Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 20760, 2024 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-39237572

RESUMEN

Embryo quality assessment by optical imaging is increasing in popularity. Among available optical techniques, light sheet microscopy has emerged as a superior alternative to confocal microscopy due to its geometry, enabling faster image acquisition with reduced photodamage to the sample. However, previous assessments of photodamage induced by imaging may have failed to measure more subtle impacts. In this study, we employed DNA damage as a sensitive indicator of photodamage. We use light sheet microscopy with excitation at a wavelength of 405 nm for imaging embryo autofluorescence and compare its performance to laser scanning confocal microscopy. At an equivalent signal-to-noise ratio for images acquired with both modalities, light sheet microscopy reduced image acquisition time by ten-fold, and did not induce DNA damage when compared to non-imaged embryos. In contrast, imaging with confocal microscopy led to significantly higher levels of DNA damage within embryos and had a higher photobleaching rate. Light sheet imaging is also capable of inducing DNA damage within the embryo but requires multiple cycles of volumetric imaging. Collectively, this study confirms that light sheet microscopy is faster and safer than confocal microscopy for imaging live embryos, indicating its potential as a label-free diagnostic for embryo quality.


Asunto(s)
Daño del ADN , Embrión de Mamíferos , Microscopía Confocal , Microscopía Confocal/métodos , Animales , Ratones , Femenino , Imagen Óptica/métodos
2.
Eur J Med Chem ; 258: 115593, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37390508

RESUMEN

17ß-hydroxysteroid dehydrogenase type 10 (17ß-HSD10) is a multifunctional mitochondrial enzyme and putative drug target for the treatment of various pathologies including Alzheimer's disease or some types of hormone-dependent cancer. In this study, a series of new benzothiazolylurea-based inhibitors were developed based on the structure-activity relationship (SAR) study of previously published compounds and predictions of their physico-chemical properties. This led to the identification of several submicromolar inhibitors (IC50 ∼0.3 µM), the most potent compounds within the benzothiazolylurea class known to date. The positive interaction with 17ß-HSD10 was further confirmed by differential scanning fluorimetry and the best molecules were found to be cell penetrable. In addition, the best compounds weren't found to have additional effects for mitochondrial off-targets and cytotoxic or neurotoxic effects. The two most potent inhibitors 9 and 11 were selected for in vivo pharmacokinetic study after intravenous and peroral administration. Although the pharmacokinetic results were not fully conclusive, it seemed that compound 9 was bioavailable after peroral administration and could penetrate into the brain (brain-plasma ratio 0.56).


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Relación Estructura-Actividad , 17-Hidroxiesteroide Deshidrogenasas , Encéfalo/metabolismo , Inhibidores Enzimáticos/química
3.
Eur J Neurosci ; 57(7): 1161-1179, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36514861

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disease and the primary cause of disability and dependency among elderly humans worldwide. AD is thought to be a disease unique to humans although several other animals develop some aspects of AD-like pathology. Odontocetes (toothed whales) share traits with humans that suggest they may be susceptible to AD. The brains of 22 stranded odontocetes of five different species were examined using immunohistochemistry to investigate the presence or absence of neuropathological hallmarks of AD: amyloid-beta plaques, phospho-tau accumulation and gliosis. Immunohistochemistry revealed that all aged animals accumulated amyloid plaque pathology. In three animals of three different species of odontocete, there was co-occurrence of amyloid-beta plaques, intraneuronal accumulation of hyperphosphorylated tau, neuropil threads and neuritic plaques. One animal showed well-developed neuropil threads, phospho-tau accumulation and neuritic plaques, but no amyloid plaques. Microglia and astrocytes were present as expected in all brain samples examined, but we observed differences in cell morphology and numbers between individual animals. The simultaneous occurrence of amyloid-beta plaques and hyperphosphorylated tau pathology in the brains of odontocetes shows that these three species develop AD-like neuropathology spontaneously. The significance of this pathology with respect to the health and, ultimately, death of the animals remains to be determined. However, it may contribute to the cause(s) of unexplained live-stranding in some odontocete species and supports the 'sick-leader' theory whereby healthy conspecifics in a pod mass strand due to high social cohesion.


Asunto(s)
Enfermedad de Alzheimer , Delfines , Enfermedades Neurodegenerativas , Anciano , Animales , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Delfines/metabolismo , Placa Amiloide/metabolismo , Ovillos Neurofibrilares/metabolismo , Proteínas tau/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo
4.
Cells ; 11(19)2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36231104

RESUMEN

Willin/FRMD6 has been reported as a potential Alzheimer's disease (AD) risk gene in a series of genome-wide association and neuroimaging studies; however, the mechanisms underlying its potential role in AD pathogenesis remain unknown. Here, we demonstrate the direct effects of Aß on Willin/FRMD6 expression and position mitochondrial oxidative stress as a novel potential mechanism underlying the role of Willin/FRMD6 in AD pathogenesis. Specifically, using mouse hippocampal HT-22 cells and primary mouse neurons, we show that Aß induces downregulation of Willin/FRMD6 protein expression. Furthermore, we demonstrate that Willin/FRMD6 knockdown leads to mitochondrial dysfunction and fragmentation, as well as upregulation of ERK1/2 signaling, both of which are reported to be key early features of AD pathogenesis. Importantly, increasing Willin/FRMD6 expression was able to rescue Aß-induced abnormalities in mitochondrial morphology, function, and energetics. Thus, enhancing Willin/FRMD6 expression holds potential as a therapeutic strategy for protecting against Aß-induced mitochondrial and neuronal dysfunction.


Asunto(s)
Péptidos beta-Amiloides , Estudio de Asociación del Genoma Completo , Péptidos beta-Amiloides/metabolismo , Animales , Ratones , Mitocondrias/metabolismo , Neuronas/metabolismo , Transducción de Señal
5.
eNeuro ; 9(5)2022.
Artículo en Inglés | MEDLINE | ID: mdl-36096650

RESUMEN

Severe brain metabolic dysfunction and amyloid-ß accumulation are key hallmarks of Alzheimer's disease (AD). While astrocytes contribute to both pathologic mechanisms, the role of their mitochondria, which is essential for signaling and maintenance of these processes, has been largely understudied. The current work provides the first direct evidence that the mitochondrial metabolic switch 17ß-hydroxysteroid dehydrogenase type 10 (17ßHSD10) is expressed and active in murine astrocytes from different brain regions. While it is known that this protein is overexpressed in the brains of AD patients, we found that 17ßHSD10 is also upregulated in astrocytes exposed to amyloidogenic and ischemic stress. Importantly, such catalytic overexpression of 17ßHSD10 inhibits mitochondrial respiration during increased energy demand. This observation contrasts with what has been found in neuronal and cancer model systems, which suggests astrocyte-specific mechanisms mediated by the protein. Furthermore, the catalytic upregulation of the enzyme exacerbates astrocytic damage, reactive oxygen species (ROS) generation and mitochondrial network alterations during amyloidogenic stress. On the other hand, 17ßHSD10 inhibition through AG18051 counters most of these effects. In conclusion, our data represents novel insights into the role of astrocytic mitochondria in metabolic and amyloidogenic stress with implications of 17ßHSD10 in multiple neurodegenerative mechanisms.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/metabolismo , Enfermedad de Alzheimer , Astrocitos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Astrocitos/metabolismo , Ratones , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
Cells ; 10(11)2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34831245

RESUMEN

The FERM domain-containing protein 6 (FRMD6), also known as Willin, is an upstream regulator of Hippo signaling that has recently been shown to modulate actin cytoskeleton dynamics and mechanical phenotype of neuronal cells through ERK signaling. Physiological functions of Willin/FRMD6 in the nervous system include neuronal differentiation, myelination, nerve injury repair, and vesicle exocytosis. The newly established neuronal role of Willin/FRMD6 is of particular interest given the mounting evidence suggesting a role for Willin/FRMD6 in Alzheimer's disease (AD), including a series of genome wide association studies that position Willin/FRMD6 as a novel AD risk gene. Here we describe recent findings regarding the role of Willin/FRMD6 in the nervous system and its actions in cellular perturbations related to the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Regulación hacia Abajo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Unión Proteica , Transducción de Señal
8.
Front Cell Neurosci ; 14: 552213, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33088261

RESUMEN

Willin/FRMD6 is part of a family of proteins with a 4.1 ezrin-radixin-moesin (FERM) domain. It has been identified as an upstream activator of the Hippo pathway and, when aberrant in its expression, is associated with human diseases and disorders. Even though Willin/FRMD6 was originally discovered in the rat sciatic nerve, most studies have focused on its functional roles in cells outside of the nervous system, where Willin/FRMD6 is involved in the formation of apical junctional cell-cell complexes and in regulating cell migration. Here, we investigate the biochemical and biophysical role of Willin/FRMD6 in neuronal cells, employing the commonly used SH-SY5Y neuronal model cell system and combining biochemical measurements with Elastic Resonator Interference Stress Micropscopy (ERISM). We present the first direct evidence that Willin/FRMD6 expression influences both the cell mechanical phenotype and neuronal differentiation. By investigating cells with increased and decreased Willin/FRMD6 expression levels, we show that Willin/FRMD6 not only affects proliferation and migration capacity of cells but also leads to changes in cell morphology and an enhanced formation of neurite-like membrane extensions. These changes were accompanied by alterations of biophysical parameters such as cell force, the organization of actin stress fibers and the formation of focal adhesions. At the biochemical level, changes in Willin/FRMD6 expression inversely affected the activity of the extracellular signal-regulated kinases (ERK) pathway and downstream transcriptional factor NeuroD1, which seems to prime SH-SY5Y cells for retinoic acid (RA)-induced neuronal differentiation.

9.
Lipids Health Dis ; 19(1): 201, 2020 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-32867761

RESUMEN

BACKGROUND: Lipid dysregulation is associated with several key characteristics of Alzheimer's disease (AD), including amyloid-ß and tau neuropathology, neurodegeneration, glucose hypometabolism, as well as synaptic and mitochondrial dysfunction. The ß-site amyloid precursor protein cleavage enzyme 1 (BACE1) is associated with increased amyloidogenesis, and has been affiliated with diabetes via its role in metabolic regulation. METHODS: The research presented herein investigates the role of hBACE1 in lipid metabolism and whether specific brain regions show increased vulnerability to lipid dysregulation. By utilising advanced mass spectrometry techniques, a comprehensive, quantitative lipidomics analysis was performed to investigate the phospholipid, sterol, and fatty acid profiles of the brain from the well-known PLB4 hBACE1 knock-in mouse model of AD, which also shows a diabetic phenotype, to provide insight into regional alterations in lipid metabolism. RESULTS: Results show extensive region - specific lipid alterations in the PLB4 brain compared to the wild-type, with decreases in the phosphatidylethanolamine content of the cortex and triacylglycerol content of the hippocampus and hypothalamus, but increases in the phosphatidylcholine, phosphatidylinositol, and diacylglycerol content of the hippocampus. Several sterol and fatty acids were also specifically decreased in the PLB4 hippocampus. CONCLUSION: Collectively, the lipid alterations observed in the PLB4 hBACE1 knock-in AD mouse model highlights the regional vulnerability of the brain, in particular the hippocampus and hypothalamus, to lipid dysregulation, hence supports the premise that metabolic abnormalities have a central role in both AD and diabetes.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Ácido Aspártico Endopeptidasas/genética , Diabetes Mellitus Experimental/metabolismo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Metabolismo de los Lípidos/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diglicéridos/metabolismo , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Femenino , Expresión Génica , Técnicas de Sustitución del Gen , Hipocampo/patología , Humanos , Hipotálamo/patología , Lipidómica/métodos , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Fosfatidilcolinas/metabolismo , Fosfatidilinositoles/metabolismo , Esteroles/metabolismo , Transgenes
10.
Sci Rep ; 10(1): 8189, 2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32424125

RESUMEN

Functionally distinct synapses exhibit diverse and complex organisation at molecular and nanoscale levels. Synaptic diversity may be dependent on developmental stage, anatomical locus and the neural circuit within which synapses reside. Furthermore, astrocytes, which align with pre and post-synaptic structures to form 'tripartite synapses', can modulate neural circuits and impact on synaptic organisation. In this study, we aimed to determine which factors impact the diversity of excitatory synapses throughout the lumbar spinal cord. We used PSD95-eGFP mice, to visualise excitatory postsynaptic densities (PSDs) using high-resolution and super-resolution microscopy. We reveal a detailed and quantitative map of the features of excitatory synapses in the lumbar spinal cord, detailing synaptic diversity that is dependent on developmental stage, anatomical region and whether associated with VGLUT1 or VGLUT2 terminals. We report that PSDs are nanostructurally distinct between spinal laminae and across age groups. PSDs receiving VGLUT1 inputs also show enhanced nanostructural complexity compared with those receiving VGLUT2 inputs, suggesting pathway-specific diversity. Finally, we show that PSDs exhibit greater nanostructural complexity when part of tripartite synapses, and we provide evidence that astrocytic activation enhances PSD95 expression. Taken together, these results provide novel insights into the regulation and diversification of synapses across functionally distinct spinal regions and advance our general understanding of the 'rules' governing synaptic nanostructural organisation.


Asunto(s)
Médula Espinal/citología , Sinapsis/metabolismo , Animales , Astrocitos/citología , Procesamiento de Imagen Asistido por Computador , Ratones , Microscopía , Relación Señal-Ruido , Médula Espinal/diagnóstico por imagen
11.
J Neurochem ; 155(3): 231-249, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32306391

RESUMEN

17ß-hydroxysteroid dehydrogenase (17ß-HSD10) is a multifunctional human enzyme with important roles both as a structural component and also as a catalyst of many metabolic pathways. This mitochondrial enzyme has important functions in the metabolism, development and aging of the neural system, where it is involved in the homeostasis of neurosteroids, especially in regard to estradiol, changes in which make it an essential part of neurodegenerative pathology. These roles therefore, indicate that 17ß-HSD10 may be a possible druggable target for neurodegenerative diseases including Alzheimer's disease (AD), and in hormone-dependent cancer. The objective of this review was to provide a summary about physiological functions and pathological roles of 17ß-HSD10 and the modulators of its activity.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/genética , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Estado de Salud , Mitocondrias/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/química , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Humanos , Mitocondrias/genética , Mutación/genética , Neoplasias/genética , Neoplasias/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Estructura Secundaria de Proteína
12.
Int J Mol Sci ; 21(6)2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32192199

RESUMEN

Human 17ß-hydroxysteroid dehydrogenase type 10 is a multifunctional protein involved in many enzymatic and structural processes within mitochondria. This enzyme was suggested to be involved in several neurological diseases, e.g., mental retardation, Parkinson's disease, or Alzheimer's disease, in which it was shown to interact with the amyloid-beta peptide. We prepared approximately 60 new compounds based on a benzothiazolyl scaffold and evaluated their inhibitory ability and mechanism of action. The most potent inhibitors contained 3-chloro and 4-hydroxy substitution on the phenyl ring moiety, a small substituent at position 6 on the benzothiazole moiety, and the two moieties were connected via a urea linker (4at, 4bb, and 4bg). These compounds exhibited IC50 values of 1-2 µM and showed an uncompetitive mechanism of action with respect to the substrate, acetoacetyl-CoA. These uncompetitive benzothiazolyl inhibitors of 17ß-hydroxysteroid dehydrogenase type 10 are promising compounds for potential drugs for neurodegenerative diseases that warrant further research and development.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/antagonistas & inhibidores , Benzotiazoles/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Urea/química , Urea/farmacología , 3-Hidroxiacil-CoA Deshidrogenasas/química , Enfermedad de Alzheimer/tratamiento farmacológico , Activación Enzimática , Humanos , Cinética , Estructura Molecular , Proteínas Recombinantes , Relación Estructura-Actividad
13.
J Neurochem ; 155(2): 191-206, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32157699

RESUMEN

Mitochondrial dysfunction has a recognised role in the progression of Alzheimer's disease (AD) pathophysiology. Cerebral perfusion becomes increasingly inefficient throughout ageing, leading to unbalanced mitochondrial dynamics. This effect is exaggerated by amyloid ß (Aß) and phosphorylated tau, two hallmark proteins of AD pathology. A neuroprotective role for the adipose-derived hormone, leptin, has been demonstrated in neuronal cells. However, its effects with relation to mitochondrial function in AD remain largely unknown. To address this question, we have used both a glucose-serum-deprived (CGSD) model of ischaemic stroke in SH-SY5Y cells and a Aß1-42 -treatment model of AD in differentiated hippocampal cells. Using a combination of 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1) and MitoRed staining techniques, we show that leptin prevents depolarisation of the mitochondrial membrane and excessive mitochondrial fragmentation induced by both CGSD and Aß1-42 . Thereafter, we used ELISAs and a number of activity assays to reveal the biochemical underpinnings of these processes. Specifically, leptin was seen to inhibit up-regulation of the mitochondrial fission protein Fis1 and down-regulation of the mitochondrial fusion protein, Mfn2. Furthermore, leptin was seen to up-regulate the expression and activity of the antioxidant enzyme, monoamine oxidase B. Herein we provide the first demonstration that leptin is sufficient to protect against aberrant mitochondrial dynamics and resulting loss of function induced by both CGSD and Aß1-42 . We conclude that the established neuroprotective actions of leptin may be facilitated through regulation of mitochondrial dynamics.


Asunto(s)
Leptina/farmacología , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Dinámicas Mitocondriales/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Péptidos beta-Amiloides/farmacología , Animales , Línea Celular , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/biosíntesis , Glucosa/deficiencia , Hipocampo/citología , Hipocampo/patología , Humanos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Ratones , Mitocondrias/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/biosíntesis , Monoaminooxidasa/metabolismo , Fragmentos de Péptidos/farmacología , Especies Reactivas de Oxígeno/metabolismo
14.
Molecules ; 24(15)2019 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-31362457

RESUMEN

: It has long been established that mitochondrial dysfunction in Alzheimer's disease (AD) patients can trigger pathological changes in cell metabolism by altering metabolic enzymes such as the mitochondrial 17ß-hydroxysteroid dehydrogenase type 10 (17ß-HSD10), also known as amyloid-binding alcohol dehydrogenase (ABAD). We and others have shown that frentizole and riluzole derivatives can inhibit 17ß-HSD10 and that this inhibition is beneficial and holds therapeutic merit for the treatment of AD. Here we evaluate several novel series based on benzothiazolylurea scaffold evaluating key structural and activity relationships required for the inhibition of 17ß-HSD10. Results show that the most promising of these compounds have markedly increased potency on our previously published inhibitors, with the most promising exhibiting advantageous features like low cytotoxicity and target engagement in living cells.


Asunto(s)
17-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , 17-Hidroxiesteroide Deshidrogenasas/química , Benzotiazoles/química , Urea/química , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Humanos , Mitocondrias/metabolismo , Estructura Molecular , Relación Estructura-Actividad
15.
Int J Mol Sci ; 20(12)2019 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-31242683

RESUMEN

Drug delivery to the central nervous system (CNS) conferred by brain barriers is a major obstacle in the development of effective neurotherapeutics. In this review, a classification of current approaches of clinical or investigational importance for the delivery of therapeutics to the CNS is presented. This classification includes the use of formulations administered systemically that can elicit transcytosis-mediated transport by interacting with transporters expressed by transvascular endothelial cells. Neurotherapeutics can also be delivered to the CNS by means of surgical intervention using specialized catheters or implantable reservoirs. Strategies for delivering drugs to the CNS have evolved tremendously during the last two decades, yet, some factors can affect the quality of data generated in preclinical investigation, which can hamper the extension of the applications of these strategies into clinically useful tools. Here, we disclose some of these factors and propose some solutions that may prove valuable at bridging the gap between preclinical findings and clinical trials.


Asunto(s)
Fármacos del Sistema Nervioso Central/farmacología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Transcitosis , Animales , Transporte Biológico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Fármacos del Sistema Nervioso Central/administración & dosificación , Ensayos Clínicos como Asunto , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Humanos , Resultado del Tratamiento
16.
J Neurosci Methods ; 319: 16-27, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30048674

RESUMEN

BACKGROUND: The functions of the central nervous system (CNS) rely on the interaction between large populations of neurons across different areas. Therefore, to comprehend CNS functions there is a need for imaging techniques providing access to the neuronal activity of large networks of neurons with very high spatiotemporal resolution. NEW METHOD: Light sheet fluorescence microscopy (LSFM) is a very promising optical sectioning technique that allows volumetric imaging over many length scales while retaining high spatial resolution and minimizing photobleaching and phototoxicity. RESULTS: The application of LSFM in neuroscience opened up the possibility of imaging in-vivo the development of the CNS and acquiring morphological images of whole cleared mammalian brains with sub-cellular resolution. The use of propagation invariant Bessel and Airy beams has shown potential for increasing the penetration depth in turbid neural tissues. COMPARISON WITH EXISTING METHODS: The lack of temporal and/or spatial resolution of traditional neuroscience imaging techniques call attention to a need for a technique capable of providing high spatio temporal resolution. LSFM, which is capable of acquiring high resolution volumetric images is increasingly becoming an interesting imaging technique for neuroscience. CONCLUSIONS: The use of different LSFM geometries has shown the potential of this technique in acquiring in-vivo functional images of the CNS and morphological images of entire cleared mammalian brains. Further development of single objective LSFM implementations and fibre based LSFM combined with the use of propagation invariant beams could allow this technique to be used for in depth in-vivo imaging.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/diagnóstico por imagen , Microscopía Fluorescente/métodos , Neuroimagen/métodos , Imagen Óptica/métodos , Animales , Humanos , Procesamiento de Imagen Asistido por Computador , Neuronas/citología
17.
Nat Commun ; 9(1): 2968, 2018 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-30061577

RESUMEN

Endophilin A1 (EP) is a protein enriched in synaptic terminals that has been linked to Alzheimer's disease (AD). Previous in vitro studies have shown that EP can bind to a variety of proteins, which elicit changes in synaptic transmission of neurotransmitters and spine formation. Additionally, we previously showed that EP protein levels are elevated in AD patients and AD transgenic animal models. Here, we establish the in vivo consequences of upregulation of EP expression in amyloid-ß peptide (Aß)-rich environments, leading to changes in both long-term potentiation and learning and memory of transgenic animals. Specifically, increasing EP augmented cerebral Aß accumulation. EP-mediated signal transduction via reactive oxygen species (ROS)/p38 mitogen-activated protein (MAP) kinase contributes to Aß-induced mitochondrial dysfunction, synaptic injury, and cognitive decline, which could be rescued by blocking either ROS or p38 MAP kinase activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Enfermedad de Alzheimer/genética , Regulación de la Expresión Génica , Adenosina Trifosfato/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Animales Modificados Genéticamente , Antioxidantes/metabolismo , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Potenciación a Largo Plazo , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Neuronas/metabolismo , Neurotransmisores/metabolismo , Fragmentos de Péptidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sinapsis/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Front Neurosci ; 12: 451, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30026684

RESUMEN

We introduce a novel all-optical assay for functional studies of biological neural networks in vitro. We created a novel optogenetic construct named OptoCaMP which is a combination of a channelrhodopsin variant (CheRiff) and a red genetically encoded calcium indicator (GECI) (jRCaMP1b). It enables simultaneous optical stimulation and recording from large population of neurons with single-cell readout. Additionally, we have developed a spatio-temporal all-optical assay to simultaneously stimulate a sub-section of a neural network and record evoked calcium activity, in both stimulated and non-stimulated neurons, thus allowing the investigation of the spread of excitation through an interconnected network. Finally, we demonstrate the sensitivity of this assay to the change of neural network connectivity.

19.
Sci Adv ; 4(4): eaar4817, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29740614

RESUMEN

Scattering and absorption limit the penetration of optical fields into tissue. We demonstrate a new approach for increased depth penetration in light-sheet microscopy: attenuation-compensation of the light field. This tailors an exponential intensity increase along the illuminating propagation-invariant field, enabling the redistribution of intensity strategically within a sample to maximize signal and minimize irradiation. A key attribute of this method is that only minimal knowledge of the specimen transmission properties is required. We numerically quantify the imaging capabilities of attenuation-compensated Airy and Bessel light sheets, showing that increased depth penetration is gained without compromising any other beam attributes. This powerful yet straightforward concept, combined with the self-healing properties of the propagation-invariant field, improves the contrast-to-noise ratio of light-sheet microscopy up to eightfold across the entire field of view in thick biological specimens. This improvement can significantly increase the imaging capabilities of light-sheet microscopy techniques using Airy, Bessel, and other propagation-invariant beam types, paving the way for widespread uptake by the biomedical community.

20.
J Enzyme Inhib Med Chem ; 33(1): 665-670, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29536773

RESUMEN

Several neurodegenerative disorders including Alzheimer's disease (AD) have been connected with deregulation of casein kinase 1 (CK1) activity. Inhibition of CK1 therefore presents a potential therapeutic strategy against such pathologies. Recently, novel class of CK1-specific inhibitors with N-(benzo[d]thiazol-2-yl)-2-phenylacetamide structural scaffold has been discovered. 1-(benzo[d]thiazol-2-yl)-3-phenylureas, on the other hand, are known inhibitors amyloid-beta binding alcohol dehydrogenase (ABAD), an enzyme also involved in pathophysiology of AD. Based on their tight structural similarity, we decided to evaluate series of previously published benzothiazolylphenylureas, originally designed as ABAD inhibitors, for their inhibitory activity towards CK1. Several compounds were found to be submicromolar CK1 inhibitors. Moreover, two compounds were found to inhibit both, ABAD and CK1. Such dual-activity could be of advantage for AD treatment, as it would simultaneously target two distinct pathological processes involved in disease's progression. Based on PAMPA testing both compounds were suggested to permeate the blood-brain barrier, which makes them, together with their unique dual activity, interesting lead compounds for further development.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Quinasa de la Caseína I/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Compuestos de Fenilurea/farmacología , Quinasa de la Caseína I/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Enfermedades Neurodegenerativas/metabolismo , Compuestos de Fenilurea/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA