Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 169(7): 1214-1227.e18, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28622508

RESUMEN

Higher eukaryotic chromosomes are organized into topologically constrained functional domains; however, the molecular mechanisms required to sustain these complex interphase chromatin structures are unknown. A stable matrix underpinning nuclear organization was hypothesized, but the idea was abandoned as more dynamic models of chromatin behavior became prevalent. Here, we report that scaffold attachment factor A (SAF-A), originally identified as a structural nuclear protein, interacts with chromatin-associated RNAs (caRNAs) via its RGG domain to regulate human interphase chromatin structures in a transcription-dependent manner. Mechanistically, this is dependent on SAF-A's AAA+ ATPase domain, which mediates cycles of protein oligomerization with caRNAs, in response to ATP binding and hydrolysis. SAF-A oligomerization decompacts large-scale chromatin structure while SAF-A loss or monomerization promotes aberrant chromosome folding and accumulation of genome damage. Our results show that SAF-A and caRNAs form a dynamic, transcriptionally responsive chromatin mesh that organizes large-scale chromosome structures and protects the genome from instability.


Asunto(s)
Cromosomas/metabolismo , Inestabilidad Genómica , Ribonucleoproteína Heterogénea-Nuclear Grupo U/metabolismo , ARN Nuclear Pequeño/metabolismo , Secuencia de Aminoácidos , Cromatina , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo U/química , Humanos , Interfase , Modelos Moleculares , Alineación de Secuencia , Transcripción Genética
2.
Immunity ; 47(4): 739-751.e5, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29045903

RESUMEN

Infection by helminth parasites is associated with amelioration of allergic reactivity, but mechanistic insights into this association are lacking. Products secreted by the mouse parasite Heligmosomoides polygyrus suppress type 2 (allergic) immune responses through interference in the interleukin-33 (IL-33) pathway. Here, we identified H. polygyrus Alarmin Release Inhibitor (HpARI), an IL-33-suppressive 26-kDa protein, containing three predicted complement control protein (CCP) modules. In vivo, recombinant HpARI abrogated IL-33, group 2 innate lymphoid cell (ILC2) and eosinophilic responses to Alternaria allergen administration, and diminished eosinophilic responses to Nippostrongylus brasiliensis, increasing parasite burden. HpARI bound directly to both mouse and human IL-33 (in the cytokine's activated state) and also to nuclear DNA via its N-terminal CCP module pair (CCP1/2), tethering active IL-33 within necrotic cells, preventing its release, and forestalling initiation of type 2 allergic responses. Thus, HpARI employs a novel molecular strategy to suppress type 2 immunity in both infection and allergy.


Asunto(s)
Proteínas del Helminto/inmunología , Interleucina-33/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Alérgenos/inmunología , Alternaria/inmunología , Secuencia de Aminoácidos , Animales , Western Blotting , Eosinófilos/inmunología , Proteínas del Helminto/genética , Proteínas del Helminto/metabolismo , Interacciones Huésped-Parásitos/inmunología , Humanos , Inmunidad Innata/inmunología , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33/genética , Interleucina-33/metabolismo , Linfocitos/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Nematospiroides dubius/genética , Nematospiroides dubius/metabolismo , Unión Proteica/inmunología , Receptores de Interleucina/inmunología , Receptores de Interleucina/metabolismo , Homología de Secuencia de Aminoácido , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología
3.
Genes Dev ; 30(19): 2173-2186, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27737961

RESUMEN

Chromosomal instability is a hallmark of cancer, but mitotic regulators are rarely mutated in tumors. Mutations in the condensin complexes, which restructure chromosomes to facilitate segregation during mitosis, are significantly enriched in cancer genomes, but experimental evidence implicating condensin dysfunction in tumorigenesis is lacking. We report that mice inheriting missense mutations in a condensin II subunit (Caph2nes) develop T-cell lymphoma. Before tumors develop, we found that the same Caph2 mutation impairs ploidy maintenance to a different extent in different hematopoietic cell types, with ploidy most severely perturbed at the CD4+CD8+ T-cell stage from which tumors initiate. Premalignant CD4+CD8+ T cells show persistent catenations during chromosome segregation, triggering DNA damage in diploid daughter cells and elevated ploidy. Genome sequencing revealed that Caph2 single-mutant tumors are near diploid but carry deletions spanning tumor suppressor genes, whereas P53 inactivation allowed Caph2 mutant cells with whole-chromosome gains and structural rearrangements to form highly aggressive disease. Together, our data challenge the view that mitotic chromosome formation is an invariant process during development and provide evidence that defective mitotic chromosome structure can promote tumorigenesis.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Unión al ADN/genética , Inestabilidad Genómica/genética , Linfoma de Células T/genética , Complejos Multiproteicos/genética , Mutación Missense/genética , Neoplasias del Timo/genética , Adenosina Trifosfatasas/metabolismo , Anafase , Animales , Células Cultivadas , Estructuras Cromosómicas/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Linfoma de Células T/fisiopatología , Masculino , Metafase , Ratones , Complejos Multiproteicos/metabolismo , Timocitos/patología , Neoplasias del Timo/fisiopatología
4.
Am J Hum Genet ; 100(5): 706-724, 2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28413018

RESUMEN

During neurotransmission, synaptic vesicles undergo multiple rounds of exo-endocytosis, involving recycling and/or degradation of synaptic proteins. While ubiquitin signaling at synapses is essential for neural function, it has been assumed that synaptic proteostasis requires the ubiquitin-proteasome system (UPS). We demonstrate here that turnover of synaptic membrane proteins via the endolysosomal pathway is essential for synaptic function. In both human and mouse, hypomorphic mutations in the ubiquitin adaptor protein PLAA cause an infantile-lethal neurodysfunction syndrome with seizures. Resulting from perturbed endolysosomal degradation, Plaa mutant neurons accumulate K63-polyubiquitylated proteins and synaptic membrane proteins, disrupting synaptic vesicle recycling and neurotransmission. Through characterization of this neurological intracellular trafficking disorder, we establish the importance of ubiquitin-mediated endolysosomal trafficking at the synapse.


Asunto(s)
Epilepsia/genética , Proteínas/genética , Espasmos Infantiles/genética , Transmisión Sináptica , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Modelos Animales de Enfermedad , Epilepsia/diagnóstico , Fibroblastos/metabolismo , Técnicas de Genotipaje , Humanos , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Ratones , Ratones Transgénicos , Mutación , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Conformación Proteica , Proteínas/metabolismo , Células de Purkinje/metabolismo , Espasmos Infantiles/diagnóstico , Vesículas Sinápticas/metabolismo , Transcriptoma , Ubiquitina/genética , Ubiquitina/metabolismo
5.
J Cell Sci ; 131(4)2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29361551

RESUMEN

Retinitis pigmentosa 2 (RP2) is the causative gene for a form of X-linked retinal degeneration. RP2 was previously shown to have GTPase-activating protein (GAP) activity towards the small GTPase ARL3 via its N-terminus, but the function of the C-terminus remains elusive. Here, we report a novel interaction between RP2 and osteoclast-stimulating factor 1 (OSTF1), an intracellular protein that indirectly enhances osteoclast formation and activity and is a negative regulator of cell motility. Moreover, this interaction is abolished by a human pathogenic mutation in RP2. We utilized a structure-based approach to pinpoint the binding interface to a strictly conserved cluster of residues on the surface of RP2 that spans both the C- and N-terminal domains of the protein, and which is structurally distinct from the ARL3-binding site. In addition, we show that RP2 is a positive regulator of cell motility in vitro, recruiting OSTF1 to the cell membrane and preventing its interaction with the migration regulator Myo1E.


Asunto(s)
Factores de Ribosilacion-ADP/genética , Actinas/genética , Proteínas del Ojo/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Proteínas/genética , Retinitis Pigmentosa/genética , Factores de Ribosilacion-ADP/química , Actinas/química , Sitios de Unión/genética , Línea Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Movimiento Celular/genética , Cilios/genética , Cilios/metabolismo , Proteínas del Ojo/química , Proteínas de Unión al GTP , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas de la Membrana/química , Simulación del Acoplamiento Molecular , Miosina Tipo I/química , Miosina Tipo I/genética , Unión Proteica/genética , Conformación Proteica , Dominios Proteicos/genética , Estructura Terciaria de Proteína , Proteínas/química , Retina/metabolismo , Retina/patología , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología
6.
J Biol Chem ; 292(16): 6468-6477, 2017 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-28249940

RESUMEN

Disrupted in Schizophrenia 1 (DISC1) is a scaffolding protein of significant importance for neurodevelopment and a prominent candidate protein in the pathology of major mental illness. DISC1 modulates a number of critical neuronal signaling pathways through protein-protein interactions; however, the mechanism by which this occurs and how DISC1 causes mental illness is unclear, partly because knowledge of the structure of DISC1 is lacking. A lack of homology with known proteins has hindered attempts to define its domain composition. Here, we employed the high-throughput Expression of Soluble Proteins by Random Incremental Truncation (ESPRIT) technique to identify discretely folded regions of human DISC1 via solubility assessment of tens of thousands of fragments of recombinant DISC1. We identified four novel structured regions, named D, I, S, and C, at amino acids 257-383, 539-655, 635-738, and 691-836, respectively. One region (D) is located in a DISC1 section previously predicted to be unstructured. All regions encompass coiled-coil or α-helical structures, and three are involved in DISC1 oligomerization. Crucially, three of these domains would be lost or disrupted by a chromosomal translocation event after amino acid 597, which has been strongly linked to major mental illness. Furthermore, we observed that a known illness-related frameshift mutation after amino acid 807 causes the C region to form aberrantly multimeric and aggregated complexes with an unstable secondary structure. This newly revealed domain architecture of DISC1, therefore, provides a powerful framework for understanding the critical role of this protein in a variety of devastating mental illnesses.


Asunto(s)
Mutación , Proteínas del Tejido Nervioso/química , Trastornos Psicóticos/genética , Esquizofrenia/genética , Mutación del Sistema de Lectura , Humanos , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Desnaturalización Proteica , Dominios Proteicos , Pliegue de Proteína , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Transducción de Señal
7.
J Biol Chem ; 292(15): 6225-6239, 2017 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-28209709

RESUMEN

Mutations in retinitis pigmentosa 2 (RP2) account for 10-20% of X-linked retinitis pigmentosa (RP) cases. The encoded RP2 protein is implicated in ciliary trafficking of myristoylated and prenylated proteins in photoreceptor cells. To date >70 mutations in RP2 have been identified. How these mutations disrupt the function of RP2 is not fully understood. Here we report a novel in-frame 12-bp deletion (c.357_368del, p.Pro120_Gly123del) in zebrafish rp2 The mutant zebrafish shows reduced rod phototransduction proteins and progressive retinal degeneration. Interestingly, the protein level of mutant Rp2 is almost undetectable, whereas its mRNA level is near normal, indicating a possible post-translational effect of the mutation. Consistent with this hypothesis, the equivalent 12-bp deletion in human RP2 markedly impairs RP2 protein stability and reduces its protein level. Furthermore, we found that a majority of the RP2 pathogenic mutations (including missense, single-residue deletion, and C-terminal truncation mutations) severely destabilize the RP2 protein. The destabilized RP2 mutant proteins are degraded via the proteasome pathway, resulting in dramatically decreased protein levels. The remaining non-destabilizing mutations T87I, R118H/R118G/R118L/R118C, E138G, and R211H/R211L are suggested to impair the interaction between RP2 and its protein partners (such as ARL3) or with as yet unknown partners. By utilizing a combination of in silico, in vitro, and in vivo approaches, our work comprehensively indicates that loss of RP2 protein structural stability is the predominating pathogenic consequence for most RP2 mutations. Our study also reveals a role of the C-terminal domain of RP2 in maintaining the overall protein stability.


Asunto(s)
Secuencia de Bases , Proteínas del Ojo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Degeneración Retiniana , Eliminación de Secuencia , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Proteínas del Ojo/genética , Proteínas de Unión al GTP , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Dominios Proteicos , Estabilidad Proteica , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Pez Cebra/genética , Proteínas de Pez Cebra/genética
8.
Hum Mutat ; 38(8): 942-946, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28493397

RESUMEN

Ocular coloboma (OC) is a defect in optic fissure closure and is a common cause of severe congenital visual impairment. Bilateral OC is primarily genetically determined and shows marked locus heterogeneity. Whole-exome sequencing (WES) was used to analyze 12 trios (child affected with OC and both unaffected parents). This identified de novo mutations in 10 different genes in eight probands. Three of these genes encoded proteins associated with actin cytoskeleton dynamics: ACTG1, TWF1, and LCP1. Proband-only WES identified a second unrelated individual with isolated OC carrying the same ACTG1 allele, encoding p.(Pro70Leu). Both individuals have normal neurodevelopment with no extra-ocular signs of Baraitser-Winter syndrome. We found this mutant protein to be incapable of incorporation into F-actin. The LCP1 and TWF1 variants each resulted in only minor disturbance of actin interactions, and no further plausibly causative variants were identified in these genes on resequencing 380 unrelated individuals with OC.


Asunto(s)
Actinas/genética , Coloboma/etiología , Coloboma/genética , Animales , Femenino , Humanos , Masculino , Ratones , Proteínas de Microfilamentos/genética , Mutación/genética , Proteínas Tirosina Quinasas/genética
9.
Hum Mol Genet ; 24(11): 3286-95, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25701875

RESUMEN

Paget's disease of bone (PDB) is a common disorder with a strong genetic component characterized by increased but disorganized bone remodelling. Previous genome-wide association studies identified a locus on chromosome 14q32 tagged by rs10498635 which was significantly associated with susceptibility to PDB in several European populations. Here we conducted fine-mapping and targeted sequencing of the candidate locus to identify possible functional variants. Imputation in 741 PDB patients and 2699 controls confirmed that the association was confined to a 60 kb region in the RIN3 gene and conditional analysis adjusting for rs10498635 identified no new independent signals. Sequencing of the RIN3 gene identified a common missense variant (p.R279C) that was strongly associated with the disease (OR = 0.64; P = 1.4 × 10(-9)), and was in strong linkage disequilibrium with rs10498635. A further 13 rare missense variants were identified, seven of which were novel and detected only in PDB cases. When combined, these rare variants were over-represented in cases compared with controls (OR = 3.72; P = 8.9 × 10(-10)). Most rare variants were located in a region that encodes a proline-rich, intrinsically disordered domain of the protein and many were predicted to be pathogenic. RIN3 was expressed in bone tissue and its expression level was ∼10-fold higher in osteoclasts compared with osteoblasts. We conclude that susceptibility to PDB at the 14q32 locus is mediated by a combination of common and rare coding variants in RIN3 and suggest that RIN3 may contribute to PDB susceptibility by affecting osteoclast function.


Asunto(s)
Proteínas Portadoras/genética , Cromosomas Humanos Par 14/genética , Factores de Intercambio de Guanina Nucleótido/genética , Osteítis Deformante/genética , Animales , Secuencia de Bases , Proteínas Portadoras/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Análisis Mutacional de ADN , Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Desequilibrio de Ligamiento , Ratones , Mutación Missense , Osteítis Deformante/metabolismo , Osteítis Deformante/patología , Osteoclastos/metabolismo , Polimorfismo de Nucleótido Simple
10.
Am J Hum Genet ; 94(6): 915-23, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24906020

RESUMEN

We identified four different missense mutations in the single-exon gene MAB21L2 in eight individuals with bilateral eye malformations from five unrelated families via three independent exome sequencing projects. Three mutational events altered the same amino acid (Arg51), and two were identical de novo mutations (c.151C>T [p.Arg51Cys]) in unrelated children with bilateral anophthalmia, intellectual disability, and rhizomelic skeletal dysplasia. c.152G>A (p.Arg51His) segregated with autosomal-dominant bilateral colobomatous microphthalmia in a large multiplex family. The fourth heterozygous mutation (c.145G>A [p.Glu49Lys]) affected an amino acid within two residues of Arg51 in an adult male with bilateral colobomata. In a fifth family, a homozygous mutation (c.740G>A [p.Arg247Gln]) altering a different region of the protein was identified in two male siblings with bilateral retinal colobomata. In mouse embryos, Mab21l2 showed strong expression in the developing eye, pharyngeal arches, and limb bud. As predicted by structural homology, wild-type MAB21L2 bound single-stranded RNA, whereas this activity was lost in all altered forms of the protein. MAB21L2 had no detectable nucleotidyltransferase activity in vitro, and its function remains unknown. Induced expression of wild-type MAB21L2 in human embryonic kidney 293 cells increased phospho-ERK (pERK1/2) signaling. Compared to the wild-type and p.Arg247Gln proteins, the proteins with the Glu49 and Arg51 variants had increased stability. Abnormal persistence of pERK1/2 signaling in MAB21L2-expressing cells during development is a plausible pathogenic mechanism for the heterozygous mutations. The phenotype associated with the homozygous mutation might be a consequence of complete loss of MAB21L2 RNA binding, although the cellular function of this interaction remains unknown.


Asunto(s)
Anoftalmos/genética , Proteínas del Ojo/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación Missense , Adulto , Alelos , Animales , Encefalopatías Metabólicas Innatas/genética , Coloboma/genética , Opacidad de la Córnea/genética , Exoma , Proteínas del Ojo/metabolismo , Femenino , Expresión Génica , Células HEK293 , Heterocigoto , Homocigoto , Humanos , Discapacidad Intelectual/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Microcefalia/genética , Microftalmía/genética , Linaje , Fenotipo , Conformación Proteica , Transducción de Señal
11.
PLoS Pathog ; 11(12): e1005289, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26624286

RESUMEN

Reverse transcription is the central defining feature of HIV-1 replication. We previously reported that the cellular eukaryotic elongation factor 1 (eEF1) complex associates with the HIV-1 reverse transcription complex (RTC) and the association is important for late steps of reverse transcription. Here we show that association between the eEF1 and RTC complexes occurs by a strong and direct interaction between the subunit eEF1A and reverse transcriptase (RT). Using biolayer interferometry and co-immunoprecipitation (co-IP) assays, we show that association between the eEF1 and RTC complexes occurs by a strong (KD ~3-4 nM) and direct interaction between eEF1A and reverse transcriptase (RT). Biolayer interferometry analysis of cell lysates with titrated levels of eEF1A indicates it is a predominant cellular RT binding protein. Both the RT thumb and connection domains are required for interaction with eEF1A. A single amino acid mutation, W252A, within the thumb domain impaired co-IP between eEF1A and RT, and also significantly reduced the efficiency of late reverse transcription and virus replication when incorporated into infectious HIV-1. Molecular modeling analysis indicated that interaction between W252 and L303 are important for RT structure, and their mutation to alanine did not impair heterodimerisation, but negatively impacted interaction with eEF1A. Didemnin B, which specifically binds eEF1A, potently inhibited HIV-1 reverse transcription by greater than 2 logs at subnanomolar concentrations, especially affecting reverse transcription late DNA synthesis. Analysis showed reduced levels of RTCs from HIV-1-infected HEK293T treated with didemnin B compared to untreated cells. Interestingly, HIV-1 with a W252A RT mutation was resistant to didemnin B negative effects showing that didemnin B affects HIV-1 by targeting the RT-eEF1A interaction. The combined evidence indicates a direct interaction between eEF1A and RT is crucial for HIV reverse transcription and replication, and the RT-eEF1A interaction is a potential drug target.


Asunto(s)
Infecciones por VIH/metabolismo , Transcriptasa Inversa del VIH/metabolismo , VIH-1/fisiología , Factor 1 de Elongación Peptídica/metabolismo , Transcripción Reversa/fisiología , Replicación Viral/fisiología , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Inmunoprecipitación
12.
Hum Mol Genet ; 23(4): 906-19, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24092329

RESUMEN

Disrupted-In-Schizophrenia 1 (DISC1) is a candidate risk factor for schizophrenia, bipolar disorder and severe recurrent depression. Here, we demonstrate that DISC1 associates robustly with trafficking-protein-Kinesin-binding-1 which is, in turn, known to interact with the outer mitochondrial membrane proteins Miro1/2, linking mitochondria to the kinesin motor for microtubule-based subcellular trafficking. DISC1 also associates with Miro1 and is thus a component of functional mitochondrial transport complexes. Consistent with these observations, in neuronal axons DISC1 promotes specifically anterograde mitochondrial transport. DISC1 thus participates directly in mitochondrial trafficking, which is essential for neural development and neurotransmission. Any factor affecting mitochondrial DISC1 function is hence likely to have deleterious consequences for the brain, potentially contributing to increased risk of psychiatric illness. Intriguingly, therefore, a rare putatively causal human DISC1 sequence variant, 37W, impairs the ability of DISC1 to promote anterograde mitochondrial transport. This is likely related to a number of mitochondrial abnormalities induced by expression of DISC1-37W, which redistributes mitochondrial DISC1 and enhances kinesin mitochondrial association, while also altering protein interactions within the mitochondrial transport complex.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Axones/metabolismo , Células COS , Chlorocebus aethiops , Células HEK293 , Hipocampo/patología , Humanos , Cinesinas/metabolismo , Trastornos Mentales/metabolismo , Dinámicas Mitocondriales , Mutación Missense , Proteínas del Tejido Nervioso/genética , Transporte de Proteínas
13.
Exp Eye Res ; 138: 32-41, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26093275

RESUMEN

Mammalian photoreceptors contain specialised connecting cilia that connect the inner (IS) to the outer segments (OS). Dysfunction of the connecting cilia due to mutations in ciliary proteins are a common cause of the inherited retinal dystrophy retinitis pigmentosa (RP). Mutations affecting the Retinitis Pigmentosa GTPase Regulator (RPGR) protein is one such cause, affecting 10-20% of all people with RP and the majority of those with X-linked RP. RPGR is located in photoreceptor connecting cilia. It interacts with a wide variety of ciliary proteins, but its exact function is unknown. Recently, there have been important advances both in our understanding of RPGR function and towards the development of a therapy. This review summarises the existing literature on human RPGR function and dysfunction, and suggests that RPGR plays a role in the function of the ciliary gate, which controls access of both membrane and soluble proteins to the photoreceptor outer segment. We discuss key models used to investigate and treat RPGR disease and suggest that gene augmentation therapy offers a realistic therapeutic approach, although important questions still remain to be answered, while cell replacement therapy based on retinal progenitor cells represents a more distant prospect.


Asunto(s)
Proteínas del Ojo/fisiología , Terapia Genética , Células Fotorreceptoras de Vertebrados/fisiología , Retinitis Pigmentosa/fisiopatología , Retinitis Pigmentosa/terapia , Animales , Modelos Animales de Enfermedad , Humanos , Cilio Conector de los Fotorreceptores/fisiología
14.
Hum Mol Genet ; 21(15): 3374-86, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22547224

RESUMEN

Disrupted-In-Schizophrenia 1 (DISC1) was identified as a risk factor for psychiatric illness through its disruption by a balanced chromosomal translocation, t(1;11)(q42.1;q14.3), that co-segregates with schizophrenia, bipolar disorder and depression. We previously reported that the translocation reduces DISC1 expression, consistent with a haploinsufficiency disease model. Here we report that, in lymphoblastoid cell lines, the translocation additionally results in the production of abnormal transcripts due to the fusion of DISC1 with a disrupted gene on chromosome 11 (DISC1FP1/Boymaw). These chimeric transcripts encode abnormal proteins, designated CP1, CP60 and CP69, consisting of DISC1 amino acids 1-597 plus 1, 60 or 69 amino acids, respectively. The novel 69 amino acids in CP69 induce increased α-helical content and formation of large stable protein assemblies. The same is predicted for CP60. Both CP60 and CP69 exhibit profoundly altered functional properties within cell lines and neurons. Both are predominantly targeted to mitochondria, where they induce clustering and loss of membrane potential, indicative of severe mitochondrial dysfunction. There is currently no access to neural material from translocation carriers to confirm these findings, but there is no reason to suppose that these chimeric transcripts will not also be expressed in the brain. There is thus potential for the production of abnormal chimeric proteins in the brains of translocation carriers, although at substantially lower levels than for native DISC1. The mechanism by which inheritance of the translocation increases risk of psychiatric illness may therefore involve both DISC1 haploinsufficiency and mitochondrial deficiency due to the effects of abnormal chimeric protein expression. GenBank accession numbers: DISC1FP1 (EU302123), Boymaw (GU134617), der 11 chimeric transcript DISC1FP1 exon 2 to DISC1 exon 9 (JQ650115), der 1 chimeric transcript DISC1 exon 4 to DISC1FP1 exon 4 (JQ650116), der 1 chimeric transcript DISC1 exon 6 to DISC1FP1 exon 3a (JQ650117).


Asunto(s)
Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 1/genética , Trastornos del Humor/genética , Proteínas del Tejido Nervioso/genética , Esquizofrenia/genética , Translocación Genética , Animales , Células COS , Chlorocebus aethiops , Haploinsuficiencia , Humanos , Proteínas Mitocondriales/genética , Proteínas del Tejido Nervioso/química , Transfección
15.
Gut ; 62(5): 695-707, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22684479

RESUMEN

OBJECTIVE: Mutations in the nucleotide-binding oligomerisation domain-containing protein 2 (NOD2) gene remain the strongest genetic determinants for Crohn's disease (CD). Having previously identified vimentin as a novel NOD2-interacting protein, the authors aimed to investigate the regulatory effects of vimentin on NOD2 function and the association of variants in Vim with CD susceptibility. DESIGN: Coimmunoprecipitation, fluorescent microscopy and fractionation were used to confirm the interaction between NOD2 and vimentin. HEK293 cells stably expressing wild-type NOD2 or a NOD2 frameshift variant (L1007fs) and SW480 colonic epithelial cells were used alongside the vimentin inhibitor, withaferin A (WFA), to assess effects on NOD2 function using the nuclear factor-kappaB (NF-κB) reporter gene, green fluorescent protein-LC3-based autophagy, and bacterial gentamicin protection assays. International genome-wide association meta-analysis data were used to test for associations of single-nucleotide polymorphisms in Vim with CD susceptibility. RESULTS: The leucine-rich repeat domain of NOD2 contained the elements required for vimentin binding; CD-associated polymorphisms disrupted this interaction. NOD2 and vimentin colocalised at the cell plasma membrane, and cytosolic mislocalisation of the L1007fs and R702W variants correlated with an inability to interact with vimentin. Use of WFA demonstrated that vimentin was required for NOD2-dependent NF-κB activation and muramyl dipeptide-induced autophagy induction, and that NOD2 and vimentin regulated the invasion and survival properties of a CD-associated adherent-invasive Escherichia coli strain. Genetic analysis revealed an association signal across the haplotype block containing Vim. CONCLUSION: Vimentin is an important regulator of NOD2 function and a potential novel therapeutic target in the treatment of CD. In addition, Vim is a candidate susceptibility gene for CD, supporting the functional data.


Asunto(s)
Enfermedad de Crohn/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Vimentina/metabolismo , Colon/metabolismo , Colon/patología , Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Enfermedad de Crohn/fisiopatología , Susceptibilidad a Enfermedades , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Mutación del Sistema de Lectura , Células HEK293 , Humanos , Inmunoprecipitación , Microscopía Fluorescente , Proteína Adaptadora de Señalización NOD2/genética , Polimorfismo de Nucleótido Simple , Vimentina/genética , Witanólidos/farmacología
16.
J Biol Chem ; 287(39): 32381-93, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22843697

RESUMEN

Paralogs NDE1 (nuclear distribution element 1) and NDEL1 (NDE-like 1) are essential for mitosis and neurodevelopment. Both proteins are predicted to have similar structures, based upon high sequence similarity, and they co-complex in mammalian cells. X-ray diffraction studies and homology modeling suggest that their N-terminal regions (residues 8-167) adopt continuous, extended α-helical coiled-coil structures, but no experimentally derived information on the structure of their C-terminal regions or the architecture of the full-length proteins is available. In the case of NDE1, no biophysical data exists. Here we characterize the structural architecture of both full-length proteins utilizing negative stain electron microscopy along with our established paradigm of chemical cross-linking followed by tryptic digestion, mass spectrometry, and database searching, which we enhance using isotope labeling for mixed NDE1-NDEL1. We determined that full-length NDE1 forms needle-like dimers and tetramers in solution, similar to crystal structures of NDEL1, as well as chain-like end-to-end polymers. The C-terminal domain of each protein, required for interaction with key protein partners dynein and DISC1 (disrupted-in-schizophrenia 1), includes a predicted disordered region that allows a bent back structure. This facilitates interaction of the C-terminal region with the N-terminal coiled-coil domain and is in agreement with previous results showing N- and C-terminal regions of NDEL1 and NDE1 cooperating in dynein interaction. It sheds light on recently identified mutations in the NDE1 gene that cause truncation of the encoded protein. Additionally, analysis of mixed NDE1-NDEL1 complexes demonstrates that NDE1 and NDEL1 can interact directly.


Asunto(s)
Proteínas Portadoras/química , Proteínas Asociadas a Microtúbulos/química , Modelos Moleculares , Pliegue de Proteína , Multimerización de Proteína/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
17.
J Neurosci ; 31(24): 9043-54, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21677187

RESUMEN

Nuclear distribution factor E-homolog 1 (NDE1), Lissencephaly 1 (LIS1), and NDE-like 1 (NDEL1) together participate in essential neurodevelopmental processes, including neuronal precursor proliferation and differentiation, neuronal migration, and neurite outgrowth. NDE1/LIS1/NDEL1 interacts with Disrupted in Schizophrenia 1 (DISC1) and the cAMP-hydrolyzing enzyme phosphodiesterase 4 (PDE4). DISC1, PDE4, NDE1, and NDEL1 have each been implicated as genetic risk factors for major mental illness. Here, we demonstrate that DISC1 and PDE4 modulate NDE1 phosphorylation by cAMP-dependent protein kinase A (PKA) and identify a novel PKA substrate site on NDE1 at threonine-131 (T131). Homology modeling predicts that phosphorylation at T131 modulates NDE1-LIS1 and NDE1-NDEL1 interactions, which we confirm experimentally. DISC1-PDE4 interaction thus modulates organization of the NDE1/NDEL1/LIS1 complex. T131-phosphorylated NDE1 is present at the postsynaptic density, in proximal axons, within the nucleus, and at the centrosome where it becomes substantially enriched during mitosis. Mutation of the NDE1 T131 site to mimic PKA phosphorylation inhibits neurite outgrowth. Thus PKA-dependent phosphorylation of the NDE1/LIS1/NDEL1 complex is DISC1-PDE4 modulated and likely to regulate its neural functions.


Asunto(s)
1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Humanos , Inmunoprecipitación/métodos , Proteínas Asociadas a Microtúbulos/genética , Modelos Moleculares , Mutación/genética , Proteínas del Tejido Nervioso/genética , Neuritas/fisiología , Fosforilación/fisiología , Densidad Postsináptica/metabolismo , Treonina/genética , Treonina/metabolismo , Transfección/métodos
18.
Am J Hum Genet ; 85(6): 833-46, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19944402

RESUMEN

Schizophrenia and bipolar disorder are leading causes of morbidity across all populations, with heritability estimates of approximately 80% indicating a substantial genetic component. Population genetics and genome-wide association studies suggest an overlap of genetic risk factors between these illnesses but it is unclear how this genetic component is divided between common gene polymorphisms, rare genomic copy number variants, and rare gene sequence mutations. We report evidence that the lipid transporter gene ABCA13 is a susceptibility factor for both schizophrenia and bipolar disorder. After the initial discovery of its disruption by a chromosome abnormality in a person with schizophrenia, we resequenced ABCA13 exons in 100 cases with schizophrenia and 100 controls. Multiple rare coding variants were identified including one nonsense and nine missense mutations and compound heterozygosity/homozygosity in six cases. Variants were genotyped in additional schizophrenia, bipolar, depression (n > 1600), and control (n > 950) cohorts and the frequency of all rare variants combined was greater than controls in schizophrenia (OR = 1.93, p = 0.0057) and bipolar disorder (OR = 2.71, p = 0.00007). The population attributable risk of these mutations was 2.2% for schizophrenia and 4.0% for bipolar disorder. In a study of 21 families of mutation carriers, we genotyped affected and unaffected relatives and found significant linkage (LOD = 4.3) of rare variants with a phenotype including schizophrenia, bipolar disorder, and major depression. These data identify a candidate gene, highlight the genetic overlap between schizophrenia, bipolar disorder, and depression, and suggest that rare coding variants may contribute significantly to risk of these disorders.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/fisiología , Predisposición Genética a la Enfermedad , Polimorfismo Genético , Secuencia de Aminoácidos , Trastorno Bipolar/genética , Estudios de Casos y Controles , Codón sin Sentido , Citogenética , Análisis Mutacional de ADN , Depresión/genética , Exones , Femenino , Ligamiento Genético , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación Missense , Esquizofrenia/genética , Homología de Secuencia de Aminoácido
19.
Nucleic Acids Res ; 38(5): 1723-37, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20007596

RESUMEN

Plasmids, conjugative transposons and phage frequently encode anti-restriction proteins to enhance their chances of entering a new bacterial host that is highly likely to contain a Type I DNA restriction and modification (RM) system. The RM system usually destroys the invading DNA. Some of the anti-restriction proteins are DNA mimics and bind to the RM enzyme to prevent it binding to DNA. In this article, we characterize ArdB anti-restriction proteins and their close homologues, the KlcA proteins from a range of mobile genetic elements; including an ArdB encoded on a pathogenicity island from uropathogenic Escherichia coli and a KlcA from an IncP-1b plasmid, pBP136 isolated from Bordetella pertussis. We show that all the ArdB and KlcA act as anti-restriction proteins and inhibit the four main families of Type I RM systems in vivo, but fail to block the restriction endonuclease activity of the archetypal Type I RM enzyme, EcoKI, in vitro indicating that the action of ArdB is indirect and very different from that of the DNA mimics. We also present the structure determined by NMR spectroscopy of the pBP136 KlcA protein. The structure shows a novel protein fold and it is clearly not a DNA structural mimic.


Asunto(s)
Proteínas Bacterianas/química , Desoxirribonucleasas de Localización Especificada Tipo I/antagonistas & inhibidores , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bordetella pertussis/química , Enzimas de Restricción del ADN/metabolismo , Desoxirribonucleasas de Localización Especificada Tipo I/metabolismo , Endopeptidasa Clp/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Pliegue de Proteína , Homología de Secuencia de Aminoácido , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/metabolismo
20.
Physiol Genomics ; 42(3): 319-30, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20571110

RESUMEN

11beta-Hydroxysteroid dehydrogenase type 2 (11betaHSD2) is a short-chain dehydrogenase/reductase (SDR) responsible for inactivating cortisol and preventing its binding to the mineralocorticoid receptor (MR). Nonfunctional mutations in HSD11B2, the gene encoding 11betaHSD2, cause the hypertensive syndrome of apparent mineralocorticoid excess (AME). Like other such Mendelian disorders, AME is rare but has nevertheless helped to illuminate principles fundamental to the regulation of blood pressure. Furthermore, polymorphisms in HSD11B2 have been associated with salt sensitivity, a major risk factor for cardiovascular mortality. It is therefore highly likely that sequence variation in HSD11B2, having subtle functional ramifications, will affect blood pressure in the wider population. In this study, a three-dimensional homology model of 11betaHSD2 was created and used to hypothesize the functional consequences in terms of protein structure of published mutations in HSD11B2. This approach underscored the strong genotype-phenotype correlation of AME: severe forms of the disease, associated with little in vivo enzyme activity, arise from mutations occurring in invariant alignment positions. These were predicted to exert gross structural changes in the protein. In contrast, those mutations causing a mild clinical phenotype were in less conserved regions of the protein that were predicted to be relatively more tolerant to substitution. Finally, a number of pathogenic mutations are shown to be associated with regions predicted to participate in dimer formation, and in protein stabilization, which may therefore suggest molecular mechanisms of disease.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/fisiología , Biología Computacional , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/química , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Secuencia de Aminoácidos , Animales , Biología Computacional/métodos , Análisis Mutacional de ADN/métodos , Bases de Datos Genéticas , Familia , Predisposición Genética a la Enfermedad , Humanos , Hipertensión/genética , Síndrome de Exceso Aparente de Mineralocorticoides/genética , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Polimorfismo Genético , Conformación Proteica , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA