Proteínas Represoras/metabolismo , Homología de Secuencia de Aminoácido , Huso Acromático/metabolismo , Espectrometría de Masas en Tándem , Complejos de Ubiquitina-Proteína Ligasa/aislamiento & purificación , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
19.
Nat Chem Biol ; 6(5): 359-68, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20383151

RESUMEN

Mps1, a dual-specificity kinase, is required for the proper functioning of the spindle assembly checkpoint and for the maintenance of chromosomal stability. As Mps1 function has been implicated in numerous phases of the cell cycle, the development of a potent, selective small-molecule inhibitor of Mps1 should facilitate dissection of Mps1-related biology. We describe the cellular effects and Mps1 cocrystal structures of new, selective small-molecule inhibitors of Mps1. Consistent with RNAi studies, chemical inhibition of Mps1 leads to defects in Mad1 and Mad2 establishment at unattached kinetochores, decreased Aurora B kinase activity, premature mitotic exit and gross aneuploidy, without any evidence of centrosome duplication defects. However, in U2OS cells having extra centrosomes (an abnormality found in some cancers), Mps1 inhibition increases the frequency of multipolar mitoses. Lastly, Mps1 inhibitor treatment resulted in a decrease in cancer cell viability.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Ciclo Celular , Inhibidores Enzimáticos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Modelos Moleculares , Inhibidores de Proteínas Quinasas/química , Proteínas Tirosina Quinasas
20.
PLoS One ; 17(6): e0268857, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35704642

RESUMEN

Factor quinolinone inhibitors (FQIs), a first-in-class set of small molecule inhibitors targeted to the transcription factor LSF (TFCP2), exhibit promising cancer chemotherapeutic properties. FQI1, the initial lead compound identified, unexpectedly induced a concentration-dependent delay in mitotic progression. Here, we show that FQI1 can rapidly and reversibly lead to mitotic arrest, even when added directly to mitotic cells, implying that FQI1-mediated mitotic defects are not transcriptionally based. Furthermore, treatment with FQIs resulted in a striking, concentration-dependent diminishment of spindle microtubules, accompanied by a concentration-dependent increase in multi-aster formation. Aberrant γ-tubulin localization was also observed. These phenotypes suggest that perturbation of spindle microtubules is the primary event leading to the mitotic delays upon FQI1 treatment. Previously, FQIs were shown to specifically inhibit not only LSF DNA-binding activity, which requires LSF oligomerization to tetramers, but also other specific LSF-protein interactions. Other transcription factors participate in mitosis through non-transcriptional means, and we recently reported that LSF directly binds α-tubulin and is present in purified cellular tubulin preparations. Consistent with a microtubule role for LSF, here we show that LSF enhanced the rate of tubulin polymerization in vitro, and FQI1 inhibited such polymerization. To probe whether the FQI1-mediated spindle abnormalities could result from inhibition of mitotic LSF-protein interactions, mass spectrometry was performed using as bait an inducible, tagged form of LSF that is biotinylated by endogenous enzymes. The global proteomics analysis yielded expected associations for a transcription factor, notably with RNA processing machinery, but also to nontranscriptional components. In particular, and consistent with spindle disruption due to FQI treatment, mitotic, FQI1-sensitive interactions were identified between the biotinylated LSF and microtubule-associated proteins that regulate spindle assembly, positioning, and dynamics, as well as centrosome-associated proteins. Probing the mitotic LSF interactome using small molecule inhibitors therefore supported a non-transcriptional role for LSF in mediating progression through mitosis.


Asunto(s)
Proteínas Asociadas a Microtúbulos , Quinolonas , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitosis , Quinolonas/metabolismo , Quinolonas/farmacología , Huso Acromático/metabolismo , Factores de Transcripción/metabolismo , Tubulina (Proteína)/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA