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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(21): 3980-3991.e18, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36182704

RESUMEN

Simian arteriviruses are endemic in some African primates and can cause fatal hemorrhagic fevers when they cross into primate hosts of new species. We find that CD163 acts as an intracellular receptor for simian hemorrhagic fever virus (SHFV; a simian arterivirus), a rare mode of virus entry that is shared with other hemorrhagic fever-causing viruses (e.g., Ebola and Lassa viruses). Further, SHFV enters and replicates in human monocytes, indicating full functionality of all of the human cellular proteins required for viral replication. Thus, simian arteriviruses in nature may not require major adaptations to the human host. Given that at least three distinct simian arteriviruses have caused fatal infections in captive macaques after host-switching, and that humans are immunologically naive to this family of viruses, development of serology tests for human surveillance should be a priority.


Asunto(s)
Arterivirus , Fiebres Hemorrágicas Virales , Animales , Arterivirus/fisiología , Fiebres Hemorrágicas Virales/veterinaria , Fiebres Hemorrágicas Virales/virología , Humanos , Macaca , Primates , Zoonosis Virales , Internalización del Virus , Replicación Viral
2.
FASEB J ; 36(5): e22306, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35385164

RESUMEN

As aortic valve stenosis develops, valve tissue becomes stiffer. In response to this change in environmental mechanical stiffness, valvular interstitial cells (VICs) activate into myofibroblasts. We aimed to investigate the role of mechanosensitive calcium channel Transient Receptor Potential Vanilloid type 4 (TRPV4) in stiffness induced myofibroblast activation. We verified TRPV4 functionality in VICs using live calcium imaging during application of small molecule modulators of TRPV4 activity. We designed hydrogel biomaterials that mimic mechanical features of healthy or diseased valve tissue microenvironments, respectively, to investigate the role of TRPV4 in myofibroblast activation and proliferation. Our results show that TRPV4 regulates VIC proliferation in a microenvironment stiffness-independent manner. While there was a trend toward inhibiting myofibroblast activation on soft microenvironments during TRPV4 inhibition, we observed near complete deactivation of myofibroblasts on stiff microenvironments. We further identified Yes-activated protein (YAP) as a downstream target for TRPV4 activity on stiff microenvironments. Mechanosensitive TRPV4 channels regulate VIC myofibroblast activation, whereas proliferation regulation is independent of the microenvironmental stiffness. Collectively, the data suggests differential regulation of stiffness-induced proliferation and myofibroblast activation. Our data further suggest a regulatory role for TRPV4 regarding YAP nuclear localization. TRPV4 is an important regulator for VIC myofibroblast activation, which is linked to the initiation of valve fibrosis. Although more validation studies are necessary, we suggest TRPV4 as a promising pharmaceutical target to slow aortic valve stenosis progression.


Asunto(s)
Estenosis de la Válvula Aórtica , Calcinosis , Miofibroblastos , Animales , Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/metabolismo , Calcinosis/metabolismo , Proliferación Celular , Células Cultivadas , Hidrogeles , Miofibroblastos/metabolismo , Porcinos , Canales Catiónicos TRPV/metabolismo
3.
PLoS Pathog ; 16(3): e1008403, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32203554

RESUMEN

The replication of small DNA viruses requires both host DNA replication and repair factors that are often recruited to subnuclear domains termed viral replication centers (VRCs). Aside from serving as a spatial focus for viral replication, little is known about these dynamic areas in the nucleus. We investigated the organization and function of VRCs during murine polyomavirus (MuPyV) infection using 3D structured illumination microscopy (3D-SIM). We localized MuPyV replication center components, such as the viral large T-antigen (LT) and the cellular replication protein A (RPA), to spatially distinct subdomains within VRCs. We found that viral DNA (vDNA) trafficked sequentially through these subdomains post-synthesis, suggesting their distinct functional roles in vDNA processing. Additionally, we observed disruption of VRC organization and vDNA trafficking during mutant MuPyV infections or inhibition of DNA synthesis. These results reveal a dynamic organization of VRC components that coordinates virus replication.


Asunto(s)
Núcleo Celular/virología , ADN Viral/metabolismo , Infecciones por Polyomavirus/metabolismo , Poliomavirus/fisiología , Replicación Viral/fisiología , Transporte Activo de Núcleo Celular/genética , Animales , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/metabolismo , Línea Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , ADN Viral/genética , Ratones , Infecciones por Polyomavirus/genética , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo
4.
Int Immunol ; 29(6): 277-289, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28814066

RESUMEN

Specific variants of endoplasmic reticulum-associated aminopeptidase 1 (ERAP1) identified by genome-wide association study modify the risk for developing ankylosing spondylitis. We previously confirmed that disease-associated ERAP1 variants have altered enzymatic abilities that can impact upon the production of pro-inflammatory cytokines from cells expressing the same ERAP1 variants. To determine if these ERAP1 variants also impacted immune responses in vivo, we generated two strains of transgenic mice expressing human ERAP1 genes containing non-synonymous single-nucleotide polymorphisms associated with an increased (ERAP1-High) or decreased (ERAP1-Low) risk for developing autoimmune disease. After vaccination with foreign antigens, ERAP1-High mice generated unique populations of antigen-specific T-cell clones. The expression of ERAP1-High also reduced MHC-I expression on the surface of multiple cell types, demonstrating a global impact on the MHC-I peptidome. ERAP1 variants also affected the innate immune system, because NK cells from murine ERAP1 (mERAP1) knockout mice and ERAP1-High/mERAP1-/- mice had decreased surface expression of the activating receptor NKG2D on their NK and T cells, and NK cells derived from mERAP1-/- mice or ERAP1-Low mice demonstrated more active NK cell killing than NK cells derived from wild-type or ERAP1-High mice. Finally, these studies were conducted in female mice, as all male ERAP1-High mice died in utero or shortly after birth, making ERAP1-High one of the only dominant lethal autosomal genes known in mammals. Together, these results present the first direct evidence that human disease-associated ERAP1 variants can greatly alter survival, as well as antigen presentation, T-cell repertoire and NK cell responses in vivo.


Asunto(s)
Aminopeptidasas/genética , Citotoxicidad Inmunológica/genética , Células Asesinas Naturales/fisiología , Antígenos de Histocompatibilidad Menor/genética , Espondilitis Anquilosante/genética , Linfocitos T/fisiología , Inmunidad Adaptativa/genética , Animales , Presentación de Antígeno , Células Clonales , Femenino , Predisposición Genética a la Enfermedad , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunidad Innata/genética , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Polimorfismo de Nucleótido Simple , Receptores de Antígenos de Linfocitos T/genética , Riesgo , Transgenes/genética
5.
Neurobiol Dis ; 81: 49-65, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26303889

RESUMEN

The dysregulation of iron metabolism in Alzheimer's disease is not accounted for in the current framework of the amyloid cascade hypothesis. Accumulating evidence suggests that impaired iron homeostasis is an early event in Alzheimer's disease progression. Iron dyshomeostasis leads to a loss of function in several enzymes requiring iron as a cofactor, the formation of toxic oxidative species, and the elevated production of beta-amyloid proteins. Several common genetic polymorphisms that cause increased iron levels and dyshomeostasis have been associated with Alzheimer's disease but the pathoetiology is not well understood. A full picture is necessary to explain how heterogeneous circumstances lead to iron loading and amyloid deposition. There is evidence to support a causative interplay between the concerted loss of iron homeostasis and amyloid plaque formation. We hypothesize that iron misregulation and beta-amyloid plaque pathology are synergistic in the process of neurodegeneration and ultimately cause a downward cascade of events that spiral into the manifestation of Alzheimer's disease. In this review, we amalgamate recent findings of brain iron metabolism in healthy versus Alzheimer's disease brains and consider unique mechanisms of iron transport in different brain cells as well as how disturbances in iron regulation lead to disease etiology and propagate Alzheimer's pathology.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/metabolismo , Hierro/metabolismo , Péptidos beta-Amiloides/metabolismo , Homeostasis/fisiología , Humanos , Placa Amiloide/etiología , Placa Amiloide/metabolismo
6.
NMR Biomed ; 28(3): 297-305, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25530083

RESUMEN

Dysregulation of neural iron is known to occur during the progression of Alzheimer's disease. The visualization of amyloid-beta (Aß) plaques with MRI has largely been credited to rapid proton relaxation in the vicinity of plaques as a result of focal iron deposition. The goal of this work was to determine the relationship between local relaxation and related focal iron content associated with Aß plaques. Alzheimer's disease (n=5) and control tissue (n=3) sample slices from the entorhinal cortex were treated overnight with the iron chelator deferoxamine or saline, and microscopic gradient-echo MRI datasets were taken. Subsequent to imaging, the same slices were stained for Aß and iron, and then compared with regard to parametric R2 * relaxation maps and gradient-echo-weighted MR images. Aß plaques in both chelated and unchelated tissue generated MR hypo-intensities and showed relaxation rates significantly greater than the surrounding tissue. The transverse relaxation rate associated with amyloid plaques was determined not to be solely a result of iron load, as much of the relaxation associated with Aß plaques remained following iron chelation. The data indicate a dual relaxation mechanism associated with Aß plaques, such that iron and plaque composition synergistically produce transverse relaxation.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Hierro/farmacología , Imagen por Resonancia Magnética , Placa Amiloide/metabolismo , Anciano , Enfermedad de Alzheimer/patología , Estudios de Casos y Controles , Deferoxamina/farmacología , Humanos , Placa Amiloide/patología
7.
Bioeng Transl Med ; 8(1): e10358, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36684107

RESUMEN

Patients with aortic valve stenosis (AVS) have sexually dimorphic phenotypes in their valve tissue, where male valvular tissue adopts a calcified phenotype and female tissue becomes more fibrotic. The molecular mechanisms that regulate sex-specific calcification in valvular tissue remain poorly understood. Here, we explored the role of osteopontin (OPN), a pro-fibrotic but anti-calcific bone sialoprotein, in regulating the calcification of female aortic valve tissue. Recognizing that OPN mediates calcification processes, we hypothesized that aortic valvular interstitial cells (VICs) in female tissue have reduced expression of osteogenic markers in the presence of elevated OPN relative to male VICs. Human female valve leaflets displayed reduced and smaller microcalcifications, but increased OPN expression relative to male leaflets. To understand how OPN expression contributes to observed sex dimorphisms in valve tissue, we employed enzymatically degradable hydrogels as a 3D cell culture platform to recapitulate male or female VIC interactions with the extracellular matrix. Using this system, we recapitulated sex differences observed in human tissue, specifically demonstrating that female VICs exposed to calcifying medium have smaller mineral deposits within the hydrogel relative to male VICs. We identified a change in OPN dynamics in female VICs in the presence of calcification stimuli, where OPN deposition localized from the extracellular matrix to perinuclear regions. Additionally, exogenously delivered endothelin-1 to encapsulated VICs increased OPN gene expression in male cells, which resulted in reduced calcification. Collectively, our results suggest that increased OPN in female valve tissue may play a sex-specific role in mitigating mineralization during AVS progression.

8.
Biomater Sci ; 10(22): 6341-6353, 2022 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-36226463

RESUMEN

Aortic valve stenosis (AVS) is a sexually dimorphic cardiovascular disease that is driven by fibrosis and calcification of the aortic valve leaflets. Circulating inflammatory factors present in serum from AVS patients contribute to sex differences in valve fibro-calcification by driving the activation of valvular interstitial cells (VICs) to myofibroblasts and/or osteoblast-like cells. However, the molecular mechanisms by which inflammatory factors contribute to sex-specific valve fibro-calcification remain largely unknown. In this study, we identified inflammatory factors present in serum samples from AVS patients that regulate sex-specific myofibroblast activation and osteoblast-like differentiation. After correlating serum proteomic datasets with clinical and in vitro myofibroblast datasets, we identified annexin A2 and cystatin C as candidate inflammatory factors that correlate with both AVS patient severity and myofibroblast activation measurements in vitro. Validation experiments utilizing hydrogel biomaterials as cell culture platforms that mimic the valve extracellular matrix confirmed that annexin A2 and cystatin C promote sex-specific VIC activation to myofibroblasts via p38 MAPK signaling. Additionally, annexin A2 and cystatin C increase osteoblast-like differentiation primarily in male VICs. Our results implicate serum inflammatory factors as potential AVS biomarkers that also contribute to sexually dimorphic AVS progression by driving VIC myofibroblast activation and/or osteoblast-like differentiation. Collectively, the results herein further our overall understanding as to how biological sex may impact inflammation-driven AVS and may lead to the development of sex-specific drug treatment strategies.


Asunto(s)
Anexina A2 , Estenosis de la Válvula Aórtica , Calcinosis , Humanos , Masculino , Femenino , Miofibroblastos , Válvula Aórtica , Cistatina C , Caracteres Sexuales , Proteómica , Células Cultivadas , Osteoblastos
9.
J Neuroinflammation ; 8(1): 4, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21235806

RESUMEN

BACKGROUND: Complement proteins and activation products have been found associated with neuropathology in Alzheimer's disease (AD). Recently, a C5a receptor antagonist was shown to suppress neuropathology in two murine models of AD, Tg2576 and 3xTg. Previously, a genetic deficiency of C1q in the Tg2576 mouse model showed an accumulation of fibrillar plaques similar to the complement sufficient Tg2576, but reactive glia were significantly decreased and neuronal integrity was improved suggesting detrimental consequences for complement activation in AD. The goal of this study was to define the role of the classical complement activation pathway in the progression of pathology in the 3xTg mouse that develops tangles in addition to fibrillar plaques (more closely reflecting human AD pathology) and to assess the influence of complement in a model of AD with a higher level of complement hemolytic activity. METHODS: 3xTg mice deficient in C1q (3xTgQ-/-) were generated, and both 3xTg and 3xTgQ-/- were backcrossed to the BUB mouse strain which has higher in vitro hemolytic complement activity. Mice were aged and perfused, and brain sections stained for pathological markers or analyzed for proinflammatory marker expression. RESULTS: 3xTgQ-/- mice showed similar amounts of fibrillar amyloid, reactive glia and hyperphosphorylated tau as the C1q-sufficient 3xTg at the ages analyzed. However, 3xTg and 3xTgQ-/- on the BUB background developed pathology earlier than on the original 3xTg background, although the presence of C1q had no effect on neuropathological and pro-inflammatory markers. In contrast to that seen in other transgenic models of AD, C1q, C4 and C3 immunoreactivity was undetectable on the plaques of 3xTg in any background, although C3 was associated with reactive astrocytes surrounding the plaques. Importantly, properdin a component of the alternative complement pathway was associated with plaques in all models. CONCLUSIONS: In contrast to previously investigated transgenic models of AD, development of neuropathology in 3xTg mice, which progresses much slower than other murine models, may not be influenced by fibrillar amyloid mediated activation of the classical complement pathway, suggesting that the alternative complement pathway activation or a C3-independent cleavage of C5 could account for the detrimental effects in these mice that are prevented by the C5a receptor antagonist. Furthermore, the paucity of complement activation may be a factor in the slower kinetics of progression of pathology in the 3xTg model of this disease.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Encéfalo/inmunología , Encéfalo/patología , Activación de Complemento , Modelos Animales de Enfermedad , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Biomarcadores/metabolismo , Femenino , Humanos , Factores Inmunológicos/inmunología , Masculino , Ratones , Ratones Transgénicos , Properdina/inmunología , Proteínas tau/genética , Proteínas tau/metabolismo
10.
Viruses ; 12(10)2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33023278

RESUMEN

During polyomavirus (PyV) infection, host proteins localize to subnuclear domains, termed viral replication centers (VRCs), to mediate viral genome replication. Although the protein composition and spatial organization of VRCs have been described using high-resolution immunofluorescence microscopy, little is known about the temporal dynamics of VRC formation over the course of infection. We used live cell fluorescence microscopy to analyze VRC formation during murine PyV (MuPyV) infection of a mouse fibroblast cell line that constitutively expresses a GFP-tagged replication protein A complex subunit (GFP-RPA32). The RPA complex forms a heterotrimer (RPA70/32/14) that regulates cellular DNA replication and repair and is a known VRC component. We validated previous observations that GFP-RPA32 relocalized to sites of cellular DNA damage in uninfected cells and to VRCs in MuPyV-infected cells. We then used GFP-RPA32 as a marker of VRC formation and expansion during live cell microscopy of infected cells. VRC formation occurred at variable times post-infection, but the rate of VRC expansion was similar between cells. Additionally, we found that the early viral protein, small TAg (ST), was required for VRC expansion but not VRC formation, consistent with the role of ST in promoting efficient vDNA replication. These results demonstrate the dynamic nature of VRCs over the course of infection and establish an approach for analyzing viral replication in live cells.


Asunto(s)
Microscopía/métodos , Infecciones por Polyomavirus/virología , Poliomavirus/fisiología , Proteína de Replicación A/metabolismo , Replicación Viral/fisiología , Animales , Línea Celular/citología , Daño del ADN , Replicación del ADN , ADN Viral/genética , Genoma Viral , Cinética , Ratones , Ratones Endogámicos C57BL , Poliomavirus/genética , Infecciones por Polyomavirus/patología , Proteína de Replicación A/genética
11.
Brain Struct Funct ; 223(3): 1519-1536, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29168009

RESUMEN

Impaired brain iron homeostatic mechanisms, independent of pathological hallmarks, are harmful to the brain because excess free iron can cause DNA, protein, and lipid damage via oxidative stress. The goal of this study was to evaluate the longitudinal effect of chronic iron overload and deficiency in the vertebrate brain. Ten-week-old C57BL6 male mice were randomly assigned to one of four unique dietary regiments for 1 year: iron-deficient, normal iron, and two different concentrations of lipophilic iron diet containing 3,5,5-trimethylhexanoyl ferrocene (TMHF). Longitudinal MRI parametrics were used to assess the location and extent of ferric iron distribution. Tissue collected at 12 months was used to directly measure iron-load, protein alterations, and histological metrics. While the iron-deficient diet did not alter brain iron stores, 0.11% TMHF and early exposure with 0.5% TMHF elevated brain iron by roughly 40 and 100%, respectively. R 2 rate increased more in the TMHF groups within iron rich brain regions. Increased brain iron concentration was linearly correlated with an increase in L-ferritin expression, and TMHF diet was found to increase L-ferritin within the olfactory bulb, neocortex, pallidum, thalamus, corpus callosum, CA3 regions of the hippocampus, and substantia nigra. Moreover, gliosis and oxidative stress were detected in the TMHF groups in the regions associated with iron-load. Spatial memory impairment was evident in the iron-loaded mice. This work illustrates that lipophilic iron elevates brain iron in a regionally specific fashion and positions dietary TMHF administration as a model for brain iron overloading.


Asunto(s)
Encéfalo/metabolismo , Compuestos Ferrosos/administración & dosificación , Hierro/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Suplementos Dietéticos , Compuestos Ferrosos/química , Procesamiento de Imagen Asistido por Computador , Hígado/efectos de los fármacos , Hígado/enzimología , Imagen por Resonancia Magnética , Masculino , Espectrometría de Masas , Aprendizaje por Laberinto , Trastornos de la Memoria/etiología , Trastornos de la Memoria/patología , Trastornos de la Memoria/fisiopatología , Metalocenos , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
12.
Metallomics ; 10(3): 426-443, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29424844

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized pathologically by amyloid beta (Aß) deposition, microgliosis, and iron dyshomeostasis. Increased labile iron due to homeostatic dysregulation is believed to facilitate amyloidogenesis. Free iron is incorporated into aggregating amyloid peptides during Aß plaque formation and increases potential for oxidative stress surrounding plaques. The goal of this work was to observe how brain iron levels temporally influence Aß plaque formation, plaque iron concentration, and microgliosis. We fed humanized APPNL-F and APPNL-G-F knock-in mice lipophilic iron compound 3,5,5-trimethylhexanoyl ferrocene (TMHF) and iron deficient diets for twelve months. TMHF elevated brain iron by 22% and iron deficiency decreased brain iron 21% relative to control diet. Increasing brain iron with TMHF accelerated plaque formation, increased Aß staining, and increased senile morphology of amyloid plaques. Increased brain iron was associated with increased plaque-iron loading and microglial iron inclusions. TMHF decreased IBA1+ microglia branch length while increasing roundness indicative of microglial activation. This body of work suggests that increasing mouse brain iron with TMHF potentiates a more human-like Alzheimer's disease phenotype with iron integration into Aß plaques and associated microgliosis.


Asunto(s)
Enfermedad de Alzheimer/patología , Dieta , Modelos Animales de Enfermedad , Hierro/metabolismo , Microglía/patología , Placa Amiloide/patología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Femenino , Humanos , Hierro/administración & dosificación , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Fenotipo , Placa Amiloide/metabolismo
13.
Front Neuroanat ; 11: 58, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28790895

RESUMEN

Vanadium is a potentially toxic environmental pollutant and induces oxidative damage in biological systems including the central nervous system (CNS). Its deposition in brain tissue may be involved in the pathogenesis of certain neurological disorders which after prolonged exposure can culminate into more severe pathology. Most studies on vanadium neurotoxicity have been done after acute exposure but in reality some populations are exposed for a lifetime. This work was designed to ascertain neurodegenerative consequences of chronic vanadium administration and to investigate the progressive changes in the brain after withdrawal from vanadium treatment. A total of 85 male BALB/c mice were used for the experiment and divided into three major groups of vanadium treated (intraperitoneally (i.p.) injected with 3 mg/kg body weight of sodium metavanadate and sacrificed every 3 months till 18 months); matched controls; and animals that were exposed to vanadium for 3 months and thereafter the metal was withdrawn. Brain tissues were obtained after animal sacrifice. Sagittal cut sections of paraffin embedded tissue (5 µm) were analyzed by the Laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) to show the absorption and distribution of vanadium metal. Also, Haematoxylin and Eosin (H&E) staining of brain sections, and immunohistochemistry for Microglia (Iba-1), Astrocytes (GFAP), Neurons (Neu-N) and Neu-N + 4',6-diamidine-2'-pheynylindole dihydrochloride (Dapi) Immunofluorescent labeling were observed for morphological and morphometric parameters. The LA-ICP-MS results showed progressive increase in vanadium uptake with time in different brain regions with prediction for regions like the olfactory bulb, brain stem and cerebellum. The withdrawal brains still show presence of vanadium metal in the brain slightly more than the controls. There were morphological alterations (of the layering profile, nuclear shrinkage) in the prefrontal cortex, cellular degeneration (loss of dendritic arborization) and cell death in the Hippocampal CA1 pyramidal cells and Purkinje cells of the cerebellum, including astrocytic and microglial activation in vanadium exposed brains which were all attenuated in the withdrawal group. With exposure into old age, the evident neuropathology was microgliosis, while progressive astrogliosis became more attenuated. We have shown that chronic administration of vanadium over a lifetime in mice resulted in metal accumulation which showed regional variabilities with time. The metal profile and pathological effects were not completely eliminated from the brain even after a long time withdrawal from vanadium metal.

14.
Brain Imaging Behav ; 10(4): 1231-1242, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26660104

RESUMEN

Mutations within the HFE protein gene sequence have been associated with increased risk of developing a number of neurodegenerative disorders. To this effect, an animal model has been created which incorporates the mouse homologue to the human H63D-HFE mutation: the H67D-HFE knock-in mouse. These mice exhibit alterations in iron management proteins, have increased neuronal oxidative stress, and a disruption in cholesterol regulation. However, it remains undetermined how these differences translate to human H63D carriers in regards to white matter (WM) integrity. To this endeavor, MRI transverse relaxation rate (R2) parametrics were employed to test the hypothesis that WM alterations are present in H63D human carriers and are recapitulated in the H67D mice. H63D carriers exhibit widespread reductions in brain R2 compared to non-carriers within white matter association fibers in the brain. Similar R2 decreases within white matter tracts were observed in the H67D mouse brain. Additionally, an exacerbation of age-related R2 decrease is found in the H67D animal model in white matter regions of interest. The decrease in R2 within white matter tracts of both species is speculated to be multifaceted. The R2 changes are hypothesized to be due to alterations in axonal biochemical tissue composition. The R2 changes observed in both the human-H63D and mouse-H67D data suggest that modified white matter myelination is occurring in subjects with HFE mutations, potentially increasing vulnerability to neurodegenerative disorders.


Asunto(s)
Proteína de la Hemocromatosis/genética , Imagen por Resonancia Magnética , Sustancia Blanca/diagnóstico por imagen , Anciano , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Estudios Transversales , Interpretación Estadística de Datos , Modelos Animales de Enfermedad , Femenino , Técnicas de Sustitución del Gen , Predisposición Genética a la Enfermedad , Técnicas de Genotipaje , Heterocigoto , Humanos , Procesamiento de Imagen Asistido por Computador , Estudios Longitudinales , Masculino , Escala del Estado Mental , Ratones Endogámicos C57BL , Ratones Transgénicos , Pruebas Neuropsicológicas , Sustancia Blanca/fisiopatología
15.
Clin Transl Sci ; 8(6): 729-33, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26663505

RESUMEN

As part of the Clinical and Translational Science Institute predoctoral TL1 training program at the Pennsylvania State University, a multidisciplinary team of predoctoral trainees representing the Chemistry, Neurosurgery, Nutritional Sciences, and Public Health Sciences departments were introduced to the NIH-sponsored Informatics for Integrating Biology and the Bedside (i2b2) database to test the following student-generated hypothesis: children with iron deficiency anemia (IDA) are at increased risk of attention deficit-hyperactivity disorder (ADHD). Children aged 4-12 and 4-17 years were categorized into IDA and control groups. De-identified medical records from the Penn State Milton S. Hershey Medical Center (HMC) and the Virginia Commonwealth University Medical Center (VCUMC) were used for the analysis. Overall, ADHD prevalence at each institution was lower than 2011 state estimates. There was a significant association between IDA and ADHD in the 4-17-year-old age group for all children (OR: 1.902 [95% CI: 1.363-2.656]), Caucasian children (OR: 1.802 [95% CI: 1.133-2.864]), and African American children (OR: 1.865 [95% CI: 1.152-3.021]). Clinical and Translational Science Award (CTSA) infrastructure is particularly useful for trainees to answer de novo scientific questions with minimal additional training and technical expertise. Moreover, projects can be expanded by collaborating within the CTSA network.


Asunto(s)
Anemia Ferropénica/complicaciones , Trastorno por Déficit de Atención con Hiperactividad/complicaciones , Curriculum , Investigación Biomédica Traslacional/educación , Investigación Biomédica Traslacional/métodos , Adolescente , Niño , Preescolar , Registros Electrónicos de Salud , Femenino , Humanos , Masculino , Informática Médica/métodos , National Institutes of Health (U.S.) , Pennsylvania , Prevalencia , Proyectos de Investigación , Programa de VERF , Estudiantes de Medicina , Estados Unidos , Virginia
16.
J Air Waste Manag Assoc ; 54(2): 229-41, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14977324

RESUMEN

Protocols have been developed and applied for the generation of aerosols that are likely to be comparable to those encountered in field settings for the calibration of easily transportable/portable real-time particle monitors. Aerosols generated were simulated environmental tobacco smoke, cedar wood smoke, cooking oil fumes, and propane stove particles. The time-integrated responses of three nephelometers and a monitor for particle-bound polynuclear aromatic hydrocarbons (PAH) were compared with gravimetric respirable suspended particulate matter (RSP) in a controlled-atmosphere chamber. In general, the monitor responses increased linearly with increasing mass concentration. However, the two monitors that reported mass per volume concentrations tended to overreport the actual RSP concentrations by factors up to 4.4. The real-time PAH monitor did not respond to cooking oil fumes, indicative of little PAH being present in the aerosol. One of the monitors that has been used in a variety of studies reported in the literature (DustTrak) was collocated with gravimetric RSP samplers in several hospitality venues in the Louisville, KY, area. Field studies indicated that the units overreported actual RSP concentrations by factors of 2.6-3.1, depending on whether the sampling was conducted in the nonsmoking or smoking sections of the facilities.


Asunto(s)
Aerosoles/análisis , Contaminación del Aire Interior/análisis , Monitoreo del Ambiente/métodos , Calibración , Monitoreo del Ambiente/instrumentación , Tamaño de la Partícula , Reproducibilidad de los Resultados , Factores de Tiempo
17.
Adv Neurobiol ; 9: 33-45, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25151373

RESUMEN

The brain consists of neurons and glial cells. Neurons are responsible for integrating input and responding to stimuli from both the internal and the external environment. The integration occurs via electrical and chemical signals that impinge on the receptive area of neurons known as dendrites, and the response is via propagation of an axonal potential. Glial cells have three functionally distinct subtypes, astrocytes, oligodendrocytes, and microglia. Astrocytes perform a variety of functions responsible for maintaining homeostasis in the brain through functions such as formation of the blood-brain barrier, preserving osmolarity, and the uptake, degradation, and secretion of neurotransmitters. Oligodendrocytes are responsible for the production of myelin, a lipid-rich substance that encapsulates neuronal axons. Microglia are responsible for immune surveillance and remodeling of the CNS during both normal development and injury. Together the cells of the brain form a highly metabolic and dynamic unit with robust requirements for oxygen and nutrients.

18.
Neuropsychopharmacology ; 36(7): 1375-84, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21389980

RESUMEN

Brain iron increases with age and is abnormally elevated early in the disease process in several neurodegenerative disorders that impact memory including Alzheimer's disease (AD). Higher brain iron levels are associated with male gender and presence of highly prevalent allelic variants in genes encoding for iron metabolism proteins (hemochromatosis H63D (HFE H63D) and transferrin C2 (TfC2)). In this study, we examined whether in healthy older individuals memory performance is associated with increased brain iron, and whether gender and gene variant carrier (IRON+) vs noncarrier (IRON-) status (for HFE H63D/TfC2) modify the associations. Tissue iron deposited in ferritin molecules can be measured in vivo with magnetic resonance imaging utilizing the field-dependent relaxation rate increase (FDRI) method. FDRI was assessed in hippocampus, basal ganglia, and white matter, and IRON+ vs IRON- status was determined in a cohort of 63 healthy older individuals. Three cognitive domains were assessed: verbal memory (delayed recall), working memory/attention, and processing speed. Independent of gene status, worse verbal-memory performance was associated with higher hippocampal iron in men (r=-0.50, p=0.003) but not in women. Independent of gender, worse verbal working memory performance was associated with higher basal ganglia iron in IRON- group (r=-0.49, p=0.005) but not in the IRON+ group. Between-group interactions (p=0.006) were noted for both of these associations. No significant associations with white matter or processing speed were observed. The results suggest that in specific subgroups of healthy older individuals, higher accumulations of iron in vulnerable gray matter regions may adversely impact memory functions and could represent a risk factor for accelerated cognitive decline. Combining genetic and MRI biomarkers may provide opportunities to design primary prevention clinical trials that target high-risk groups.


Asunto(s)
Envejecimiento , Encéfalo/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Hierro/metabolismo , Proteínas de la Membrana/genética , Memoria/fisiología , Caracteres Sexuales , Transferrina/genética , Anciano , Atención/fisiología , Encéfalo/anatomía & histología , Femenino , Proteína de la Hemocromatosis , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Pruebas Neuropsicológicas , Relajación , Aprendizaje Verbal
19.
J Alzheimers Dis ; 20(1): 333-41, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20164577

RESUMEN

Prevalent gene variants involved in iron metabolism [hemochromatosis (HFE) H63D and transferrin C2 (TfC2)] have been associated with higher risk and earlier age at onset of Alzheimer's disease (AD), especially in men. Brain iron increases with age, is higher in men, and is abnormally elevated in several neurodegenerative diseases, including AD and Parkinson's disease, where it has been reported to contribute to younger age at onset in men. The effects of the common genetic variants (HFE H63D and/or TfC2) on brain iron were studied across eight brain regions (caudate, putamen, globus pallidus, thalamus, hippocampus, white matter of frontal lobe, genu, and splenium of corpus callosum) in 66 healthy adults (35 men, 31 women) aged 55 to 76. The iron content of ferritin molecules (ferritin iron) in the brain was measured with MRI utilizing the Field Dependent Relaxation Rate Increase (FDRI) method. 47% of the sample carried neither genetic variant (IRON-) and 53% carried one and/or the other (IRON+). IRON+ men had significantly higher FDRI compared to IRON- men (p=0.013). This genotype effect was not observed in women who, as expected, had lower FDRI than men. This is the first published evidence that these highly prevalent genetic variants in iron metabolism genes can influence brain iron levels in men. Clinical phenomena such as differential gender-associated risks of developing neurodegenerative diseases and age at onset may be associated with interactions between iron genes and brain iron accumulation. Clarifying mechanisms of brain iron accumulation may help identify novel interventions for age-related neurodegenerative diseases.


Asunto(s)
Encéfalo/metabolismo , Ferritinas/metabolismo , Variación Genética/genética , Antígenos de Histocompatibilidad Clase I/genética , Proteínas de la Membrana/genética , Mutación/genética , Caracteres Sexuales , Anciano , Envejecimiento/genética , Envejecimiento/patología , Encéfalo/anatomía & histología , Mapeo Encefálico , Femenino , Regulación de la Expresión Génica/genética , Proteína de la Hemocromatosis , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Análisis Multivariante , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/metabolismo
20.
Neurobiol Aging ; 31(9): 1554-62, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18926601

RESUMEN

OBJECTIVE: Myelination of the human brain results in roughly quadratic trajectories of myelin content and integrity, reaching a maximum in mid-life and then declining in older age. This trajectory is most evident in vulnerable later myelinating association regions such as frontal lobes and may be the biological substrate for similar trajectories of cognitive processing speed. Speed of movement, such as maximal finger tapping speed (FTS), requires high-frequency action potential (AP) bursts and is associated with myelin integrity. We tested the hypothesis that the age-related trajectory of FTS is related to brain myelin integrity. METHODS: A sensitive in vivo MRI biomarker of myelin integrity (calculated transverse relaxation rates (R(2))) of frontal lobe white matter (FLwm) was measured in a sample of very healthy males (N=72) between 23 and 80 years of age. To assess specificity, R(2) of a contrasting early-myelinating region (splenium of the corpus callosum) was also measured. RESULTS: FLwm R(2) and FTS measures were significantly correlated (r=.45, p<.0001) with no association noted in the early-myelinating region (splenium). Both FLwm R(2) and FTS had significantly quadratic lifespan trajectories that were virtually indistinguishable and both reached a peak at 39 years of age and declined with an accelerating trajectory thereafter. CONCLUSIONS: The results suggest that in this very healthy male sample, maximum motor speed requiring high-frequency AP burst may depend on brain myelin integrity. To the extent that the FLwm changes assessed by R(2) contribute to an age-related reduction in AP burst frequency, it is possible that other brain functions dependent on AP bursts may also be affected. Non-invasive measures of myelin integrity together with testing of basic measures of processing speed may aid in developing and targeting anti-aging treatments to mitigate age-related functional declines.


Asunto(s)
Envejecimiento/fisiología , Corteza Motora/fisiología , Movimiento/fisiología , Vaina de Mielina/fisiología , Vaina de Mielina/ultraestructura , Resistencia Física/fisiología , Esfuerzo Físico/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Envejecimiento/patología , Humanos , Masculino , Persona de Mediana Edad , Corteza Motora/ultraestructura , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA