Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Mol Cytogenet ; 11: 26, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29736186

RESUMEN

BACKGROUND: Ring chromosome instability may influence a patient's phenotype and challenge its interpretation. RESULTS: Here, we report a 4-year-old girl with a compound phenotype. Cytogenetic analysis revealed her karyotype to be 46,XX,r(22). aCGH identified a 180 kb 22q13.32 duplication, a de novo 2.024 Mb subtelomeric 22q13.32-q13.33 deletion, which is associated with Phelan-McDermid syndrome, and a maternal single gene 382-kb TUSC7 deletion of uncertain clinical significance located in the region of the 3q13.31 deletion syndrome. All chromosomal aberrations were confirmed by real-time PCR in lymphocytes and detected in skin fibroblasts. The deletions were also found in the buccal epithelium. According to FISH analysis, 8% and 24% of the patient's lymphocytes and skin fibroblasts, respectively, had monosomy 22. CONCLUSIONS: We believe that a combination of 22q13.32-q13.33 deletion and monosomy 22 in a portion of cells can better define the clinical phenotype of the patient. Importantly, the in vivo presence of monosomic cells indicates ring chromosome instability, which may favor karyotype correction that is significant for the development of chromosomal therapy protocols.

2.
Oncotarget ; 8(47): 82006-82026, 2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-29137240

RESUMEN

The intramembrane proteolytic activities of presenilins (PSEN1/PS1 and PSEN2/PS2) underlie production of ß-amyloid, the key process in Alzheimer's disease (AD). Dysregulation of presenilin-mediated signaling is linked to cancers. Inhibition of the γ-cleavage activities of PSENs that produce Aß, but not the ε-like cleavage activity that release physiologically essential transcription activators, is a potential approach for the development of rational therapies for AD. In order to identify whether different activities of PSEN1 can be dissociated, we designed multiple mutations in the evolutionary conserved sites of PSEN1. We tested them in vitro and in vivo assays and compared their activities with mutant isoforms of presenilin-related intramembrane di-aspartyl protease (IMPAS1 (IMP1)/signal peptide peptidase (SPP)). PSEN1 auto-cleavage was more resistant to the mutation remodeling than the ε-like proteolysis. PSEN1 with a G382A or a P433A mutation in evolutionary invariant sites retains functionally important APP ε- and Notch S3- cleavage activities, but G382A inhibits APP γ-cleavage and Aß production and a P433A elevates Aß. The G382A variant cannot restore the normal cellular ER Ca2+ leak in PSEN1/PSEN2 double knockout cells, but efficiently rescues the loss-of-function (Egl) phenotype of presenilin in C. elegans. We found that, unlike in PSEN1 knockout cells, endoplasmic reticulum (ER) Ca2+ leak is not changed in the absence of IMP1/SPP. IMP1/SPP with the analogous mutations retained efficiency in cleavage of transmembrane substrates and rescued the lethality of Ce-imp-2 knockouts. In summary, our data show that mutations near the active catalytic sites of intramembrane di-aspartyl proteases have different consequences on proteolytic and signaling functions.

3.
Hum Mol Genet ; 25(11): 2295-2313, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27056978

RESUMEN

Polycystic kidney disease (PKD) is a common cause of renal failure with few effective treatments. INPP5E is an inositol polyphosphate 5-phosphatase that dephosphorylates phosphoinositide 3-kinase (PI3K)-generated PI(3,4,5)P3 and is mutated in ciliopathy syndromes. Germline Inpp5e deletion is embryonically lethal, attributed to cilia stability defects, and is associated with polycystic kidneys. However, the molecular mechanisms responsible for PKD development upon Inpp5e loss remain unknown. Here, we show conditional inactivation of Inpp5e in mouse kidney epithelium results in severe PKD and renal failure, associated with a partial reduction in cilia number and hyperactivation of PI3K/Akt and downstream mammalian target of rapamycin complex 1 (mTORC1) signaling. Treatment with an mTORC1 inhibitor improved kidney morphology and function, but did not affect cilia number or length. Therefore, we identify Inpp5e as an essential inhibitor of the PI3K/Akt/mTORC1 signaling axis in renal epithelial cells, and demonstrate a critical role for Inpp5e-dependent mTORC1 regulation in PKD suppression.


Asunto(s)
Riñón/metabolismo , Complejos Multiproteicos/genética , Monoéster Fosfórico Hidrolasas/genética , Enfermedades Renales Poliquísticas/genética , Serina-Treonina Quinasas TOR/genética , Animales , Ciliopatías/tratamiento farmacológico , Ciliopatías/genética , Ciliopatías/patología , Modelos Animales de Enfermedad , Elafina/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Mutación de Línea Germinal , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Enfermedades Renales Poliquísticas/patología , Proteínas Proto-Oncogénicas c-akt/genética , Eliminación de Secuencia , Transducción de Señal/efectos de los fármacos , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
4.
J Cell Sci ; 128(2): 364-72, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25395580

RESUMEN

Mutations in inositol polyphosphate 5-phosphatase E (INPP5E) cause the ciliopathies known as Joubert and MORM syndromes; however, the role of INPP5E in ciliary biology is not well understood. Here, we describe an interaction between INPP5E and AURKA, a centrosomal kinase that regulates mitosis and ciliary disassembly, and we show that this interaction is important for the stability of primary cilia. Furthermore, AURKA phosphorylates INPP5E and thereby increases its 5-phosphatase activity, which in turn promotes transcriptional downregulation of AURKA, partly through an AKT-dependent mechanism. These findings establish the first direct link between AURKA and phosphoinositide signaling and suggest that the function of INPP5E in cilia is at least partly mediated by its interactions with AURKA.


Asunto(s)
Aurora Quinasa A/metabolismo , Cilios/metabolismo , Fosfatidilinositoles/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Aurora Quinasa A/genética , Cerebelo/anomalías , Cerebelo/patología , Cilios/genética , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Regulación de la Expresión Génica , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Mitosis/genética , Mutación , Monoéster Fosfórico Hidrolasas/genética , Mapas de Interacción de Proteínas/genética , Retina/anomalías , Retina/patología , Transducción de Señal
5.
Proc Natl Acad Sci U S A ; 111(35): 12859-64, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25139996

RESUMEN

Mutations inactivating the cilia-localized Pkd1 protein result in autosomal dominant polycystic kidney disease (ADPKD), a serious inherited syndrome affecting ∼ 1 in 500 people, in which accumulation of renal cysts eventually destroys kidney function. Severity of ADPKD varies throughout the population, for reasons thought to involve differences both in intragenic Pkd1 mutations and in modifier alleles. The scaffolding protein NEDD9, commonly dysregulated during cancer progression, interacts with Aurora-A (AURKA) kinase to control ciliary resorption, and with Src and other partners to influence proliferative signaling pathways often activated in ADPKD. We here demonstrate Nedd9 expression is deregulated in human ADPKD and a mouse ADPKD model. Although genetic ablation of Nedd9 does not independently influence cystogenesis, constitutive absence of Nedd9 strongly promotes cyst formation in the tamoxifen-inducible Pkd1fl/fl;Cre/Esr1(+) mouse model of ADPKD. This cystogenic effect is associated with striking morphological defects in the cilia of Pkd1(-/-);Nedd9(-/-) mice, associated with specific loss of ciliary localization of adenylase cyclase III in the doubly mutant genotype. Ciliary phenotypes imply a failure of Aurora-A activation: Compatible with this idea, Pkd1(-/-);Nedd9(-/-) mice had ciliary resorption defects, and treatment of Pkd1(-/-) mice with a clinical Aurora-A kinase inhibitor exacerbated cystogenesis. In addition, activation of the ADPKD-associated signaling effectors Src, Erk, and the mTOR effector S6 was enhanced, and Ca(2+) response to external stimuli was reduced, in Pkd1(-/-);Nedd9(-/-) versus Pkd1(-/-) mice. Together, these results indicated an important modifier action of Nedd9 on ADPKD pathogenesis involving failure to activate Aurora-A.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Señalización del Calcio/fisiología , Riñón/patología , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Canales Catiónicos TRPP/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Aurora Quinasa A/antagonistas & inhibidores , Aurora Quinasa A/metabolismo , Cilios/fisiología , Modelos Animales de Enfermedad , Células Epiteliales/citología , Femenino , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cultivo Primario de Células , Triazoles/farmacología
6.
Mol Biol Cell ; 23(14): 2658-70, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22621899

RESUMEN

The centrosomal Aurora-A kinase (AURKA) regulates mitotic progression, and overexpression and hyperactivation of AURKA commonly promotes genomic instability in many tumors. Although most studies of AURKA focus on its role in mitosis, some recent work identified unexpected nonmitotic activities of AURKA. Among these, a role for basal body-localized AURKA in regulating ciliary disassembly in interphase cells has highlighted a role in regulating cellular responsiveness to growth factors and mechanical cues. The mechanism of AURKA activation involves interactions with multiple partner proteins and is not well understood, particularly in interphase cells. We show here that AURKA activation at the basal body in ciliary disassembly requires interactions with Ca(2+) and calmodulin (CaM) and that Ca(2+)/CaM are important mediators of the ciliary disassembly process. We also show that Ca(2+)/CaM binding is required for AURKA activation in mitosis and that inhibition of CaM activity reduces interaction between AURKA and its activator, NEDD9. Finally, mutated derivatives of AURKA impaired for CaM binding and/or CaM-dependent activation cause defects in mitotic progression, cytokinesis, and ciliary resorption. These results define Ca(2+)/CaM as important regulators of AURKA activation in mitotic and nonmitotic signaling.


Asunto(s)
Calmodulina/metabolismo , Ciclo Celular/fisiología , Cilios/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aurora Quinasa A , Aurora Quinasas , Calcio/metabolismo , Línea Celular , Centrosoma/metabolismo , Citocinesis , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Mutación Puntual , Unión Proteica/genética , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , ARN Interferente Pequeño , Transducción de Señal
7.
J Cell Biol ; 193(6): 1021-32, 2011 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-21670214

RESUMEN

Most studies of Aurora A (AurA) describe it as a mitotic centrosomal kinase. However, we and others have recently identified AurA functions as diverse as control of ciliary resorption, cell differentiation, and cell polarity control in interphase cells. In these activities, AurA is transiently activated by noncanonical signals, including Ca(2+)-dependent calmodulin binding. These and other observations suggested that AurA might be involved in pathological conditions, such as polycystic kidney disease (PKD). In this paper, we show that AurA is abundant in normal kidney tissue but is also abnormally expressed and activated in cells lining PKD-associated renal cysts. PKD arises from mutations in the PKD1 or PKD2 genes, encoding polycystins 1 and 2 (PC1 and PC2). AurA binds, phosphorylates, and reduces the activity of PC2, a Ca(2+)-permeable nonselective cation channel and, thus, limits the amplitude of Ca(2+) release from the endoplasmic reticulum. These and other findings suggest AurA may be a relevant new biomarker or target in the therapy of PKD.


Asunto(s)
Señalización del Calcio/fisiología , Riñón/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Aurora Quinasa A , Aurora Quinasas , Calcio/metabolismo , Células HEK293 , Humanos , Riñón/citología , Riñón/patología , Ratones , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/patología , Proteínas Serina-Treonina Quinasas/genética , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
8.
Nat Commun ; 1: 64, 2010 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-20842194

RESUMEN

Oncogenic hyperactivation of the mitotic kinase Aurora-A (AurA) in cancer is associated with genomic instability. Increasing evidence indicates that AurA also regulates critical processes in normal interphase cells, but the source of such activity has been obscure. We report here that multiple stimuli causing release of Ca(2+) from intracellular endoplasmic reticulum stores rapidly and transiently activate AurA, without requirement for second messengers. This activation is mediated by direct Ca(2+)-dependent calmodulin (CaM) binding to multiple motifs on AurA. On the basis of structure-function analysis and molecular modelling, we map two primary regions of CaM-AurA interaction to unfolded sequences in the AurA N- and C-termini. This unexpected mechanism for AurA activation provides a new context for evaluating the function of AurA and its inhibitors in normal and cancerous cells.


Asunto(s)
Calcio/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Aurora Quinasas , Western Blotting , Calmodulina/metabolismo , Línea Celular , Retículo Endoplásmico/metabolismo , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Inmunoprecipitación , Espectrometría de Masas , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Homología de Secuencia de Aminoácido
9.
Cancer Res ; 69(18): 7198-206, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19738060

RESUMEN

In the past 3 years, altered expression of the HEF1/CAS-L/NEDD9 scaffolding protein has emerged as contributing to cancer metastasis in multiple cancer types. However, whereas some studies have identified elevated NEDD9 expression as prometastatic, other work has suggested a negative role in tumor progression. We here show that the Nedd9-null genetic background significantly limits mammary tumor initiation in the MMTV-polyoma virus middle T genetic model. Action of NEDD9 is tumor cell intrinsic, with immune cell infiltration, stroma, and angiogenesis unaffected. The majority of the late-appearing mammary tumors of MMTV-polyoma virus middle T;Nedd9(-/-) mice are characterized by depressed activation of proteins including AKT, Src, FAK, and extracellular signal-regulated kinase, emphasizing an important role of NEDD9 as a scaffolding protein for these prooncogenic proteins. Analysis of cells derived from primary Nedd9(+/+) and Nedd9(-/-) tumors showed persistently reduced FAK activation, attachment, and migration, consistent with a role for NEDD9 activation of FAK in promoting tumor aggressiveness. This study provides the first in vivo evidence of a role for NEDD9 in breast cancer progression and suggests that NEDD9 expression may provide a biomarker for tumor aggressiveness.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias Mamarias Experimentales/genética , Proteínas/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Movimiento Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Quinasa 1 de Adhesión Focal/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Endogámicos C57BL , Proteínas/metabolismo , Transducción de Señal
10.
Methods Cell Biol ; 94: 137-60, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20362089

RESUMEN

Cilia are microtubule-based structures that protrude from the cell surface and function as sensors for mechanical and chemical environmental cues that regulate cellular differentiation or division. In metazoans, ciliary signaling is important during organismal development and in the homeostasis controls of adult tissues, with receptors for the Hedgehog, platelet derived growth factor (PDGF), Wnt, and other signaling cascades arrayed and active along the ciliary membrane. In normal cells, cilia are dynamically regulated during cell cycle progression: present in G0 and G1 cells, and usually in S/G2 cells, but almost invariably resorbed before mitotic entry, to reappear post-cytokinesis. This periodic resorption and reassembly of cilia, specified by the intrinsic cell cycle the intrinsic cell cycle machinery, influences the susceptibility of cells to the influence of extrinsic signals with cilia-associated receptors. Pathogenic conditions of mammals associated with loss of or defects in ciliary integrity include a number of developmental disorders, cystic syndromes in adults, and some cancers. With the continuing expansion of the list of human diseases associated with ciliary abnormalities, the identification of the cellular mechanisms regulating ciliary growth and disassembly has become a topic of intense research interest. Although these mechanisms are far from being understood, a number of recent studies have begun to identify key regulatory factors that may begin to offer insight into disease pathogenesis and treatment. In this chapter we will discuss the current state of knowledge regarding cell cycle control of ciliary dynamics, and provide general methods that can be applied to investigate cell cycle-dependent ciliary growth and disassembly.


Asunto(s)
Ciclo Celular/fisiología , Cilios , Animales , Proteínas de Ciclo Celular/metabolismo , Fraccionamiento Celular/métodos , Células Cultivadas , Cilios/metabolismo , Cilios/ultraestructura , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Transducción de Señal/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
11.
Cancer Res ; 68(7): 2058-61, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381407

RESUMEN

In mammals, most cell types have primary cilia, protruding structures involved in sensing mechanical and chemical signals from the extracellular environment that act as major communication hubs for signaling controlling cell differentiation and polarity. The list of clinical disorders associated with ciliary dysfunction has expanded from polycystic kidney disease to include many others. Transformed cells commonly lack cilia, but whether this lack is cause or consequence of transformation is not well understood. Here we discuss work addressing recently identified actions of the cancer-promoting proteins Aurora A and HEF1/NEDD9/CAS-L at cilia. Together with older studies, this work suggests that loss of cilia in cancer may contribute to the insensitivity of cancer cells to environmental repressive signals, based in part on derangement of cell cycle checkpoints governed by cilia and centrosomes.


Asunto(s)
Ciclo Celular/fisiología , Cilios/fisiología , Neoplasias/patología , Animales , Cilios/metabolismo , Humanos , Neoplasias/metabolismo
12.
Am J Hum Genet ; 81(1): 32-43, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17564961

RESUMEN

We identified a mutation in the CRYGD gene (P23S) of the gamma-crystallin gene cluster that is associated with a polymorphic congenital cataract that occurs with frequency of approximately 0.3% in a human population. To gain insight into the molecular mechanism of the pathogenesis of gamma-crystallin isoforms, we undertook an evolutionary analysis of the available mammalian and newly obtained primate sequences of the gamma-crystallin genes. The cataract-associated serine at site 23 corresponds to the ancestral state, since it was found in CRYGD of a lower primate and all the surveyed nonprimate mammals. Crystallin proteins include two structurally similar domains, and substitutions in mammalian CRYGD protein at site 23 of the first domain were always associated with substitutions in the structurally reciprocal sites 109 and 136 of the second domain. These data suggest that the cataractogenic effect of serine at site 23 in the N-terminal domain of CRYGD may be compensated indirectly by amino acid changes in a distal domain. We also found that gene conversion was a factor in the evolution of the gamma-crystallin gene cluster throughout different mammalian clades. The high rate of gene conversion observed between the functional CRYGD gene and two primate gamma-crystallin pseudogenes (CRYGEP1 and CRYGFP1) coupled with a surprising finding of apparent negative selection in primate pseudogenes suggest a deleterious impact of recently derived pseudogenes involved in gene conversion in the gamma-crystallin gene cluster.


Asunto(s)
Catarata/genética , Cristalinas/genética , Evolución Molecular , gamma-Cristalinas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cristalinas/química , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Linaje , Filogenia , Conformación Proteica , Análisis de Secuencia de ADN , Serina/química , Serina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA