Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Alzheimers Res Ther ; 15(1): 201, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37968719

RESUMEN

BACKGROUND: Progression of Alzheimer's disease leads to synapse loss, neural network dysfunction and cognitive failure. Accumulation of protein aggregates and brain immune activation have triggering roles in synaptic failure but the neuronal mechanisms underlying synapse loss are unclear. On the neuronal surface, cellular prion protein (PrPC) is known to be a high-affinity binding site for Amyloid-ß oligomers (Aßo). However, PrPC's dependence in knock-in AD models for tau accumulation, transcriptomic alterations and imaging biomarkers is unknown. METHODS: The necessity of PrPC was examined as a function of age in homozygous AppNL-G-F/hMapt double knock-in mice (DKI). Phenotypes of AppNL-G-F/hMapt mice with a deletion of Prnp expression (DKI; Prnp-/-) were compared with DKI mice with intact Prnp, mice with a targeted deletion of Prnp (Prnp-/-), and mice with intact Prnp (WT). Phenotypes examined included behavioral deficits, synapse loss by PET imaging, synapse loss by immunohistology, tau pathology, gliosis, inflammatory markers, and snRNA-seq transcriptomic profiling. RESULTS: By 9 months age, DKI mice showed learning and memory impairment, but DKI; Prnp-/- and Prnp-/- groups were indistinguishable from WT. Synapse loss in DKI brain, measured by [18F]SynVesT-1 SV2A PET or anti-SV2A immunohistology, was prevented by Prnp deletion. Accumulation of Tau phosphorylated at aa 217 and 202/205, C1q tagging of synapses, and dystrophic neurites were all increased in DKI mice but each decreased to WT levels with Prnp deletion. In contrast, astrogliosis, microgliosis and Aß levels were unchanged between DKI and DKI; Prnp-/- groups. Single-nuclei transcriptomics revealed differential expression in neurons and glia of DKI mice relative to WT. For DKI; Prnp-/- mice, the majority of neuronal genes differentially expressed in DKI mice were no longer significantly altered relative to WT, but most glial DKI-dependent gene expression changes persisted. The DKI-dependent neuronal genes corrected by Prnp deletion associated bioinformatically with synaptic function. Additional genes were uniquely altered only in the Prnp-/- or the DKI; Prnp-/- groups. CONCLUSIONS: Thus, PrPC-dependent synapse loss, phospho-tau accumulation and neuronal gene expression in AD mice can be reversed without clearing Aß plaque or preventing gliotic reaction. This supports targeting the Aßo-PrPC interaction to prevent Aßo-neurotoxicity and pathologic tau accumulation in AD.


Asunto(s)
Enfermedad de Alzheimer , Priones , Ratones , Animales , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Proteínas Priónicas/genética , Transcriptoma , Péptidos beta-Amiloides/metabolismo , Ratones Transgénicos , Priones/metabolismo , Sinapsis/patología , Neuronas/metabolismo , Modelos Animales de Enfermedad , Proteínas tau/genética , Proteínas tau/metabolismo
2.
medRxiv ; 2023 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-36865305

RESUMEN

Alzheimer's disease, the most common age-related neurodegenerative disease, is closely associated with both amyloid-ß plaque and neuroinflammation. Two thirds of Alzheimer's disease patients are females and they have a higher disease risk. Moreover, women with Alzheimer's disease have more extensive brain histological changes than men along with more severe cognitive symptoms and neurodegeneration. To identify how sex difference induces structural brain changes, we performed unbiased massively parallel single nucleus RNA sequencing on Alzheimer's disease and control brains focusing on the middle temporal gyrus, a brain region strongly affected by the disease but not previously studied with these methods. We identified a subpopulation of selectively vulnerable layer 2/3 excitatory neurons that that were RORB-negative and CDH9-expressing. This vulnerability differs from that reported for other brain regions, but there was no detectable difference between male and female patterns in middle temporal gyrus samples. Disease-associated, but sex-independent, reactive astrocyte signatures were also present. In clear contrast, the microglia signatures of diseased brains differed between males and females. Combining single cell transcriptomic data with results from genome-wide association studies (GWAS), we identified MERTK genetic variation as a risk factor for Alzheimer's disease selectively in females. Taken together, our single cell dataset revealed a unique cellular-level view of sex-specific transcriptional changes in Alzheimer's disease, illuminating GWAS identification of sex-specific Alzheimer's risk genes. These data serve as a rich resource for interrogation of the molecular and cellular basis of Alzheimer's disease.

3.
Front Neurol ; 14: 1045644, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36846134

RESUMEN

Introduction: Synapse loss is one of the hallmarks of Alzheimer's disease (AD) and is associated with cognitive decline. In this study, we tested [18F]SDM-16, a novel metabolically stable SV2A PET imaging probe, in the transgenic APPswe/PS1dE9 (APP/PS1) mouse model of AD and age-matched wild-type (WT) mice at 12 months of age. Methods: Based on previous preclinical PET imaging studies using [11C]UCB-J and [18F]SynVesT-1 in the same strain animals, we used the simplified reference tissue model (SRTM), with brain stem as the pseudo reference region to calculate distribution volume ratios (DVRs). Results: To simplify and streamline the quantitative analysis, we compared the standardized uptake value ratios (SUVRs) from different imaging windows to DVRs and found that the averaged SUVRs from 60-90 min post-injection (p.i.) are most consistent with the DVRs. Thus, we used averaged SUVRs from 60-90 min for group comparisons and found statistically significant differences in the tracer uptake in different brain regions, e.g., hippocampus (p = 0.001), striatum (p = 0.002), thalamus (p = 0.003), and cingulate cortex (p = 0.0003). Conclusions: In conclusion, [18F]SDM-16 was used to detect decreased SV2A levels in the brain of APP/PS1 AD mouse model at one year old. Our data suggest that [18F]SDM-16 has similar statistical power in detecting the synapse loss in APP/PS1 mice as [11C]UCB-J and [18F]SynVesT-1, albeit later imaging window (60-90 min p.i.) is needed when SUVR is used as a surrogate for DVR for [18F]SDM-16 due to its slower brain kinetics.

4.
Science ; 377(6614): eabo7257, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36007006

RESUMEN

The granular dorsolateral prefrontal cortex (dlPFC) is an evolutionary specialization of primates that is centrally involved in cognition. We assessed more than 600,000 single-nucleus transcriptomes from adult human, chimpanzee, macaque, and marmoset dlPFC. Although most cell subtypes defined transcriptomically are conserved, we detected several that exist only in a subset of species as well as substantial species-specific molecular differences across homologous neuronal, glial, and non-neural subtypes. The latter are exemplified by human-specific switching between expression of the neuropeptide somatostatin and tyrosine hydroxylase, the rate-limiting enzyme in dopamine production in certain interneurons. The above molecular differences are also illustrated by expression of the neuropsychiatric risk gene FOXP2, which is human-specific in microglia and primate-specific in layer 4 granular neurons. We generated a comprehensive survey of the dlPFC cellular repertoire and its shared and divergent features in anthropoid primates.


Asunto(s)
Corteza Prefontal Dorsolateral , Evolución Molecular , Primates , Somatostatina , Tirosina 3-Monooxigenasa , Adulto , Animales , Dopamina/metabolismo , Corteza Prefontal Dorsolateral/citología , Corteza Prefontal Dorsolateral/metabolismo , Humanos , Pan troglodytes , Primates/genética , Análisis de la Célula Individual , Somatostatina/genética , Somatostatina/metabolismo , Transcriptoma , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
5.
Sci Transl Med ; 14(647): eabi8593, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35648810

RESUMEN

Microglia-mediated synaptic loss contributes to the development of cognitive impairments in Alzheimer's disease (AD). However, the basis for this immune-mediated attack on synapses remains to be elucidated. Treatment with the metabotropic glutamate receptor 5 (mGluR5) silent allosteric modulator (SAM), BMS-984923, prevents ß-amyloid oligomer-induced aberrant synaptic signaling while preserving physiological glutamate response. Here, we show that oral BMS-984923 effectively occupies brain mGluR5 sites visualized by [18F]FPEB positron emission tomography (PET) at doses shown to be safe in rodents and nonhuman primates. In aged mouse models of AD (APPswe/PS1ΔE9 overexpressing transgenic and AppNL-G-F/hMapt double knock-in), SAM treatment fully restored synaptic density as measured by [18F]SynVesT-1 PET for SV2A and by histology, and the therapeutic benefit persisted after drug washout. Phospho-TAU accumulation in double knock-in mice was also reduced by SAM treatment. Single-nuclei transcriptomics demonstrated that SAM treatment in both models normalized expression patterns to a far greater extent in neurons than glia. Last, treatment prevented synaptic localization of the complement component C1Q and synaptic engulfment in AD mice. Thus, selective modulation of mGluR5 reversed neuronal gene expression changes to protect synapses from damage by microglial mediators in rodents.


Asunto(s)
Enfermedad de Alzheimer , Receptor del Glutamato Metabotropico 5 , Enfermedad de Alzheimer/patología , Animales , Complemento C1q/metabolismo , Complemento C1q/uso terapéutico , Modelos Animales de Enfermedad , Ratones , Receptor del Glutamato Metabotropico 5/metabolismo , Receptor del Glutamato Metabotropico 5/uso terapéutico , Sinapsis/metabolismo
6.
Neuron ; 110(3): 452-469.e14, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-34798047

RESUMEN

The hippocampal-entorhinal system supports cognitive functions, has lifelong neurogenic capabilities in many species, and is selectively vulnerable to Alzheimer's disease. To investigate neurogenic potential and cellular diversity, we profiled single-nucleus transcriptomes in five hippocampal-entorhinal subregions in humans, macaques, and pigs. Integrated cross-species analysis revealed robust transcriptomic and histologic signatures of neurogenesis in the adult mouse, pig, and macaque but not humans. Doublecortin (DCX), a widely accepted marker of newly generated granule cells, was detected in diverse human neurons, but it did not define immature neuron populations. To explore species differences in cellular diversity and implications for disease, we characterized subregion-specific, transcriptomically defined cell types and transitional changes from the three-layered archicortex to the six-layered neocortex. Notably, METTL7B defined subregion-specific excitatory neurons and astrocytes in primates, associated with endoplasmic reticulum and lipid droplet proteins, including Alzheimer's disease-related proteins. This resource reveals cell-type- and species-specific properties shaping hippocampal-entorhinal neurogenesis and function.


Asunto(s)
Macaca , Transcriptoma , Animales , Proteína Doblecortina , Hipocampo/patología , Humanos , Ratones , Neurogénesis/genética , Porcinos
7.
Nat Commun ; 12(1): 6906, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34824275

RESUMEN

Astrocytes play critical roles after brain injury, but their precise function is poorly defined. Utilizing single-nuclei transcriptomics to characterize astrocytes after ischemic stroke in the visual cortex of the marmoset monkey, we observed nearly complete segregation between stroke and control astrocyte clusters. Screening for the top 30 differentially expressed genes that might limit stroke recovery, we discovered that a majority of astrocytes expressed RTN4A/ NogoA, a neurite-outgrowth inhibitory protein previously only associated with oligodendrocytes. NogoA upregulation on reactive astrocytes post-stroke was significant in both the marmoset and human brain, whereas only a marginal change was observed in mice. We determined that NogoA mediated an anti-inflammatory response which likely contributes to limiting the infiltration of peripheral macrophages into the surviving parenchyma.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Macrófagos/metabolismo , Proteínas Nogo/metabolismo , Animales , Callithrix , Femenino , Proteína GAP-43 , Glicoproteínas de Membrana , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Proteínas Nogo/genética , Oligodendroglía , Receptores Inmunológicos , Núcleo Solitario , Accidente Cerebrovascular , Transcriptoma , Regulación hacia Arriba , Corteza Visual
9.
Mol Imaging Biol ; 23(3): 372-381, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33258040

RESUMEN

PURPOSE: Synapse loss is a hallmark of Alzheimer's disease (AD) and correlates with cognitive decline. The validation of a noninvasive in vivo imaging approach to quantify synapse would greatly facilitate our understanding of AD pathogenesis and assist drug developments for AD. As animal models of neurodegenerative and neuropsychiatric disorders play a critical role in the drug discovery and development process, a robust, objective, and translational method for quantifying therapeutic drug efficacy in animal models will facilitate the drug development process. In this study, we tested the quantification reliability of the SV2A PET tracer, [18F]SynVesT-1, in a mouse model of AD (APP/PS1) and wild-type controls, and developed a simplified quantification method to facilitate large cohort preclinical imaging studies. PROCEDURES: We generated nondisplaceable binding potential (BPND) and distribution volume ratio (DVR) values using the simplified reference tissue model (SRTM) on the 90-min dynamic PET imaging data, with brain stem and cerebellum as the reference region, respectively. Then, we correlated the standardized uptake value ratio (SUVR)-1 and SUVR averaged from different imaging windows with BPND and DVR, using brain stem and cerebellum as the reference region, respectively. We performed homologous competitive binding assay and autoradiographic saturation binding assay using [18F]SynVesT-1 to calculate the Bmax and Kd. RESULTS: Using brain stem as the reference region, the averaged SUVR-1 from 30 to 60 min postinjection correlated well with the BPND calculated using SRTM. Using cerebellum as the reference region, the averaged SUVR from 30 to 60 min postinjection correlated well with the SRTM DVR. From the homologous competitive binding assay and autoradiographic saturation binding assay, the calculated the Bmax and Kd were 4.5-18 pmol/mg protein and 9.8-19.6 nM, respectively, for rodent brain tissue. CONCLUSIONS: This simplified SUVR method provides reasonable SV2A measures in APP/PS1 mice and their littermate controls. Our data indicate that, in lieu of a full 90-min dynamic scan, a 30-min static PET scan (from 30 to 60 min postinjection) would be sufficient to provide quantification data on SV2A expression, equivalent to the data generated from kinetic modeling. The methods developed here are readily applicable to the evaluation of therapeutic effects of novel drugs in this rodent model using [18F]SynVesT-1 and small animal PET.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Radioisótopos de Flúor/química , Tomografía de Emisión de Positrones/métodos , Piridinas/química , Pirrolidinas/química , Radiofármacos/farmacología , Animales , Encéfalo/metabolismo , Tronco Encefálico/diagnóstico por imagen , Tronco Encefálico/metabolismo , Cerebelo/diagnóstico por imagen , Cerebelo/metabolismo , Diseño de Fármacos , Femenino , Concentración 50 Inhibidora , Cinética , Glicoproteínas de Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica
10.
Mol Neurobiol ; 56(8): 5689-5700, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30666562

RESUMEN

Neurodegenerative stimuli are often associated with perturbation of the axon initial segment (AIS), but it remains unclear whether AIS disruption is causative for neurodegeneration or is a downstream step in disease progression. Here, we demonstrate that either of two separate, genetically parallel pathways that disrupt the AIS induce axonal degeneration and loss of neurons in the central brain of Drosophila. Expression of a portion of the C-terminal tail of the Ank2-L isoform of Ankyrin severely shortens the AIS in Drosophila mushroom body (MB) neurons, and this shortening occurs through a mechanism that is genetically separate from the previously described Cdk5α-dependent pathway of AIS regulation. Further, either manipulation triggers morphological degeneration of MB axons and is accompanied by neuron loss. Taken together, our results are consistent with the hypothesis that disruption of the AIS is causally related to degeneration of fly central brain neurons, and we suggest that similar mechanisms may contribute to neurodegeneration in mammals.


Asunto(s)
Ancirinas/metabolismo , Segmento Inicial del Axón/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Animales , Ancirinas/química , Biomarcadores/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteínas de Drosophila/química
11.
Cell Rep ; 26(1): 131-144.e4, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30605670

RESUMEN

Innate immunity is central to the pathophysiology of neurodegenerative disorders, but it remains unclear why immunity is altered in the disease state and whether changes in immunity are a cause or a consequence of neuronal dysfunction. Here, we identify a molecular pathway that links innate immunity to age-dependent loss of dopaminergic neurons in Drosophila. We find, first, that altering the expression of the activating subunit of the Cdk5 protein kinase (Cdk5α) causes severe disruption of autophagy. Second, this disruption of autophagy is both necessary and sufficient to cause the hyperactivation of innate immunity, particularly expression of anti-microbial peptides. Finally, it is the upregulation of immunity that induces the age-dependent death of dopaminergic neurons. Given the dysregulation of Cdk5 and innate immunity in human neurodegeneration and the conserved role of the kinase in the regulation of autophagy, this sequence is likely to have direct application to the chain of events in human neurodegenerative disease.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/inmunología , Neuronas Dopaminérgicas/enzimología , Neuronas Dopaminérgicas/patología , Proteínas de Drosophila/inmunología , Factores de Edad , Animales , Autofagia/fisiología , Quinasa 5 Dependiente de la Ciclina/biosíntesis , Quinasa 5 Dependiente de la Ciclina/genética , Neuronas Dopaminérgicas/inmunología , Drosophila , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/genética , Inmunidad Innata , Inmunohistoquímica , Masculino
12.
Dis Model Mech ; 11(3)2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29469033

RESUMEN

Aging is the greatest risk factor for neurodegeneration, but the connection between the two processes remains opaque. This is in part for want of a rigorous way to define physiological age, as opposed to chronological age. Here, we develop a comprehensive metric for physiological age in Drosophila, based on genome-wide expression profiling. We applied this metric to a model of adult-onset neurodegeneration, increased or decreased expression of the activating subunit of the Cdk5 protein kinase, encoded by the gene Cdk5α, the ortholog of mammalian p35. Cdk5α-mediated degeneration was associated with a 27-150% acceleration of the intrinsic rate of aging, depending on the tissue and genetic manipulation. Gene ontology analysis and direct experimental tests revealed that affected age-associated processes included numerous core phenotypes of neurodegeneration, including enhanced oxidative stress and impaired proteostasis. Taken together, our results suggest that Cdk5α-mediated neurodegeneration results from accelerated aging, in combination with cell-autonomous neuronal insults. These data fundamentally recast our picture of the relationship between neurodegeneration and its most prominent risk factor, natural aging.


Asunto(s)
Envejecimiento/fisiología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Degeneración Nerviosa/patología , Proteínas del Tejido Nervioso/metabolismo , Envejecimiento/genética , Animales , Animales Modificados Genéticamente , Autofagia , Biomarcadores/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Degeneración Nerviosa/genética , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Estrés Oxidativo , Fenotipo
13.
Dev Dyn ; 244(12): 1550-63, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26394609

RESUMEN

BACKGROUND: During metamorphosis, axons and dendrites of the mushroom body (MB) in the Drosophila central brain are remodeled extensively to support the transition from larval to adult behaviors. RESULTS: We show here that the neuronal cyclin-dependent kinase, Cdk5, regulates the timing and rate of mushroom body remodeling: reduced Cdk5 activity causes a delay in pruning of MB neurites, while hyperactivation accelerates it. We further show that Cdk5 cooperates with the ubiquitin-proteasome system in this process. Finally, we show that Cdk5 modulates the first overt step in neurite disassembly, dissolution of the neuronal tubulin cytoskeleton, and provide evidence that it also acts at additional steps of MB pruning. CONCLUSIONS: These data show that Cdk5 regulates the onset and extent of remodeling of the Drosophila MB. Given the wide phylogenetic conservation of Cdk5, we suggest that it is likely to play a role in developmental remodeling in other systems, as well. Moreover, we speculate that the well-established role of Cdk5 in neurodegeneration may involve some of the same cellular mechanisms that it uses during developmental remodeling.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteínas de Drosophila/metabolismo , Cuerpos Pedunculados/citología , Neuronas/metabolismo , Animales , Axones/metabolismo , Dendritas/metabolismo , Drosophila melanogaster , Cuerpos Pedunculados/metabolismo , Filogenia , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinación/fisiología
14.
PLoS One ; 9(1): e86998, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24489825

RESUMEN

Human and Simian Immunodeficiency virus (HIV-1, HIV-2, and SIV) encode an accessory protein, Nef, which is a pathogenesis and virulence factor. Nef is a multivalent adapter that dysregulates the trafficking of many immune cell receptors, including chemokine receptors (CKRs). Physiological endocytic itinerary of agonist occupied CXCR4 involves ubiquitinylation of the phosphorylated receptor at three critical lysine residues and dynamin-dependent trafficking through the ESCRT pathway into lysosomes for degradation. Likewise, Nef induced CXCR4 degradation was critically dependent on the three lysines in the C-terminal -SSLKILSKGK- motif. Nef directly recruits the HECT domain E3 ligases AIP4 or NEDD4 to CXCR4 in the resting state. This mechanism was confirmed by ternary interactions of Nef, CXCR4 and AIP4 or NEDD4; by reversal of Nef effect by expression of catalytically inactive AIP4-C830A mutant; and siRNA knockdown of AIP4, NEDD4 or some ESCRT-0 adapters. However, ubiquitinylation dependent lysosomal degradation was not the only mechanism by which Nef downregulated CKRs. Agonist and Nef mediated CXCR2 (and CXCR1) degradation was ubiquitinylation independent. Nef also profoundly downregulated the naturally truncated CXCR4 associated with WHIM syndrome and engineered variants of CXCR4 that resist CXCL12 induced internalization via an ubiquitinylation independent mechanism.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , VIH-1/química , Monocitos/metabolismo , Receptores CXCR4/genética , Ubiquitina/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/farmacología , Secuencias de Aminoácidos , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Endocitosis , Complejos de Clasificación Endosomal Requeridos para el Transporte/antagonistas & inhibidores , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , VIH-1/genética , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , Datos de Secuencia Molecular , Monocitos/efectos de los fármacos , Monocitos/virología , Ubiquitina-Proteína Ligasas Nedd4 , Cultivo Primario de Células , Proteolisis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores CXCR4/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo
15.
Environ Mol Mutagen ; 53(5): 392-407, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22674528

RESUMEN

Both gene methylation changes and genetic instability have been noted in offspring of male rodents exposed to radiation or chemicals, but few specific gene targets have been established. Previously, we identified the gene for ribosomal RNA, rDNA, as showing methylation change in sperm of mice treated with the preconceptional carcinogen, chromium(III) chloride. rDNA is a critical cell growth regulator. Here, we investigated the effects of paternal treatments on rDNA in offspring tissue. A total of 93 litters and 758 offspring were obtained, permitting rigorous mixed-effects models statistical analysis of the results. We show that the offspring of male mice treated with Cr(III) presented increased methylation in a promoter sequence of the rDNA gene, specifically in lung. Furthermore polymorphic variants of the multi-copy rDNA genes displayed altered frequencies indicative of structural changes, as a function of both tissue type and paternal treatments. Organismal effects also occurred: some groups of offspring of male mice treated with either Cr(III) or its vehicle, acidic saline, compared with those of untreated mice, had altered average body and liver weights and levels of serum glucose and leptin. Males treated directly with Cr(III) or acidic saline presented serum hormone changes consistent with a stress response. These results establish for the first time epigenetic and genetic instability effects in a gene of central physiological importance, in offspring of male mice exposed preconceptionally to chemicals, possibly related to a stress response in these males.


Asunto(s)
Estrés Fisiológico/efectos de los fármacos , Animales , Metilación de ADN , ADN Ribosómico/genética , Genotipo , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Secuencias Reguladoras de Ácidos Nucleicos
16.
PLoS One ; 6(10): e26112, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22022528

RESUMEN

Prostate cancers of luminal adenocarcinoma histology display a range of clinical behaviors. Although most prostate cancers are slow-growing and indolent, a proportion is aggressive, developing metastasis and resistance to androgen deprivation treatment. One hypothesis is that a portion of aggressive cancers initiate from stem-like, androgen-independent tumor-propagating cells. Here we demonstrate the in vitro creation of a mouse cell line, selected for growth as self-renewing stem/progenitor cells, which manifests many in vivo properties of aggressive prostate cancer. Normal mouse prostate epithelium containing floxed Pten and TP53 alleles was subjected to CRE-mediated deletion in vitro followed by serial propagation as protospheres. A polyclonal cell line was established from dissociated protospheres and subsequently a clonal daughter line was derived. Both lines demonstrate a mature luminal phenotype in vitro. The established lines contain a stable minor population of progenitor cells with protosphere-forming ability and multi-lineage differentiation capacity. Both lines formed orthotopic adenocarcinoma tumors with metastatic potential to lung. Intracardiac inoculation resulted in brain and lung metastasis, while intra-tibial injection induced osteoblastic bone formation, recapitulating the bone metastatic phenotype of human prostate cancer. The cells showed androgen receptor dependent growth in vitro. Importantly, in vivo, the deprivation of androgens from established orthotopic tumors resulted in tumor regression and eventually castration-resistant growth. These data suggest that transformed prostate progenitor cells preferentially differentiate toward luminal cells and recapitulate many characteristics of the human disease.


Asunto(s)
Adenocarcinoma/patología , Células Madre Multipotentes/patología , Fosfohidrolasa PTEN/deficiencia , Esferoides Celulares/patología , Proteína p53 Supresora de Tumor/deficiencia , Adenocarcinoma/metabolismo , Andrógenos/farmacología , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/patología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Castración , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Separación Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Técnicas de Inactivación de Genes , Inmunohistoquímica , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Modelos Biológicos , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/metabolismo , Metástasis de la Neoplasia , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/patología , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
17.
PLoS One ; 6(7): e22266, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21765958

RESUMEN

Gene rearrangement occurs during development in some cell types and this genome dynamics is modulated by intrinsic and extrinsic factors, including growth stimulants and nutrients. This raises a possibility that such structural change in the genome and its subsequent epigenetic modifications may also take place during mammalian ontogeny, a process undergoing finely orchestrated cell division and differentiation. We tested this hypothesis by comparing single nucleotide polymorphism-defined haplotype frequencies and DNA methylation of the rDNA multicopy gene between two mouse ontogenic stages and among three adult tissues of individual mice. Possible influences to the genetic and epigenetic dynamics by paternal exposures were also examined for Cr(III) and acid saline extrinsic factors. Variables derived from litters, individuals, and duplicate assays in large mouse populations were examined using linear mixed-effects model. We report here that active rDNA rearrangement, represented by changes of haplotype frequencies, arises during ontogenic progression from day 8 embryos to 6-week adult mice as well as in different tissue lineages and is modifiable by paternal exposures. The rDNA methylation levels were also altered in concordance with this ontogenic progression and were associated with rDNA haplotypes. Sperm showed highest level of methylation, followed by lungs and livers, and preferentially selected haplotypes that are positively associated with methylation. Livers, maintaining lower levels of rDNA methylation compared with lungs, expressed more rRNA transcript. In vitro transcription demonstrated haplotype-dependent rRNA expression. Thus, the genome is also dynamic during mammalian ontogeny and its rearrangement may trigger epigenetic changes and subsequent transcriptional controls, that are further influenced by paternal exposures.


Asunto(s)
Metilación de ADN/genética , ADN Ribosómico/genética , Reordenamiento Génico/genética , Exposición Paterna , Transcripción Genética , Animales , Secuencia de Bases , Islas de CpG/genética , Epigénesis Genética , Haplotipos/genética , Masculino , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ADN
18.
Protein Expr Purif ; 62(1): 21-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18713650

RESUMEN

Recombinant protein expression in insect cells varies greatly from protein to protein. A fusion tag that is not only a tool for detection and purification, but also enhances expression and/or solubility would greatly facilitate both structure/function studies and therapeutic protein production. We have shown that fusion of SUMO (small ubiquitin-related modifier) to several test proteins leads to enhanced expression levels in Escherichia coli. In eukaryotic expression systems, however, the SUMO tag could be cleaved by endogenous desumoylase. In order to adapt SUMO-fusion technology to these systems, we have developed an alternative SUMO-derived tag, designated SUMOstar, which is not processed by native SUMO proteases. In the present study, we tested the SUMOstar tag in a baculovirus/insect cell system with several proteins, i.e. mouse UBP43, human tryptase beta II, USP4, USP15, and GFP. Our results demonstrate that fusion to SUMOstar enhanced protein expression levels at least 4-fold compared to either the native or His(6)-tagged proteins. We isolated active SUMOstar tagged UBP43, USP4, USP15, and GFP. Tryptase was active following cleavage with a SUMOstar specific protease. The SUMOstar system will make significant impact in difficult-to-express proteins and especially to those proteins that require the native N-terminal residue for function.


Asunto(s)
Baculoviridae/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína SUMO-1/genética , Animales , Baculoviridae/metabolismo , Clonación Molecular , Endopeptidasas/genética , Endopeptidasas/aislamiento & purificación , Endopeptidasas/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes , Humanos , Ratones , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteína SUMO-1/metabolismo , Spodoptera/citología , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/aislamiento & purificación , Ubiquitina Tiolesterasa/metabolismo , Proteasas Ubiquitina-Específicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...