Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Analyst ; 147(23): 5274-5282, 2022 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-36346247

RESUMEN

Magnetic resonance imaging (MRI) is the gold standard method to study brain anatomy in vivo. Using MRI, subtle alterations to white matter structures in the brain are observed prior to cognitive decline associated with the ageing process, and neurodegenerative diseases such as Alzheimer's disease. Detection of such alterations provides hope for early clinical diagnosis. While MRI is essential to detect subtle alterations to brain structure in vivo, the technique is less suited to study and image the distribution of biochemical markers within specific brain structures. Consequently, the chemical changes that drive, or are associated with MRI-detectable alterations to white matter are not well understood. Herein, we describe (to the best of our knowledge) the first application of a complementary imaging approach that incorporates in vivo MRI with ex vivo Fourier transform infrared (FTIR) spectroscopic imaging on the same brain tissue. The combined workflow is used to detect and associate markers of altered biochemistry (FTIR) with anatomical changes to brain white matter (MRI). We have applied this combination of techniques to the senescence accelerated murine prone strain 8 (SAMP8) mouse model (n = 6 animals in each group, analysed across two ageing time points, 6 and 12 months). The results have demonstrated alterations to lipid composition and markers of disturbed metabolism during ageing are associated with loss of white matter volume.


Asunto(s)
Sustancia Blanca , Animales , Ratones , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Química Encefálica , Análisis de Fourier , Espectroscopía Infrarroja por Transformada de Fourier , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Imagen por Resonancia Magnética/métodos , Envejecimiento , Neuroimagen
2.
Analyst ; 146(11): 3516-3525, 2021 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-33881057

RESUMEN

Visualising direct biochemical markers of cell physiology and disease pathology at the sub-cellular level is an ongoing challenge in the biological sciences. A suite of microscopies exists to either visualise sub-cellular architecture or to indirectly view biochemical markers (e.g. histochemistry), but further technique developments and innovations are required to increase the range of biochemical parameters that can be imaged directly, in situ, within cells and tissue. Here, we report our continued advancements in the application of synchrotron radiation attenuated total reflectance Fourier transform infrared (SR-ATR-FTIR) microspectroscopy to study sub-cellular biochemistry. Our recent applications demonstrate the much needed capability to map or image directly sub-cellular protein aggregates within degenerating neurons as well as lipid inclusions within bacterial cells. We also characterise the effect of spectral acquisition parameters on speed of data collection and the associated trade-offs between a realistic experimental time frame and spectral/image quality. Specifically, the study highlights that the choice of 8 cm-1 spectral resolutions provide a suitable trade-off between spectral quality and collection time, enabling identification of important spectroscopic markers, while increasing image acquisition by ∼30% (relative to 4 cm-1 spectral resolution). Further, this study explores coupling a focal plane array detector with SR-ATR-FTIR, revealing a modest time improvement in image acquisition time (factor of 2.8). Such information continues to lay the foundation for these spectroscopic methods to be readily available for, and adopted by, the biological science community to facilitate new interdisciplinary endeavours to unravel complex biochemical questions and expand emerging areas of study.


Asunto(s)
Agregado de Proteínas , Sincrotrones , Lípidos , Proteínas , Espectroscopía Infrarroja por Transformada de Fourier
3.
Biochemistry ; 56(32): 4107-4116, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28718623

RESUMEN

Alzheimer's disease (AD) is a major international health and economic concern. A key pathological feature of AD is so-called "amyloid-ß-plaques", or "Aß-plaques", which are deposits of aggregated protein, enriched with the Aß fragment of amyloid precursor protein. Despite their name, the deposits are not pure Aß and have a heterogeneous, chemically complex composition that can include multiple proteins, lipids, and metal ions (Fe, Cu, or Zn). Despite extensive research, it is still uncertain whether Aß-plaques are a cause or a consequence of AD pathology. Further characterization of the elemental and biochemical composition within and surrounding Aß-plaques, and knowledge of how composition varies with disease state or progression, may provide important insight into the relationship between Aß-plaques and AD pathology. With this aim in mind, herein we demonstrate a multimodal spectroscopic imaging workflow to better characterize the complex composition of Aß-plaques. Our approach incorporates several spectroscopic imaging techniques, such as Fourier transform infrared spectroscopic imaging (FTIR), Raman microscopy, and X-ray fluorescence microscopy (XFM). While FTIR, Raman, and XFM have been used previously, mostly in isolation, to study Aß-plaques, application of all three techniques, in combination with histology and fluorescence microscopy, has not been reported previously. We demonstrate that a multimodal workflow, incorporating all three methods on adjacent or serial tissue sections, can reveal substantial complementary information about the biochemical and elemental composition of Aß-plaques. Information revealed by the method includes the relative content and distribution of aggregated protein, total lipid, lipid esters, cholesterol, and metals (Fe, Cu, or Zn).


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Metales/metabolismo , Agregación Patológica de Proteínas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Ratones , Ratones Transgénicos , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/patología , Espectrometría por Rayos X , Espectroscopía Infrarroja por Transformada de Fourier , Espectrometría Raman
4.
BBA Adv ; 2: 100038, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37082604

RESUMEN

It is now well established that transition metals, such as Iron (Fe), Copper (Cu), and Zinc (Zn) are necessary for healthy brain function. Although Fe, Cu, and Zn are essential to the brain, imbalances in the amount, distribution, or chemical form ("metallome") of these metals is linked to the pathology of numerous brain diseases or disorders. Despite the known importance of metal ions for both brain health and disease, the metallome that exists within specific types of brain cells is yet to be fully characterised. The aim of this mini-review is to present an overview of the current knowledge of the metallome found within specific brain cells (oligodendrocytes, astrocytes, microglia, and neurons), as revealed by direct elemental mapping techniques. It is hoped this review will foster continued research using direct elemental mapping techniques to fully characterise the brain cell metallome.

5.
Metallomics ; 14(10)2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-36066906

RESUMEN

Metal ions (Fe, Cu, and Zn) are essential to a healthy brain function, with the amount, localisation, and chemical form often tightly controlled. Evidence points towards loss of metal ion homeostasis within the ageing brain; in particular brain Fe accumulation appears to be a hallmark of ageing, which may place the brain at a greater risk of neurodegenerative disease. Unfortunately, the cause or consequence of altered brain metal ion homeostasis during ageing remains unknown, and there is a lack of data comparing brain metal ion homeostasis with other events of the ageing process (e.g. brain metabolism, brain inflammation). This study has utilised a multi-modal approach that incorporated: X-ray fluorescence microscopy for elemental mapping of metal ion homeostasis, Perl's Fe histochemistry, FTIR spectroscopic biochemical imaging of lactate and protein aggregates, and immuno-fluorescence analysis of markers of brain inflammation and Fe storage proteins (heavy-chain ferritin, light-chain ferritin, and mitochondrial ferritin). Interestingly, while age-related Fe accumulation was observed in corpus callosum white matter of murine (C56BL/6J) brain tissue (concomitant with elevated levels of markers of brain inflammation and altered metabolism), Fe content was not altered within the hippocampus (a decrease in total Zn within the mossy fibres was observed). Ultimately, the results of this study demonstrate an important association between elevated brain Fe and brain inflammation during natural ageing. This study also highlights that future research is required to image different chemical forms of Fe with respect to changes in brain metabolism and inflammation, as well as localising these changes to specific cell types.


Asunto(s)
Encefalitis , Enfermedades Neurodegenerativas , Envejecimiento , Animales , Biomarcadores/metabolismo , Encefalitis/metabolismo , Ferritinas/metabolismo , Hipocampo/metabolismo , Homeostasis , Hierro/metabolismo , Lactatos/análisis , Lactatos/metabolismo , Ratones , Enfermedades Neurodegenerativas/metabolismo , Agregado de Proteínas
6.
Yakugaku Zasshi ; 141(6): 835-842, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34078791

RESUMEN

Dementia has no cure and is an international health crisis. In addition to the immeasurable loss of QOL caused by dementia, the global economic cost is predicted to reach $2 trillion (USD) by 2030. Although much remains unknown about the biochemical pathways driving cognitive decline and memory loss during dementia, metals have been implicated in neurodegenerative disease. For example, total levels of Fe and Cu increase, which has been proposed to drive oxidative stress; and Fe, Cu, and Zn can bind amyloid-ß, catalysing aggregation and formation of amyloid plaques. Unfortunately, despite these known facets through which metal ions may induce pathology, studies in greater detail have been hampered by a lack of microscopy methods to directly visualise metal ions, and their chemical form, within brain cells. Herein we report the use of synchrotron X-ray fluorescence microscopy to simultaneously image Fe, Cu, and Zn within neurons in ex vivo brain tissue sections. Using animal models of dementia, we now demonstrate for the first time that despite global increases in brain metal content and metal ion accumulation within amyloid plaques, key brain regions may also become metal ion deficient. Such deficiency could contribute to cognitive decline because of the essential roles metal ions play in neurotransmitter synthesis and energy metabolism. These recent findings are discussed in the context of memory loss, and the impact that metal ion dis-homeostasis may have on diagnostic and therapeutic development.


Asunto(s)
Demencia/etiología , Demencia/metabolismo , Hipocampo/metabolismo , Metales/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Cobre/metabolismo , Demencia/psicología , Modelos Animales de Enfermedad , Metabolismo Energético , Humanos , Iones , Hierro/metabolismo , Memoria , Ratones , Microscopía Fluorescente , Neurotransmisores/metabolismo , Unión Proteica , Deficiencias en la Proteostasis/etiología , Zinc/metabolismo
7.
Adv Sci (Weinh) ; 8(19): e2101902, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34338438

RESUMEN

Analysis of the epicuticular wax layer on the surface of plant leaves can provide a unique window into plant physiology and responses to environmental stimuli. Well-established analytical methodologies can quantify epicuticular wax composition, yet few methods are capable of imaging wax distribution in situ or in vivo. Here, the first report of Fourier transform infrared (FTIR) reflectance spectroscopic imaging as a non-destructive, in situ, method to investigate variation in epicuticular wax distribution at 25 µm spatial resolution is presented. The authors demonstrate in vivo imaging of alterations in epicuticular waxes during leaf development and in situ imaging during plant disease or exposure to environmental stressors. It is envisaged that this new analytical capability will enable in vivo studies of plants to provide insights into how the physiology of plants and crops respond to environmental stresses such as disease, soil contamination, drought, soil acidity, and climate change.


Asunto(s)
Enfermedades de las Plantas , Epidermis de la Planta/química , Fenómenos Fisiológicos de las Plantas , Estrés Fisiológico/fisiología , Ceras/química , Microscopía Electrónica de Rastreo , Hojas de la Planta/química
8.
Metallomics ; 12(12): 2134-2144, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33300524

RESUMEN

Zinc is a prominent trace metal required for normal memory function. Memory loss and cognitive decline during natural ageing and neurodegenerative disease have been associated with altered brain-Zn homeostasis. Yet, the exact chemical pathways through which Zn influences memory function during health, natural ageing, or neurodegenerative disease remain unknown. The gap in the literature may in part be due to the difficulty to simultaneously image, and therefore, study the different chemical forms of Zn within the brain (or biological samples in general). To this extent, we have begun developing and optimising protocols that incorporate X-ray absorption near-edge structure (XANES) spectroscopic analysis of tissue at the Zn K-edge as an analytical tool to study Zn speciation in the brain. XANES is ideally suited for this task as all chemical forms of Zn are detected, the technique requires minimal sample preparation that may otherwise redistribute or alter the chemical form of Zn, and the Zn K-edge has known sensitivity to coordination geometry and ligand type. Herein, we report our initial results where we fit K-edge spectra collected from micro-dissected flash-frozen brain tissue, to a spectral library prepared from standard solutions, to demonstrate differences in the chemical form of Zn that exist between two brain regions, the hippocampus and cerebellum. Lastly, we have used an X-ray microprobe to demonstrate differences in Zn speciation within sub-regions of thin air-dried sections of the murine hippocampus; but, the corresponding results highlight that the chemical form of Zn is easily perturbed by sample preparation such as tissue sectioning or air-drying, which must be a critical consideration for future work.


Asunto(s)
Química Encefálica , Zinc/análisis , Animales , Cationes Bivalentes/análisis , Masculino , Ratas Sprague-Dawley , Espectroscopía de Absorción de Rayos X
9.
ACS Chem Neurosci ; 11(3): 248-257, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31850738

RESUMEN

There is increased recognition of the effects of diffuse traumatic brain injury (dTBI), which can initiate yet unknown biochemical cascades, resulting in delayed secondary brain degeneration and long-term neurological sequela. There is limited availability of therapies that minimize the effect of secondary brain damage on the quality of life of people who have suffered TBI, many of which were otherwise healthy adults. Understanding the cascade of biochemical events initiated in specific brain regions in the acute phase of dTBI and how this spreads into adjacent brain structures may provide the necessary insight into drive development of improved therapies. In this study, we have used direct biochemical imaging techniques (Fourier transform infrared spectroscopic imaging) and elemental mapping (X-ray fluorescence microscopy) to characterize biochemical and elemental alterations that occur in corpus callosum white matter in the acute phase of dTBI. The results provide direct visualization of differential biochemical and ionic changes that occur in the highly vulnerable medial corpus callosum white matter relative to the less vulnerable lateral regions of the corpus callosum. Specifically, the results suggest that altered ionic gradients manifest within mechanically damaged medial corpus callosum, potentially spreading to and inducing lipid alterations to white matter structures in lateral brain regions.


Asunto(s)
Lesiones Encefálicas/metabolismo , Cuerpo Calloso/metabolismo , Lípidos , Sustancia Blanca/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Iones/química , Masculino , Pruebas Neuropsicológicas , Ratas Sprague-Dawley
10.
ACS Chem Neurosci ; 10(5): 2533-2540, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30855947

RESUMEN

Non-insulin-dependent diabetes mellitus (NIDDM) is reported to increase the risk of cognitive impairment and dementia. However, the underlying mechanisms are not fully understood. While the brain homeostasis of metals and lipids is pivotal to maintaining energy metabolism and redox homeostasis for healthy brain function, no studies have reported hippocampal metal and biochemical changes in NIDDM. Therefore, we here utilized direct spectroscopic imaging to reveal the elemental distribution within the hippocampal subregions of an established murine model of NIDDM, db/db mice. In 26-week-old insulin resistant db/db mice, X-ray fluorescence microscopy revealed that the Cu content within the dentate gyrus and CA3 was significantly greater than that of the age-matched nondiabetic control mice. In addition, Fourier transform infrared (FTIR) spectroscopy analysis indicated a significant increase in the abundance of lactate within the corpus callosum (CC), dentate gyrus, CA1, and CA3 regions of diabetic db/db mice compared to that of the control, indicating altered energy metabolism. FTIR analysis also showed a significant decrease in the level of lipid methylene and ester within the CC of db/db mice. Furthermore, immunomicroscopy analyses demonstrated the increase in the level of glial fibrillary acidic protein expression and peri-vascular extravasation of IgG, indicating astrogliosis and blood-brain barrier dysfunction, respectively. These data suggest that astrogliosis-induced alterations in the supply of Cu, lipids, and energy substrates may be involved in the mechanisms of NIDDM-associated cognitive decline.


Asunto(s)
Cobre/deficiencia , Diabetes Mellitus Tipo 2/metabolismo , Hipocampo/metabolismo , Lactatos/metabolismo , Metabolismo de los Lípidos/fisiología , Animales , Glucemia/metabolismo , Cobre/metabolismo , Metabolismo Energético/fisiología , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Masculino , Ratones Endogámicos , Microscopía Fluorescente/métodos , Imagen Multimodal/métodos , Espectroscopía Infrarroja por Transformada de Fourier/métodos
11.
ACS Chem Neurosci ; 9(11): 2774-2785, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29901988

RESUMEN

Western society is facing a health epidemic due to the increasing incidence of dementia in aging populations, and there are still few effective diagnostic methods, minimal treatment options, and no cure. Aging is the greatest risk factor for memory loss that occurs during the natural aging process, as well as being the greatest risk factor for neurodegenerative disease such as Alzheimer's disease. Greater understanding of the biochemical pathways that drive a healthy aging brain toward dementia (pathological aging or Alzheimer's disease), is required to accelerate the development of improved diagnostics and therapies. Unfortunately, many animal models of dementia model chronic amyloid precursor protein overexpression, which although highly relevant to mechanisms of amyloidosis and familial Alzheimer's disease, does not model well dementia during the natural aging process. A promising animal model reported to model mechanisms of accelerated natural aging and memory impairments, is the senescence accelerated murine prone strain 8 (SAMP8), which has been adopted by many research group to study the biochemical transitions that occur during brain aging. A limitation to traditional methods of biochemical characterization is that many important biochemical and elemental markers (lipid saturation, lactate, transition metals) cannot be imaged at meso- or microspatial resolution. Therefore, in this investigation, we report the first multimodal biospectroscopic characterization of the SAMP8 model, and have identified important biochemical and elemental alterations, and colocalizations, between 4 month old SAMP8 mice and the relevant control (SAMR1) mice. Specifically, we demonstrate direct evidence of Zn deficiency within specific subregions of the hippocampal CA3 sector, which colocalize with decreased lipid unsaturation. Our findings also revealed colocalization of decreased lipid unsaturation and increased lactate in the corpus callosum white matter, adjacent to the hippocampus. Such findings may have important implication for future research aimed at elucidating specific biochemical pathways for therapeutic intervention.


Asunto(s)
Envejecimiento , Región CA3 Hipocampal/diagnóstico por imagen , Cuerpo Calloso/diagnóstico por imagen , Demencia , Ácidos Grasos Insaturados/metabolismo , Ácido Láctico/metabolismo , Sustancia Blanca/diagnóstico por imagen , Zinc/deficiencia , Animales , Región CA3 Hipocampal/química , Región CA3 Hipocampal/metabolismo , Cuerpo Calloso/química , Cuerpo Calloso/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos Insaturados/análisis , Hipocampo/química , Hipocampo/diagnóstico por imagen , Hipocampo/metabolismo , Ácido Láctico/análisis , Metabolismo de los Lípidos , Ratones , Análisis Espectral , Sustancia Blanca/química , Sustancia Blanca/metabolismo , Zinc/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA